US20050043227A1 - Use of hypoxia inducible factor 2alpha for curing neonatal respiratory distress syndrome and as a target for the treatment of pulmonary hypertension - Google Patents

Use of hypoxia inducible factor 2alpha for curing neonatal respiratory distress syndrome and as a target for the treatment of pulmonary hypertension Download PDF

Info

Publication number
US20050043227A1
US20050043227A1 US10/475,403 US47540304A US2005043227A1 US 20050043227 A1 US20050043227 A1 US 20050043227A1 US 47540304 A US47540304 A US 47540304A US 2005043227 A1 US2005043227 A1 US 2005043227A1
Authority
US
United States
Prior art keywords
hif
mice
vegf
hypoxia
lung
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.)
Abandoned
Application number
US10/475,403
Other languages
English (en)
Inventor
Veerle Compernolle
Peter Carmeliet
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.)
Vlaams Instituut voor Biotechnologie VIB
Original Assignee
Vlaams Instituut voor Biotechnologie VIB
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 Vlaams Instituut voor Biotechnologie VIB filed Critical Vlaams Instituut voor Biotechnologie VIB
Assigned to VLAAMS INTERUNIVERSITAIR INSTITUUT VOOR BIOTECHNOLOGIE VZW reassignment VLAAMS INTERUNIVERSITAIR INSTITUUT VOOR BIOTECHNOLOGIE VZW ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CARMELIET, PETER, COMPERNOLLE, VEERLE
Publication of US20050043227A1 publication Critical patent/US20050043227A1/en
Priority to US11/226,771 priority Critical patent/US7307060B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1858Platelet-derived growth factor [PDGF]
    • A61K38/1866Vascular endothelial growth factor [VEGF]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/74Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving hormones or other non-cytokine intercellular protein regulatory factors such as growth factors, including receptors to hormones and growth factors
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • G01N2333/4701Details
    • G01N2333/4703Regulators; Modulating activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/12Pulmonary diseases
    • G01N2800/125Adult respiratory distress syndrome
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/32Cardiovascular disorders
    • G01N2800/321Arterial hypertension
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/38Pediatrics

Definitions

  • the current invention relates to the field of hypoxia-induced disorders and more specifically to the use of hypoxia inducible factor 2 ⁇ (HIF-2 ⁇ ) as a target in a method for the screening for molecules that can be used for the treatment of pulmonary hypertension.
  • the invention further relates to the use of HIF-2 ⁇ and/or the HIF-2 ⁇ inducible protein VEGF for the treatment of neonatal respiratory distress syndrome.
  • Preterm delivery is the chief problem in obstetrics today, affecting 10% of all births 1 . It accounts for more than 70% of perinatal mortality and nearly half of long-term neurological morbidity, especially in infants who are born at less than 32 weeks of gestation and weigh less than 1,000 g. With ⁇ 60% of these newborns developing respiratory distress syndrome (RDS) and a 50% lethality, RDS is the leading cause of neonatal mortality 2 .
  • RDS results from insufficient production of surfactant by immature type 2 pneumocytes in preterm infants, but can also result from dysfunction or deficiency of surfactant in term infants due to inherited mutations, meconium aspiration, hemorrhage, infections and others 3 .
  • Surfactant is a mixture of phospholipids and surfactant-associated proteins (SP-A to SP-D), which lowers surface tension at the air-water interface and thereby prevents alveolar collapse and respiratory failure.
  • Surfactant phospholipids are synthetized from substrates, provided by glycogen stores in fetal immature pneumocytes 4 .
  • Neonatal intensive care has improved the survival of infants with RDS, but often at the expense of the development of bronchopulmonary dysplasia or chronic lung disease of prematurity 5 .
  • Treatment with oxygen may irreversibly damage lung parenchyma and angiogenesis, while prenatal steroid treatment causes neurological, metabolic, cardiovascular and hormonal side-effects, and impairs growth 6 .
  • Surfactant treatment is effective, but expensive and only symptomatic 7 .
  • VEGF vascular endothelial growth factor
  • Flk-1 Flk-1
  • VEGF-R1 Flt-1
  • VEGF-R2 Flk-1
  • VEGF-R1 Flt-1
  • VEGF is deposited at the leading edge of branching airways, where it stimulates vascularization 10 . Indirect evidence suggests, however, that VEGF also affects epithelial growth and differentiation.
  • Type 2 pneumocytes and bronchiolar epithelial cell produce VEGF and possess VEGF receptors 11,12 VEGF levels are also considerably higher in the bronchoalveolar fluid than in the blood 12 , suggesting that epithelial cells affect their own function by releasing VEGF into the airway lumen.
  • the lung is one of the few organs where VEGF levels remain elevated in the adult, even though no active angiogenesis occurs. Previous studies provided circumstantial evidence for a role of VEGF in lung development, but did not provide functional in vivo proof for a role of VEGF in lung maturation and surfactant production.
  • VEGF levels in tracheal aspirate were lower in infants with lung immaturity developing bronchopulmonary dysplasia than in those surviving without pulmonary complications in some 13 -15 but not in other studies 16 .
  • Exogenous VEGF stimulates growth of epithelial cells in early embryonic lung explants in vitro 17 , but the relevance of endogenous VEGF for lung maturation just prior to birth in vivo and the possible therapeutic potential of VEGF in preventing RDS in preterm infants remain unknown.
  • loss of HIF-2 ⁇ causes fatal RDS in newborn due to insufficient surfactant production.
  • VEGF plays an important role in lung maturation since VEGF levels are reduced in HIF-2 ⁇ deficient mice, neonates expressing only the VEGF 120 isoform or with impaired HIF-2 ⁇ -dependent VEGF expression die of RDS, and intra-amniotic administration of anti-Flk-1 antibodies aggravated lung prematurity.
  • intra-uterine delivery of VEGF before birth or intra-tracheal injection of VEGF after birth stimulates conversion of glycogen to surfactant, improved lung function and prevented RDS in premature.
  • one aspect of the invention shows the use of VEGF for the manufacture of a medicament to treat RDS in premature infants.
  • hypoxia HIF-1 ⁇ upregulates the expression of a number of genes involved in erythropoiesis, glycolysis and angiogenesis by formation of a heterodimer with HIF-1 ⁇ (also termed aryl hydrocarbon receptor nuclear translocator; ARNT), which binds to a hypoxia-response element (HRE) in the promoter of these target genes.
  • HIF-1 ⁇ has also been implicated in the induction of apoptosis in hypoxic and hypoglycaemic conditions.
  • HIF-2 ⁇ hypoxia-inducible factor
  • HIF-2 ⁇ also known as EPAS-1, HLF, HRF or MOP2
  • EPAS-1, HLF, HRF or MOP2 hypoxia-inducible factor-2 ⁇
  • HIF-2 ⁇ is a homologue of HIF-1 ⁇
  • the role of HIF-2 ⁇ in glycolytic, angiogenic, apoptotic or possible disease processes is unknown and unpredictable.
  • A Top, targeting vector pPNT.HIF-2 ⁇ ; middle, map of the wild type (WT) gene; bottom, homologously recombined (HR) HIF-2 ⁇ allele.
  • Analytical restriction digests and hybridization probes A (0.6-kb NcoI-EcoRV fragment) and B (2.3-kb NheI-EcoRI fragment) for genotyping are indicated.
  • B Southern blot analysis (probe A) of StuI-digested genomic DNA from ES cells generating a 7-kb WT and 7.5-kb HR HIF-2 ⁇ allele.
  • C RTPCR analysis of total RNA of ES cells for HIF-2 ⁇ gene expression. HPRT gene expression was used as an internal control.
  • D Immunoblot analysis on total cell extract from WT, HIF-1 ⁇ ⁇ / ⁇ and HIF-2 ⁇ ⁇ / ⁇ ES cells for HIF-2 ⁇ gene expression during normoxia (N) and hypoxia (H).
  • HIF-2 ⁇ +/ ⁇ mice were protected against hypoxia-induced pulmonary hypertension, vascular remodeling and right ventricle hypertrophy.
  • the right ventricular hypertrophy is surprisingly absent in the HIF-2 ⁇ +/ ⁇ mice.
  • HIF-1 ⁇ +/ ⁇ mice lose weight than WT mice during chronic hypoxia, despite their reduced pulmonary hypertension.
  • WT mice lost more weight than HIF-2 ⁇ +/ ⁇ mice in hypoxia, which is in agreement with the prevention of pulmonary hypertension in HIF-2 ⁇ +/ ⁇ mice.
  • HIF-2 ⁇ plays, to our surprise, a more important role than HIF-1 ⁇ in hypoxic lungs and hence the invention demonstrates a crucial role for HIF-2 ⁇ in the development of pulmonary hypertension and vascular remodeling and implies the use of HIF-2 ⁇ inhibitors to prevent hypoxia-induced pulmonary hypertension, a pathology with high morbidity and mortality.
  • the invention provides the use of the hypoxia inducible factor 2 ⁇ (HIF-2 ⁇ ) or fragments thereof as a target in a method to screen for molecules that are able to inhibit the development of pulmonary hypertension
  • said method to screen for molecules comprises the following steps a) incubating a mixture comprising HIF-2 ⁇ or a fragment thereof and at least one molecule, b) allowing binding between HIF-2 ⁇ or a fragment thereof and said molecule, c) isolating said molecule binding to HIF-2 ⁇ , or a fragment thereof and d) determining the ability of said molecule to inhibit the development of pulmonary hypertension.
  • antibody or ‘antibodies’ relates to an antibody characterized as being specifically directed against HIF-2 ⁇ or any functional derivative thereof, with said antibodies being preferably monoclonal antibodies; or an antigen-binding fragment thereof, of the F(ab′) 2 , F(ab) or single chain Fv type, or any type of recombinant antibody derived thereof.
  • These antibodies of the invention including specific polyclonal antisera prepared against HIF-2 ⁇ or any functional derivative thereof, have no cross-reactivity to others proteins.
  • the monoclonal antibodies of the invention can for instance be produced by any hybridoma liable to be formed according to classical methods from splenic cells of an animal, particularly of a mouse or rat immunized against HIF-2 ⁇ or any functional derivative thereof, and of cells of a myeloma cell line, and to be selected by the ability of the hybridoma to produce the monoclonal antibodies recognizing HIF-2 ⁇ or any functional derivative thereof which have been initially used for the immunization of the animals.
  • the monoclonal antibodies according to this embodiment of the invention may be humanized versions of the mouse monoclonal antibodies made by means of recombinant DNA technology, departing from the mouse and/or human genomic DNA sequences coding for H and L chains or from cDNA clones coding for H and L chains.
  • the monoclonal antibodies according to this embodiment of the invention may be human monoclonal antibodies.
  • Such human monoclonal antibodies are prepared, for instance, by means of human peripheral blood lymphocytes (PBL) repopulation of severe combined immune deficiency (SCID) mice as described in PCT/EP 99/03605 or by using transgenic non-human animals capable of producing human antibodies as described in U.S. Pat. No. 5,545,806.
  • PBL peripheral blood lymphocytes
  • SCID severe combined immune deficiency
  • fragments derived from these monoclonal antibodies such as Fab, F(ab)′ 2 and scFv (“single chain variable fragment”), providing they have retained the original binding properties, form part of the present invention.
  • Such fragments are commonly generated by, for instance, enzymatic digestion of the antibodies with papain, pepsin, or other proteases. It is well known to the person skilled in the art that monoclonal antibodies, or fragments thereof, can be modified for various uses.
  • the antibodies involved in the invention can be labeled by an appropriate label of the enzymatic, fluorescent, or radioactive type.
  • the inhibitor of HIF-2 ⁇ can be a camel antibody or a functional fragment thereof.
  • Camel antibodies are fully described in WO94/25591, WO94/04678 and in WO97/49805.
  • the camel antibody can be directed to HIF-2 ⁇ , preventing the binding and/or subsequent signal transduction of HIF-2 ⁇ .
  • Antibodies can be used to hit intracellular targets such as HIF-2 ⁇ by processes as for example ‘antibody lipidation’, the latter is fully described in WO94/01131.
  • Random peptide libraries consisting of all possible combinations of amino acids attached to a solid phase support may be used to identify peptides that are able to bind to the ligand binding site of a given receptor or other functional domains of a receptor such as kinase domains (Lam K S et al., 1991, Nature 354, 82).
  • the screening of peptide libraries may also have therapeutic value in the discovery of pharmaceutical agents that act to inhibit the biological activity of HIF-2 ⁇ . Identification of molecules that are able to bind to HIF-2 ⁇ may be accomplished by for example screening a peptide library with recombinant soluble HIF-2 ⁇ protein.
  • oligoribonucleotide sequences that include anti-sense RNA and DNA molecules and ribozymes that function to inhibit the translation of HIF-2 ⁇ mRNA.
  • Anti-sense RNA and DNA molecules act to directly block the translation of mRNA by binding to targeted mRNA and preventing protein translation.
  • antisense DNA oligodeoxyribonucleotides derived from the translation initiation site, e.g., between ⁇ 10 and +10 regions of the HIF-2 ⁇ nucleotide sequence, are preferred.
  • Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA.
  • ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by a endonucleolytic cleavage.
  • engineered hammerhead motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of HIF-2 ⁇ RNA sequences.
  • Specific ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites which include the following sequences, GUA, GUU and GUC.
  • RNA sequences of between 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site may be evaluated for predicted structural features such as secondary structure that may render the oligonucleotide sequence unsuitable.
  • the suitability of candidate targets may also be evaluated by testing their accessibility to hybridization with complementary oligonucleotides, using ribonuclease protection assays.
  • RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding the antisense RNA molecule. Such DNA sequences may be incorporated into a wide variety of vectors which incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters.
  • antisense cDNA constructs that synthesize anti-sense RNA constitutively or inducibly, depending on the promoter used, can be introduced stably into cell lines.
  • the invention also provides methods for identifying molecules that bind on HIF-2 ⁇ and antagonize the signal transduction. These methods are also referred to as ‘drug screening assays’ or ‘bioassays’ and typically include the step of screening a candidate/test compound or agent for the ability to interact with HIF-2 ⁇ .
  • Candidate compounds or agents, which have this ability, can be used as drugs to combat or prevent pulmonary hypertension.
  • Candidate/test compounds such as small molecules, e.g. small organic molecules, and other drug candidates can be obtained, for example, from combinatorial and natural product libraries as described above.
  • the assays are cell-free assays which include the steps of combining HIF-2 ⁇ and a candidate/test compound, e.g., under conditions which allow for interaction of (e.g. binding of) the candidate/test compound with HIF-2 ⁇ to form a complex, and detecting the formation of a complex, in which the ability of the candidate compound to interact with HIF-2 ⁇ is indicated by the presence of the candidate compound in the complex. Formation of complexes between the HIF-2 ⁇ and the candidate compound can be quantitated, for example, using standard immunoassays.
  • the HIF-2 ⁇ employed in such a test may be free in solution, affixed to a solid support, borne on a cell surface, or located intracellularly.
  • HIF-2 ⁇ or its (their) target molecule(s) to facilitate separation of complexes from uncomplexed forms of one or both of the proteins, as well as to accommodate automation of the assay.
  • Interaction e.g., binding on of HIF-2 ⁇ to a target molecule, can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtiter plates, test tubes, and microcentrifuge tubes.
  • a fusion protein can be provided which adds a domain that allows the protein to be bound to a matrix.
  • HIF-2 ⁇ tagged can be adsorbed onto Ni-NTA microtiter plates, or HIF-2 ⁇ -ProtA fusions adsorbed to IgG, which are then combined with the cell lysates (e.g., 35 S-labeled) and the candidate compound, and the mixture incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). Following incubation, the plates are washed to remove any unbound label, and the matrix immobilized and radiolabel determined directly, or in the supernatant after the complexes are dissociated.
  • cell lysates e.g., 35 S-labeled
  • the candidate compound e.g., 35 S-labeled
  • the mixture incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH).
  • the plates are washed to remove any unbound label, and the matrix immobilized and radiolabel determined directly, or in the supernatant after the complexes are dissociated
  • the complexes can be dissociated from the matrix, separated by SDS-PAGE, and the level of HIF-2 ⁇ binding protein found in the bead fraction quantitated from the gel using standard electrophoretic techniques.
  • Other techniques for immobilizing protein on matrices can also be used in the drug screening assays of the invention.
  • HIF-2 ⁇ or its target molecules can be immobilized utilizing conjugation of biotin and streptavidin.
  • Biotinylated HIF-2 ⁇ can be prepared from biotin-NHS(N-hydroxy-succinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, Ill.), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical).
  • biotinylation kit Pierce Chemicals, Rockford, Ill.
  • streptavidin-coated 96 well plates Piereptavidin-coated 96 well plates
  • antibodies reactive with HIF-2 ⁇ but which do not interfere with binding of the protein to its target molecule can be derivatized to the wells of the plate, and HIF-2 ⁇ trapped in the wells by antibody conjugation.
  • preparations of a HIF-2 ⁇ -binding protein and a candidate compound are incubated in the HIF-2 ⁇ -presenting wells of the plate, and the amount of complex trapped in the well can be quantitated.
  • Methods for detecting such complexes include immunodetection of complexes using antibodies reactive with the HIF-2 ⁇ -target molecule, or which are reactive with HIF-2 ⁇ and compete with the target molecule; as well as enzyme-linked assays which rely on detecting an enzymatic activity associated with the target molecule.
  • HIF-2 ⁇ Another technique for drug screening which provides for high throughput screening of compounds having suitable binding affinity to HIF-2 ⁇ is described in detail in “Determination of Amino Acid Sequence Antigenicity” by Geysen H N, WO 84/03564, published on Sept. 13, 1984.
  • large numbers of different small peptide test compounds are synthesized on a solid substrate, such as plastic pins or some other surface.
  • the protein test compounds are reacted with fragments of HIF-2 ⁇ and washed. Bound HIF-2 ⁇ is then detected by methods well known in the art.
  • Purified HIF-2 ⁇ can also be coated directly onto plates for use in the aforementioned drug screening techniques.
  • non-neutralizing antibodies can be used to capture the peptide and immobilize it on a solid support.
  • This invention also contemplates the use of competitive drug screening assays in which neutralizing antibodies capable of binding HIF-2 ⁇ specifically compete with a test compound for binding HIF-2 ⁇ . In this manner, the antibodies can be used to detect the presence of any protein, which shares one or more antigenic determinants with HIF-2 ⁇ .
  • the invention provides the use of HIF-2 ⁇ or fragments thereof in a method to screen for molecules that are able to inhibit the development of pulmonary hypertension
  • said method comprises the following steps: a) incubating a mixture comprising HIF-2 ⁇ , or a fragment thereof, a reporter construct wherein the reporter gene is driven by the transcription factor HIF-2 ⁇ , and at least one molecule, b) determining if the latter incubation results in at least 50% reduction in expression of said reporter gene, and c) determining the ability of said molecule to inhibit the development of pulmonary hypertension.
  • a molecule able to reduce the expression of a reporter gene by at least 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100% will also reduce the level of HIF-2 ⁇ protein when said molecule is applied to a cell culture or an organism. Since the HIF-2 ⁇ ⁇ /+ mice are protected against pulmonary hypertension, at least 50% reduction or inhibition of its activity of HIF-2 ⁇ protein might be necessary and sufficient to inhibit pulmonary hypertension.
  • molecules it is meant peptides, proteins, organic molecules and carbohydrates.
  • Small molecules eg small organic molecules, and other drug candidates can be obtained, for example, from combinatorial and natural product libraries. Random peptide libraries consisting of all possible combinations of amino acids attached to a solid phase support may also be used to identify peptides that are able to bind to specific ligands (Lam K S et al., 1991, Nature 354, 82).
  • a “reporter gene” is a DNA molecule that expresses a detectable gene product, which may be RNA or protein.
  • the detection may be accomplished by any method known to one of skill in the art. For example, detection of mRNA expression may be accomplished by using Northern blots and detection of protein may be accomplished by staining with antibodies specific to the protein.
  • Preferred reporter genes are those that are readily detectable.
  • a reporter gene may be operably linked in a DNA construct with a regulatory DNA sequence such that detection of the reporter gene product provides a measure of the transcriptional activity of the regulatory sequence.
  • reporter genes include, but are not limited to, those coding for chloramphenicol acetyl transferase (CAT), luciferase, beta-galactosidase and alkaline phosphatase.
  • CAT chloramphenicol acetyl transferase
  • luciferase luciferase
  • beta-galactosidase alkaline phosphatase.
  • the term “operably linked” refers to linkage of a DNA segment to another DNA segment in such a way as to allow the segments to function in their intended manners.
  • a DNA sequence encoding a gene product is operably linked to a regulatory sequence when it is ligated to the regulatory sequence, such as, for example, promoters, enhancers and silencers, in a manner which allows modulation of transcription of the DNA sequence, directly or indirectly.
  • a DNA sequence is operably linked to a promoter when it is ligated to the promoter downstream with respect to the transcription initiation site of the promoter, in the correct reading frame with respect to the transcription initiation site and allows transcription elongation to proceed through the DNA sequence.
  • An enhancer or silencer is operably linked to a DNA sequence coding for a gene product when it is ligated to the DNA sequence in such a manner as to increase or decrease respectively the transcription of the DNA sequence. Enhancers and silencers may be located upstream, downstream or embedded within the coding regions of the DNA sequence.
  • a DNA for a signal sequence is operably linked to DNA coding for a polypeptide if it is expressed as a pre-protein that participates in the secretion of the polypeptide. Linkage of DNA sequences to regulatory sequences is typically accomplished by ligation at suitable restriction sites or adapters or linkers inserted in lieu thereof using restriction endonucleases known to one of skill in the art.
  • a promoter construct operably linked to for example a reporter gene may be transfected into a cell line using any technique previously described to produce for example a stable cell line, but also transiently transfected host cells can be used, containing the reporter construct integrated into the genome.
  • the cells may be grown and incubated with test compounds for varying times.
  • the cells may be grown in 96 well plates to facilitate the analysis of large numbers of compounds.
  • the cells may then be washed and the reporter gene expression analysed. For some reporters, such as luciferase the cells will be lysed and subsequently analysed.
  • the level of expression in the presence of the test compound may be compared with the level of expression in the absence of the test compound.
  • Pulmonary hypertension is a specific condition of hypertension in the lung and relates to arterial hypertension, capillary hypertension or venous-hypertension in the lung.
  • pulmonary hypertension relates to pulmonary arterial hypertension.
  • pulmonary arterial hypertension relates to—but is not restricted to—both primary arterial hypertension and to pulmonary arterial hypertension occurring secondary to pulmonary diseases such as chronic bronchitis, emphysema, kyphoscoliosis and conditions such as chronic mountain sickness.
  • Pulmonary hypertension is a serious medical condition that may lead to right ventricular hypertrophy, failure and death.
  • the term “right heart failure” relates to disorders such as cor pulmonale and congenital abnormalities of the heart. It will be appreciated that cor pulmonale often occurs secondary to certain lung diseases such as chronic bronchitis and emphysema. Congenital abnormalities of the heart include disorders, such as atrial septal defect, tetralogy of fallot, venticular septal defect and persistent ductus arteriosus.
  • the invention provides a method for the production of a pharmaceutical composition comprising the usage of HIF-2 ⁇ or a fragment thereof in a method to screen for molecules that are able to inhibit the development of pulmonary hypertension and further more mixing said molecule identified, or a derivative or homologue thereof, with a pharmaceutically acceptable carrier.
  • the invention provides in vivo evidence that HIF-2 ⁇ and its downstream target VEGF are critical for fetal lung maturation. Loss of HIF-2 ⁇ , absence of critical VEGF isoforms or inhibition of VEGF in utero all impaired lung maturation and caused RDS at birth due to insufficient surfactant production.
  • VEGF intra-amniotically to unborn fetuses or intra-tracheally after birth, it increases conversion of glycogen stores to surfactant, improves lung function, protects severely preterm mice against RDS and prolongs their survival, with a comparable efficiency as prenatal steroid treatment but without acute adverse effects.
  • the present invention demonstrates an important role for VEGF for the treatment of respiratory distress syndrome.
  • the invention provides the use of HIF-2 ⁇ or a fragment or homologue thereof for the manufacture of a medicament to treat neonatal respiratory distress syndrome.
  • the invention provides the use of VEGF or a fragment or homologue thereof, that is induced by HIF-2 ⁇ , for the manufacture of a medicament to treat neonatal respiratory distress syndrome.
  • the invention provides the use of VEGF 165 for the manufacture of a medicament to treat neonatal respiratory distress syndrome.
  • the term ‘medicament to treat’ relates to a composition comprising molecules as described above and a pharmaceutically acceptable carrier or excipient (both terms can be used interchangeably) to treat diseases as indicated above (pulmonary hypertension and respiratory distress syndrome).
  • the administration of a compound or a pharmaceutically acceptable salt thereof may be by way of oral, inhaled or parenteral administration. In a preferred embodiment inhaled administration is preferred.
  • the active compound may be administered alone or preferably formulated as a pharmaceutical composition.
  • An amount effective to treat the disorders hereinbefore described depends on the usual factors such as the nature and severity of the disorders being treated and the weight of the mammal.
  • a unit dose will normally contain 0.01 to 50 mg for example 0.01 to 10 mg, or 0.05 to 2 mg of the identified compound or a pharmaceutically acceptable salt thereof.
  • Unit doses will normally be administered once or more than once a day, for example 2, 3, or 4 times a day, more usually 1 to 3 times a day, such that the total daily dose is normally in the range of 0.0001 to 1 mg/kg; thus a suitable total daily dose for a 70 kg adult is 0.01 to 50 mg, for example 0.01 to 10 mg or more usually 0.05 to 10 mg.
  • the compound or a pharmaceutically acceptable salt thereof is administered in the form of a unit-dose composition, such as a unit dose oral, parenteral, or inhaled composition.
  • the compound (e.g. VEGF 165 ) or a pharmaceutically acceptable salt thereof is administered in an intra-uterine way (or intra-amniotically).
  • the compound (e.g. VEGF 165 ) or a pharmaceutically acceptable salt thereof is administered intra-tracheally.
  • Such compositions are prepared by admixture and are suitably adapted for oral, inhaled, intra-amniotic, intra-tracheal or parenteral administration, and as such may be in the form of tablets, capsules, oral liquid preparations, powders, granules, lozenges, reconstitutable powders, injectable and infusable solutions or suspensions or suppositories or aerosols.
  • Tablets and capsules for oral administration are usually presented in a unit dose, and contain conventional excipients such as binding agents, fillers, diluents, tabletting agents, lubricants, disintegrants, colourants, flavourings, and wetting agents.
  • the tablets may be coated according to well-known methods in the art.
  • Suitable fillers for use include cellulose, mannitol, lactose and other similar agents.
  • Suitable disintegrants include starch, polyvinylpyrrolidone and starch derivatives such as sodium starch glycollate.
  • Suitable lubricants include, for example, magnesium stearate.
  • Suitable pharmaceutically acceptable wetting agents include sodium lauryl sulphate.
  • solid oral compositions may be prepared by conventional methods of blending, filling, tabletting or the like. Repeated blending operations may be used to distribute the active agent throughout those compositions employing large quantities of fillers. Such operations are, of course, conventional in the art.
  • Oral liquid preparations may be in the form of, for example, aqueous or oily suspensions, solutions, emulsions, syrups, or elixirs, or may be presented as a dry product for reconstitution with water or other suitable vehicle before use.
  • Such liquid preparations may contain conventional additives such as suspending agents, for example sorbitol, syrup, methyl cellulose, gelatin, hydroxyethylcellulose, carboxymethyl cellulose, aluminium stearate gel or hydrogenated edible fats, emulsifying agents, for example lecithin, sorbitan monooleate, or acacia; non-aqueous vehicles (which may include edible oils), for example, almond oil, fractionated coconut oil, oily esters such as esters of glycerine, propylene glycol, or ethyl alcohol; preservatives, for example methyl or propyl p-hydroxybenzoate or sorbic acid, and if desired conventional flavouring or colouring agents.
  • suspending agents for example sorbitol, syrup, methyl cellulose, gelatin, hydroxyethylcellulose, carboxymethyl cellulose, aluminium stearate gel or hydrogenated edible fats, emulsifying agents, for example lecithin, sorbitan monooleate
  • Oral formulations also include conventional sustained release formulations, such as tablets or granules having an enteric coating.
  • compositions for inhalation are presented for administration to the respiratory tract as a snuff or an aerosol or solution for a nebulizer, or as a microfine powder for insufflation, alone or in combination with an inert carrier such as lactose.
  • the particles of active compound suitably have diameters of less than 50 microns, preferably less than 10 microns, for example between 1 and 5 microns, such as between 2 and 5 microns.
  • a favored inhaled dose will be in the range of 0.05 to 2 mg, for example 0.05 to 0.5 mg, 0.1 to 1 mg or 0.5 to 2 mg.
  • fluid unit dose forms are prepared containing a compound of the present invention and a sterile vehicle.
  • the active compound depending on the vehicle and the concentration, can be either suspended or dissolved.
  • Parenteral solutions are normally prepared by dissolving the compound in a vehicle and filter sterilising before filling into a suitable vial or ampoule and sealing.
  • adjuvants such as a local anaesthetic, preservatives and buffering agents are also dissolved in the vehicle.
  • the composition can be frozen after filling into the vial and the water removed under vacuum.
  • Parenteral suspensions are prepared in substantially the same manner except that the compound is suspended in the vehicle instead of being dissolved and sterilised by exposure to ethylene oxide before suspending in the sterile vehicle.
  • a surfactant or wetting agent is included in the composition to facilitate uniform distribution of the active compound.
  • small amounts of bronchodilators for example sympathomimetic amines such as isoprenaline, isoetharine, salbutamol, phenylephrine and ephedrine; xanthine derivatives such as theophylline and aminophylline and corticosteroids such as prednisolone and adrenal stimulants such as ACTH may be included.
  • the compositions will usually be accompanied by written or printed directions for use in the medical treatment concerned.
  • the present invention further provides a pharmaceutical composition for use in the treatment and/or prophylaxis of herein described disorders which comprises a molecule or a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable solvate thereof, and, if required, a pharmaceutically acceptable carrier thereof.
  • a pharmaceutically acceptable composition is an inhalation composition, suitably in unit dosage form. Such compositions may be prepared in the manner as hereinbefore described.
  • genetic constructs operably linked to HIF-2 ⁇ or to functional fragments thereof or genetic constructs operably linked to VEGF or homologues or fragments thereof can also be used to manufacture a medicament.
  • a ‘genetic construct’ means any genetic element, coding or non-coding or anti-sense genetic information.
  • Gene therapy means the treatment by the delivery of therapeutic nucleic acids to patient's cells. This is extensively reviewed in Lever and Goodfellow 1995 ; Br. Med Bull., 51, 1-242; Culver 1995; Ledley, F. D. 1995 . Hum. Gene Ther. 6, 1129. To achieve gene therapy there must be a method of delivering genes to the patient's cells and additional methods to ensure the effective production of any therapeutic genes.
  • Non-viral delivery There are two general approaches to achieve gene delivery; these are non-viral delivery and virus-mediated gene delivery.
  • virus-mediated gene delivery is the use of a virus-mediated gene delivery system with replication defective retroviruses to stably introduce genes into patient's cells.
  • PR-11 is a truncated form of PR-39, composed of these 11 amino acids (NH 2 -Arg-Arg-Arg-Pro-Arg-Pro-Pro-Tyr-Leu-Pro-Arg-COOH).
  • PR-11 is used for the manufacture of a medicament to treat respiratory distress syndrome.
  • PR-11 is administrated to fetus by intra-amniotic (in utero) or to preterms by intra-tracheal injection.
  • VEGF Intra-amniotic or intra-tracheal delivery of VEGF improves surfactant production and protects preterm newborns against RDS.
  • VEGF did not cause adverse effects on vascular leakage or bleeding in the lung, possibly because it barely crossed the alveolar epithelium.
  • steroids are often used to induce lung maturation but may cause serious adverse effects 6 .
  • the invention indicates that dexamethasone upregulates pulmonary VEGF expression in fetuses at low doses, but suppresses VEGF production at a high dose.
  • glucocorticoids can counteract the beneficial pneumotrophic effect of VEGF.
  • oxygen improves oxygenation of preterm infants with RDS but, since it also suppresses VEGF expression in alveolar type 2 pneumocytes 11 , it would deprive alveolar cells from pneumotrophic effects.
  • VEGF supplements can lower the toxicity of high oxygen concentrations in the neonate, as supported by indirect evidence that the protective effect of IL-13 against hyperoxic lung damage is mediated by VEGF 50 .
  • the present invention shows that the pneumotrophic effect of VEGF has a therapeutic potential for lung maturation in preterm infants at risk for RDS.
  • HIF-2 ⁇ +/ ⁇ HIF-2 ⁇ +/ ⁇
  • ES cells were screened by Southern blot analysis (probe A) for the presence of an additional 7.5-kb StuI band (homologous recombinant) besides the wild type 7-kb fragment ( FIG. 1 ).
  • HIF-2 ⁇ +/ ⁇ ES clones were also screened with a second restriction digest (KpnI+XhoI, probe B) to confirm that homozygous recombination occurred correctly at both flanks. 22 positive homologous recombinant HIF-2 ⁇ +/ ⁇ ES clones were obtained out of a total of 75 screened ES clones. HIF-2 ⁇ ⁇ / ⁇ ES cells with two inactivated HIF-2 ⁇ alleles were obtained by selection of heterozygous HIF-2 ⁇ +/ ⁇ ES cells on high G418 (1.8 mg/ml).
  • HIF-2 ⁇ +/+ two functional HIF-2 ⁇ alleles
  • WT wild type
  • Inactivation of the HIF-2 ⁇ gene was confirmed by reverse transcriptase polymerase chain reaction (RT-PCR) and immunoblot analysis ( FIG. 1 ).
  • heterozygous HIF-2 ⁇ +/ ⁇ ES clones were aggregated with diploid embryos and reimplanted in pseudo-pregnant recipient mice. High percentage chimaeras were selected from the offspring of these recipient mothers and crossed with wild types mice. When germline transmission occurred, heterozygous HIF-2 ⁇ +/ ⁇ mice were born from these matings. Offspring from intercrossings of HIF-2 ⁇ +/ ⁇ mice contained only wild types and heterozygotes, but no homozygous HIF-2 ⁇ ⁇ / ⁇ mice, indicating that deficiency of HIF-2 ⁇ causes lethality in the embryonic stage.
  • hypoxic HIF-2 ⁇ +/ ⁇ mice in which the fraction of arterioles containing both an IEL and a complete EEL was comparable with that of normoxic WT and HIF-2 ⁇ +/ ⁇ mice (Table 3), indicating that heterozygous deficiency of HIF-2 ⁇ completely abolished the hypoxic response in the development of pulmonary vascular remodeling.
  • Hypoxia-induced pulmonary vascular remodeling presence of elastic laminae.
  • Last row represents the total amount of vessels surrounded by elastic laminae. °Statistically significant (P ⁇ 0.05) versus normoxia. *Statistically significant (P ⁇ 0.05) versus WT.
  • Normoxia Hypoxia WT HIF-2 ⁇ +/ ⁇ WT HIF-2 ⁇ +/ ⁇ Partial SMC 1.32 ⁇ 0.05 1.27 ⁇ 0.10 1.23 ⁇ 0.10 1.21 ⁇ 0.04 coverage Complete SMC 0.65 ⁇ 0.06 0.60 ⁇ 0.12 1.43 ⁇ 0.09* 0.66 ⁇ 0.02 coverage 5.
  • HIF-2 ⁇ +/ ⁇ mice Hemodynamic measurements were performed to determine whether the decreased pulmonary vascular remodeling in HIF-2 ⁇ +/ ⁇ mice in hypoxia was associated with reduced pulmonary hypertension.
  • Chronic hypoxia (4 weeks) caused a significant increase of the mean and the systolic blood pressure in the right ventricle (RV) of WT mice (Table 5).
  • RV right ventricle
  • HIF-2 ⁇ +/ ⁇ mice showed comparable right ventricular blood pressures in normoxia and hypoxia, which were also comparable with pressures observed in normoxic WT mice.
  • Heart beating frequencies were comparable in WT and HIF-2 ⁇ +/ ⁇ mice under both normoxic and hypoxic conditions.
  • HIF-2 ⁇ regulates hypoxia-induced pulmonary vascular remodeling via mediating expression of ET-1 and VEGF.
  • TABLE 7 Gene expression analysis in lungs of WT and HIF-2 ⁇ +/ ⁇ mice. Values are means ⁇ SEM expressed as number of mRNA copies per 100 copies of HPRT (n is 5 to 6). °Statistically significant (P ⁇ 0.05) versus normoxia. *Statistically significant (P ⁇ 0.05) versus WT.
  • Wild type new-borns adapt quickly after birth to the postnatal life.
  • HIF-2 ⁇ ⁇ / ⁇ new-borns in contrast develop serious respiration problems shortly after birth.
  • Their breathing is slow and irregular accompanied with tirage caused by the diaphragm.
  • Their chest is lift upwards reflecting the use of accessory respiratory muscles (sternocleidomastoideus and platysma).
  • the neonates become cyanotic and eventually they die. This usually occurs within the first two hours of life.
  • the lungs appear collapsed in 50% of the HIF-2 ⁇ ⁇ / ⁇ new-borns.
  • the majority of the lungs of the other neonates appear retarded (branching structures instead of alveoli) and partially collapsed.
  • Neonatal lungs were taken out of the thorax without perfusion to avoid distension of the tissue by perfusion. Histological sections were used to calculate the percentage of the area covered by air in alveoli related to the total area covered by air and tissue.
  • RDS was not attributable to growth retardatation, respiratory muscle dysfunction, lung hypoplasia, impaired fluid clearance, hypoxic stress or other organ defects. Only the heart rate was found to be lower in HIF-2 ⁇ ⁇ / ⁇ fetuses, likely resulting from the reduced catecholamine production (confirming previous findings 23 ). However, administration of D,L -threo-3,4-dihydrophenylserine (DOPS; a substrate that is converted to noradrenaline) to pregnant HIF-2 ⁇ +/ ⁇ females increased the heart rate and reversed the lethality of HIF-2 ⁇ ⁇ / ⁇ embryos, but failed to reverse the fatal RDS after birth. Moreover, HIF-2 ⁇ +/ ⁇ mice, which had no signs of RDS, also had lower heart rates, making it unlikely that bradycardia or insufficient adrenergic stimulation is the primary cause of RDS.
  • DOPS D,L -threo-3,4-dihydrophenylserine
  • Immature lungs are characterised by thick alveolar septa, a less developed capillary bed and an immature alveolar epithelium.
  • the alveolar septa are thicker in HIF-2 ⁇ ⁇ / ⁇ neonates compared with wild types (13.6 micrometer compared with 8.1 micrometer in wild types), as shown in Table 12.
  • the alveolar epithelium consists of more immature, glycogen rich cells. (38.64 PAS-positive cells per millimeter alveolar epithelial lining in HIF-2 ⁇ ⁇ / ⁇ neonates versus 0.71 PAS-positive cells per millimeter alveolar epithelial lining in wild-type newborns, as shown in Table 13).
  • the lecithin/sphingomyeline ratio (3.53 ⁇ 0.16 in wild type lungs versus 2.84 ⁇ 0.23; p-value 0.0397) as well as the amount of phosphatidylcholine (184.736+/ ⁇ 21.82 nmol/10 mg sample in wild type versus 111.93+/ ⁇ 14.73 nmol/10 mg sample in homozygous; p-value 0.0244) is lowered in HIF-2 ⁇ ⁇ / ⁇ then in wild types.
  • Semi-thin sections show that the number of pneumocytes type 1 is declined and the number of pneumocytes type 2 is increased in the HIF-2 ⁇ / ⁇ .
  • the alveolar septa are to thick and have an abnormal composition. Ultrastructural analyse confirms an increased number pneumocytes type 2.
  • Sirius red staining increased and diffuse collagen deposition in the HIF-2 ⁇ / ⁇ .
  • MAC3 staining shows the presence of an increased number macrophages.
  • Similar numbers of CC10-positive Clara cells were present in bronchi and bronchioli in both genotypes, and no genotypic differences were found in the presence of neuroepithelial bodies, as evidenced by PGP9.5 staining.
  • Pulmonary development involves extensive vascular growth and remodeling, beginning during the pseudoglandular stage of development and continuing throughout all subsequent stages. Counting of vascular densities after thrombomoduline staining revealed that vascular development was comparable in both genotypes during the pseudoglandular and canalicular stages (11.5 ⁇ 0.7 blood vessels per alveolus in WT versus 12.4 ⁇ 1.3 vessels per alveolus in HIF-2 ⁇ ⁇ / ⁇ at E17.5).
  • HIF-2 ⁇ is involved in the remodeling and expansion of the distal capillaries in the lung, while it has a more neglectible role for vascular development in other organs.
  • VEGF mRNA is expressed in the alveolar epithelium in the perinatal period. This expression occurs at the same time as HIF-2 ⁇ expression in the alveolar epithelium. Also the protein level of VEGF gradually increases during the perinatal period (Table 15). At E16.5 there is no difference in the VEGF-protein level in wt and HIF-2 ⁇ / ⁇ . At E18.5, the HIF-2 ⁇ ⁇ / ⁇ fetus show a significantly decrease mRNA level (Table 16; Table 17). At protein level a 40 percent reduction of VEGF is observed in HIF-2 ⁇ ⁇ / ⁇ mice at 18.5 (276 (+/ ⁇ 7.3 in wild types versus 181.6+/ ⁇ 5.7 in HIF-2 ⁇ ⁇ / ⁇ foetus).
  • VEGF is regulated by HIF-2 ⁇ in the alveolar epithelium.
  • the VEGF-protein level further rises until P14.
  • In situ hybridisation shows that VEGF-mRNA is present in the alveolar epithelium of wild type as well as HIF-2 ⁇ / ⁇ lungs.
  • the alveolar epithelium expresses the VEGF receptor flk but not fit.
  • Elisa-kits R&D systems were used to determine quantitative the mouse vascular endothelial growth factor concentrations in lungs of E16.5 and E18.5 HIF-2 ⁇ ⁇ / ⁇ and wild types and in P0, P3, P8, P14 and adult wild type lungs (week 6).
  • VEGF ELISA different ages (pg/mg protein) age E16.5 E18.5 P0 P3 P8 P14 6 weeks WT 119 ⁇ 1.3 276 ⁇ 7.3 523 ⁇ 44.2 629 ⁇ 87.2 935 ⁇ 170.2 1102 ⁇ 66.5 614 ⁇ 64.1 HIF2 ⁇ / ⁇ 119 ⁇ 15.5 181.6 ⁇ 5.7 p-value 0.98 0.000003 13.
  • VEGF is a downsteam target of HIF-2 ⁇ we analyzed a number of previously generated VEGF mutant mouse strains.
  • the early embryonic lethality of VEGF deficient mice precluded analysis of their lung development.
  • VEGF 120/120 selectively expressing the VEGF 120 isoform (VEGF 120/120 ) 28 , died of RDS within a few hours after birth; their lungs were atelectatic and poorly aerated, and contained thick alveolar septa with abundant PAS-positive pneumocytes (cells per mm alveolar lining: 2 ⁇ 1 in WT neonates versus 51 ⁇ 20 in VEGF 120/120 littermates; N3-5; P ⁇ 0.05).
  • VEGF was produced by alveolar type 2 pneumocytes, while immunostaining revealed that VEGF was primarily present on the alveolar side of these cells.
  • ELISA ELISA
  • significant VEGF levels were detected in the bronchiolar alveolar lavage fluid of WT neonates (120 ⁇ 49 pg/mg protein).
  • No genotypic differences were detected in pulmonary expression of PIGF (a VEGF homologue that only binds to the VEGF receptor Flt-1 but not Flk-1 30 ) or angiopoietin-1 (another vascular-specific growth factor).
  • type 2 pneumocytes expressed both Flk-1 and HIF-2 ⁇ .
  • Fit-1 was detectable on capillaries and colocalized with the endothelial marker CD31 on adjacent sections, while the VEGF 165 -isoform selective receptor neuropilin-1 was undetectable.
  • hypoxia inducible factor 1 ⁇ The mRNA level of hypoxia inducible factor 1 ⁇ is not different in HIF-2 ⁇ ⁇ / ⁇ versus wild types.
  • HIF-2 ⁇ transcript levels were comparable in the lung, heart and kidneys in E16.5 WT fetuses. Thereafter, expression of HIF-2 ⁇ increased more than 5-fold in the lungs but less than 2-fold in the heart and kidneys.
  • Double-immunostaining for alkaline phosphatase (a marker of type 2 pneumocytes) and HIF-2 ⁇ revealed that alveolar type 2 pneumocytes abundantly expressed HIF-2 ⁇ , although other cells in alveolar septa, presumably endothelial and mesenchymal cells, also expressed HIF-2 ⁇ .
  • TGFbeta is increased in respiratory distress syndrome especially in the group predisponed for chronic lung disease. It is presumed that this higher TGFbeta levels are correlated with lung fibrosis.
  • HIF-2 ⁇ ⁇ / ⁇ lungs at E18.5 the TGFbeta mRNA is increased (40.7 versus 28.6 in wild type).
  • Platelet derived growth factor (PDGF) and its receptor (PDGFbetaR) are involved in the development of emphysematous lesion (as well in null mutations as in overexpression studies). PDGFis not different expressed in HIF-2 ⁇ ⁇ / ⁇ lungs.
  • the PDGFbetaR-mRNA level is increased in the HIF-2 ⁇ ⁇ / ⁇ lungs (Table 16).
  • the amount of hVEGF used for intra-amniotic injection was calculated as follows: the volume of the amniotic fluid and fetal lungs/guts was estimated to be ⁇ 500 ⁇ l. Taking into account that VEGF might be rapidly degraded or cleared, a dose of 500 ng was used to achieve an initial maximal dose of ⁇ 1 ⁇ g/ml, which is at least 10-fold higher than an effective concentration in vitro (when delivered by intra-amniotic route, a 10- to 100-fold excess of surfactant is administered).
  • VEGF vascular endothelial growth factor
  • VEGF was comparably effective to the glucocorticoid dexamethasone (0.8 mg/kg), administered to pregnant mice at gestational day 15.5 and 16.5.
  • intra-uterine VEGF improved fetal lung maturation and prevented RDS.
  • Intra-Tracheal VEGF Treatment Improves Pulmonary Maturation in Premature WT Newborns
  • E17.5 WT pups were delivered by caesarean section and treated intra-tracheally with saline or 500 ng hVEGF. Since injected pups were often cannibalized by their foster mothers, pups were monitored in isolation from a foster mother, which limited the clinical to maximum period of 10 hours (thereafter, pups started to become dehydrated and deprived of nutrients). After Caesarian delivery at E17.5, ⁇ 60% of preterm pups had an aerated lung area of less than 25% and died immediately after birth (category A), while another ⁇ 10% of preterm pups ventilated well and were normally oxygenated (category B).
  • preterm pups had an aerated lung area of 39 ⁇ 2% and suffered severe RDS. These pups were able to live for at least 6 hours although, ultimately, they succumbed to fatal exhaustion (category C). Only mice of class C were intratracheally injected with VEGF (500 ng/pup). To allocate pups to categories A, B and C, newborns were monitored for 30 minutes after Caesarean section, when their lung function and clinical condition was easily scored. Intra-tracheal VEGF administration prevented RDS in preterm pups of category C. Within 4-6 hours after VEGF administration, breathing became easier and more regular, skin color turned pink and pups moved more actively.
  • VEGF 500 ng/pup
  • VEGF intra-tracheally delivered VEGF could only reach ventilated lung areas ( ⁇ 50% of the lung), and taking the short duration of VEGF exposure (6 hours) and the young fetal age (E17.5) into consideration, the observed improvement of the clinical condition and lung maturation is remarkable.
  • Treatment with hVEGF did not increase the number of blood vessel (16.9 ⁇ 0.6 blood vessels/alveolus in hVEGF versus 17.2 ⁇ 0.5 in saline treated lungs).
  • the percentage of alveoli with a double layer of blood vessels was similar in both groups (97% ⁇ 2 in saline treated versus 92% ⁇ 2 in VEGF treated lungs).
  • the septa with a double layer of capillaries change into more thin septa with a single layer of blood vessels in close contact with the alveolar epithelium. Due to the increased expansion of the lungs in the hVEGF treated lungs, 8 ⁇ 2% of the alveoli in the hVEGF treated versus 0.2 ⁇ 0.2% of the alveoli in the saline treated group had a single layer of blood vessels. (p ⁇ 0.002)
  • VEGF receptor mediated lung maturation To analyze which VEGF receptor mediated lung maturation, neutralizing anti-Flk1 antibodies (preventing binding of VEGF to Flk-1 30,31 ) or anti-Flt-1 antibodies (blocking binding of VEGF and PIGF to Flt1 30,31 ) were intra-amniotically injected in WT fetuses at E17.5, and pups were delivered by Caesarean section at E18.5. Anti-Flt-1 antibodies were ineffective, but anti-Flk-1 antibodies prevented the thinning of the alveolar septa and the disappearance of PAS-positive cells (Table 3).
  • hVEGF recombinant human VEGF
  • Administration of recombinant human VEGF (hVEGF) to the airways may not necessarily cause the same adverse effects on vascular leakage as systemic administration or adenoviral VEGF gene transfer to the airway cells 33 , because the alveolar epithelium is less permeable than the endothelium.
  • 1 ⁇ g hVEGF was intra-tracheally administered, resulting in an estimated concentration of 10 pg/ml alveolar fluid, less than ⁇ 0.1% of the hVEGF was recovered in the plasma after 1 hour (500 ⁇ 60 pg/ml hVEGF as compared to 50 ⁇ 4 pg/ml murine VEGF in plasma of uninjected pups).
  • hVEGF plasma levels were undetectable ( ⁇ 2 pg/ml), confirming previous findings that VEGF remaines restricted to the alveolar compartment with minor spill-over to the interstitium and circulation 12 . Similar findings were obtained after intra-amniotic injection of hVEGF. Neither intra-amniotic nor intratracheal VEGF stimulated angiogenesis in alveolar septa (Table 18), vascular leakage (analyzed by Western blotting the amount of extravasated IgG in perfused lungs) or bronchial edema. There were also no microscopic abnormalities, leakiness or neovascular growth in the gastro-intestinal tract, placenta or fetal membrane after intra-amniotic delivery.
  • a genomic BAC clone containing the murine HIF-2 ⁇ gene was obtained from Research Genetics, Inc. (Huntsville, Ala., USA).
  • BAC-DNA preparations were performed using the following protocol: Bacterial pellets from overnight cultures were resuspended in 10 ml of 50 mM Tris-HCl pH 8.0 containing 10 mM EDTA (ethylene di-amino tetra-acetate) and 0.1 mg/ml RNase A. The cell suspension was lysed with 10 ml of 200 mM NaOH/1% SDS during 5 minutes and afterwards neutralized with 10 ml of 3.0 M potassium acetate (pH 5.5).
  • BAC-DNA was digested overnight with different restriction enzymes (single and double digests) and loaded on 1% agarose gels for electrophoresis.
  • DNA fragments in the vicinity of exon 2 of the HIF-2 ⁇ gene were digested with restriction enzymes, separated by gel electrophoresis, purified (Gel purification kit, Qiagen, Valencia, Calif., USA) and subcloned into pBluescript (Stratagene, La Jolla, Calif., USA) using the classical cloning techniques (ligation of DNA fragments at 14° C. using T4 DNA ligase (New England Biolabs Inc, Beverly, Mass., USA) in 50 mM Tris-Hcl pH 7.5 containing 1 mM DTT (dithiothreitol), 1 mM ATP and 10 mM MgCl 2 ; transformation of competent E. coli DH5 ⁇ cells with ligation products). These cloned DNA fragments were used for construction of a targeting vector with 10 kb homology to inactivate the HIF-2 ⁇ gene.
  • Undifferentiated embryonic stem (ES) cells were cultured in ES cell medium (Dulbecco's modified Eagle medium (DMEM) containing 100 IU/ml penicillin, 100 ⁇ g/ml streptomycin, 0.001% ⁇ -mercapto-ethanol, 2 mM glutamine, 1 mM Na-pyruvate, 1% of a 100 ⁇ solution of non-essential amino acids (NEAA) and 10 ng/ml leukemia inhibitory factor (LIF)), supplemented with 15% heat-inactivated fetal calf serum (FCS), on 0.1% gelatin-coated 10-cm culture dishes, containing mitomycin C-inactivated primary embryonic fibroblasts (mitomycin C-inactivation: 10 ⁇ g/ml for 3 hours).
  • DMEM Dulbecco's modified Eagle medium
  • FCS heat-inactivated fetal calf serum
  • ES cells were cultured in a humidified CO 2 incubator at 37° C., 90% humidity, and with 95% air and 5% CO 2 , and refed daily with fresh ES cell medium.
  • ES cells were washed with phosphate buffered saline (PBS), trypsinized with 0.25% trypsin and 0.02% EDTA for 5 minutes at 37° C. (5% CO 2 ) and split at a ratio 1:4 to 1:8.
  • PBS phosphate buffered saline
  • trypsinized with 0.25% trypsin and 0.02% EDTA
  • ES cells were frozen in ES cell medium containing 35% fetal calf serum and 10% dimethlyl sulfoxide (DMSO).
  • DMSO dimethlyl sulfoxide
  • DMEM fetal calf serum
  • NEAA penicillin/streptocmycin
  • Na-pyruvate glutamin
  • tissue culture-grade PBS LIF-ESGRO
  • trypsin/EDTA trypsin/EDTA
  • ⁇ -mercapto-ethanol and DMSO from Sigma, Bornem, Belgium
  • FCS from Hyclone, Logan, Utah, USA
  • MitomycinC Kyowa was from NTL, Brussel, Belgium.
  • the targeting vector was purified for electroporation in male R1 ES cells (provided by A. Nagy, Samuel Lunenfeld Institute, Toronto, Canada). Therefore, 20 ⁇ l linearized DNA (1 ⁇ g/ ⁇ l) and 6.10 6 ES cells, suspended in 0.8 ml PBS, were gently mixed in a 0.4-cm cuvette and immediately zapped at 250 V and 500 ⁇ F. The electroporated ES cells were then diluted in 10 ml ES cell culture medium and plated out on a 10-cm dish containing embryonic fibroblast feeder cells. For one electroporation, 5 cuvettes were prepared and processed in this way.
  • ES cell lysis was accomplished with Proteinase K (250 ⁇ g/ml; Boehringer Mannheim, Mannheim, Germany) in 100 mM Tris-HCl, 5 mM EDTA, 0.2% SDS and 0.2 M NaCl (0.5 ml lysis buffer per 48-well). Genomic DNA was precipitated after addition of an equal volume isopropanol and redissolved in 100-200 ⁇ l TE buffer (10 mM Tris-HCl pH 8.0 with 1 mM EDTA). Homologous recombinants were identified by diagnostic restriction digests/Southern blot analysis.
  • HIF-2 ⁇ +/ ⁇ ES cells were obtained after selection of HIF-2 ⁇ +/ ⁇ ES cells at high G418 concentration (1.8 mg/ml) during 8 days and identified by Southern blot analysis.
  • HIF-2 ⁇ +/ ⁇ ES clones were used for generation of transgenic mice via aggregation with diploid embryos. Therefore, 2.5 days old 8-cell stage embryos (morula stage) were collected from the oviduct of pregnant Swiss females and aggregated with HIF-2 ⁇ +/ ⁇ ES cell clumps containing 8-15 cells after removal of the zona pellucida with an acidic buffer (pH 2.5). Aggregated embryos were cultured during 2 days at 37° C.
  • HIF-2 ⁇ +/ ⁇ animals among the germline offspring were identified by Southern blot analysis of genomic DNA isolated from tails, and were intercrossed to obtain homozygous HIF-2 ⁇ ⁇ / ⁇ progeny.
  • RNA and antigen level respectively were confirmed at the RNA and antigen level respectively by reverse transcriptase PCR (RT-PCR) and immunoblot analysis.
  • Total RNA was extracted from wild type and HIF-2 ⁇ ⁇ / ⁇ ES cells using TRIZOL reagent (Life Technologies, Rockville, Md., USA), purified by phenol/chloroform extraction and precipitated with isopropanol.
  • DNase treatment of purified RNA was performed at 37° C. for 15 minutes with DNasel (Amersham Pharmacia Biotech, Uppsala, Sweden) in 40 mM Tris-HCl pH 7.5 and 6 mM MgCl 2 .
  • RNA was dissolved in RNase-free H 2 O and transcribed in cDNA with reverse transcriptase Superscript II (Life Technologies, Rockville, Md., USA).
  • Primer A (5′-ACGGAACTCCAGGTCCCTTGTCGCTGC-3′) (exon 1)
  • primer B (5′-ACAGGAGCTTATGTGTCCGAAGGAAGC-3′) (exon 2) were used for PCR on cDNA to amplify a HIF-2 ⁇ specific 531-bp fragment.
  • HPRT gene was used for normalization; therefore, a HPRT specific 374-bp fragment was amplified using primer hprt190 (5′-GTCGCCCTCTGTGTGCTCAAGGGGGGC-3′) and primer hprt563 (5′-AAACTTGTCTGGAATTTCAAATCCAAC-3′). Protein extraction and immunoblot analysis were performed as previously described.
  • ES cells were washed with ice-cold PBS, scraped from the dishes and homogenized with an IKA Ultra-turrax T8 homogenizer (Janke & Kunkel, Stauffen, Germany) in an 8 M urea buffer, containing 10% glycerol, 1% SDS (sodium dodecyl sulphate), 5 mM DTT and 10 mM Tris-HCl (pH 6.8).
  • IKA Ultra-turrax T8 homogenizer Janke & Kunkel, Stauffen, Germany
  • SDS sodium dodecyl sulphate
  • 5 mM DTT sodium dodecyl sulphate
  • 10 mM Tris-HCl pH 6.8
  • Protein concentrations of the extracts were quantified with the BCA protein assay (Pierce, Rockford, Ill., USA), which is based on the protein-mediated reduction of Cu 2+ to Cu 1+ in basic medium and the subsequent formation of a purple complex between Cu 1+ and bicinchoninic acid (BCA). This complex can be quantified by spectrophotometric analysis at 560 nm. Proteins were loaded on SDS/6% polyacrylamide gels for electrophoresis and afterwards transferred to Immobilon P membranes (Millipore, Bedford, Mass., USA) in 10 mM Tris, 100 mM glycine, 10% methanol and 0.05% SDS.
  • BCA protein assay Piercece, Rockford, Ill., USA
  • Membranes were blocked with PBS containing 5% fat-free milk and 0.1% Tween 20.
  • MoAb28b and 190b monoclonal antibodies (provided by Dr. P. Maxwell and Dr. P. Ratcliffe, Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK) were used for binding with HIF-1 ⁇ and HIF-2 ⁇ respectively.
  • Secondary goat anti-mouse antibodies (DAKO, Carpinteria, Calif., USA) conjugated with HRP (horseradish peroxidase) were used for detection by chemoluminescence. Immunoblot analysis was done in collaboration with Dr. P. H. Maxwell and Dr. P. J. Ratcliffe (Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK).
  • Taqman quantitative Real Time PCR was performed according to the manufacturer's protocol (Perkin Elmer, Zaventem, Belgium) using the Taqman universal PCR master mix kit, the ABI PRISM 7700 sequence detector, forward (F) and reverse (R) primers and probes, labeled with a fluorescent dye (FAM or JOE) and a quencher (TAMRA). Dyes were attached at the 5′ end while quenchers were attached to the 3′ end of the probes. Gene expression was standardized for the ⁇ -actin gene.
  • mice of 8 weeks old were weighed and placed in a tightly sealed chamber under normobaric hypoxia (10% O 2 ), which was maintained by a continuous inflow of 2 l/min N 2 and 2 l/min normal air (20% O 2 ).
  • the chamber was opened every 7 days for 10 minutes to clean the cages and to add food and water; mice were kept at these conditions for 4 weeks.
  • Control mice were kept for 4 weeks in normal air (20% O 2 ).
  • mice were weighed, anaesthetized and used for right ventricular pressure measurements. After collection of blood for hematocrit determination, hearts and lungs were dissected and used for gene expression analysis (real time PCR), histological analysis and determination of right ventricular hypertrophy.
  • mice were reoxygenated by returning them to room air for 1 hour and subsequently anaesthetized with 1.4 mg/kg urethane.
  • the right ventricular pressures were measured in anaesthetized ventilated mice by transthoracic puncture using high-fidelity pressure micromanometers (SPR-671; Millar Instruments, Houston, Tex.) at controlled normal body temperatures. Correct position of the pressure manometers in the right ventricle was verified by pressure readings and confirmed by postmortem examination.
  • mice were anaesthetized with pentobarbital (60 mg/kg) and perfused via cardiac puncture with saline to remove the blood followed by 1% paraformaldehyde in 0.1 M PBS (pH 7) at 100 cm H 2 O pressure. Subsequently, the trachea was cannulated and the airways were perfused with 1% paraformaldehyde in 0.1 M PBS (pH 7) at 25 cm H 2 O pressure. Lungs and heart were removed “en bloc” and post-fixed in 1% paraformaldehyde in 0.1 M PBS (pH 7) for 12 hours.
  • lungs and hearts were embedded in paraffin; 7 ⁇ m sections were used for histological stainings (haematoxylin and eosin). Hart's elastin staining was performed for visualization of internal elastic lamina (IEL) and external elastic lamina (EEL). Rabbit antibodies against murine thrombomodulin (a gift from Dr. R. Jackman, Harvard University, Boston, Mass., USA) and mouse antibodies against murine smooth muscle cell ⁇ -actin (DAKO, Carpinteria, Calif., USA) were used for stainings of endothelial and smooth muscle cells respectively (see section 10).
  • IEL internal elastic lamina
  • EEL external elastic lamina
  • Rabbit antibodies against murine thrombomodulin (a gift from Dr. R. Jackman, Harvard University, Boston, Mass., USA) and mouse antibodies against murine smooth muscle cell ⁇ -actin (DAKO, Carpinteria, Calif., USA) were used for stainings of endothelial and smooth muscle cells respectively
  • hypoxia-induced pulmonary vascular remodeling was assessed by counting the number of non-muscularized (only IEL), partially muscularized (IEL plus incomplete EEL) and fully muscularized (IEL and complete EEL) peripheral vessels (located distal to the bronchi) per 100 alveoli using the Quantimet Q600 imaging system (Leica imaging systems Ltd, Cambridge, UK) (30).
  • Elisa-kits were used to determine quantitative the mouse vascular endothelial growth factor concentrations in lungs of E16.5 and E18.5 HIF-2 ⁇ ⁇ / ⁇ and wild types and in P0, P3, P8, P14 and adult wild type lungs (week 6).
  • Human VEGF immunoassay was use to determine the quantity of injected hVEGF that reached the lungs.
  • Lungs of E18.5 and P0 or total foetus were fixed in 1% paraformaldehyde O.N. and paraffin embedded. Immunostaining for thrombomodulin, smooth muscle cell actin and apoprotein D, H&E and PAS-staining were performed on paraffin sections. Immunostaining for flk and fit were done on sections of unfixed in OCT frozen E18.5 lungs. Quantification of the number positive PAS cells and positive cells for apoprotein was calculated per millimetre of alveolar epithelial lining to correct for the collapse that occurred in the HIF-2 ⁇ ⁇ / ⁇ and VEGF 120/120 lungs. The septal thickness and the percentage of the total lungsurface (tissue+air) covered with air where determined using the Quantimet Q600 imaging system (Leica imaging systems Ltd, Cambridge, UK) on at least 5 optical fields.
  • Lung tissue was homogenized (Polytron PTA7 aggregate; Kinetica Benelux, Best, The Netherlands) in 1.9 ml of chloroform/methanol/water (5/10/4; v/v). The device was rinsed with 1.9 ml of the mixture and the rinse was added to the homogenate.
  • Pregnant HIF-2 ⁇ +/ ⁇ mothers at gestation day 18.5 were used for in vivo ultrasonic determination of embryonic heart rate as described 55 . Briefly, after maternal anesthesia (urethane i.p.), a midline abdominal incision was made and the uterine horns were exposed. To maintain stable embryonic and maternal temperature, the experimental set-up consisted of a temperature-regulating water bath (37° C.), filled with oxygenated (95% O 2 and 5% CO 2 ) Krebs-Henseleit buffer. The female was placed in a supine position in the water bath, allowing to expose the uterine horns on a horizontal plane.
  • Adrenal glands and organs of Zuckerkandl from neonates were snap frozen, stored at ⁇ 80° C., homogenized on ice in 1 ml of 0.1M perchloric acid containing 0.3% glutathione and 5 ⁇ g/l 3,4-dihydroxybenzylamine (as internal standard) and centrifuged at 4° C. for 10 minutes (19,000 g).
  • Catecholamines were eluted from alumina with 0.1 M perchloric acid, 0.3% glutathione, and quantitated by high pressure liquid chromatography 23 .
  • D,L -threo-3,4-dihydrophenylserine (DOPS, 1 mg/ml) was supplemented to the drinking water (containing 2 mg/ml ascorbic acid in bottles shielded from light and changed daily) of pregnant females from gestation day 8.5 until birth of the pups 23.
  • DOPS D,L -threo-3,4-dihydrophenylserine

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Hematology (AREA)
  • Zoology (AREA)
  • Urology & Nephrology (AREA)
  • Endocrinology (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • Marine Sciences & Fisheries (AREA)
  • Microbiology (AREA)
  • Vascular Medicine (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biotechnology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
US10/475,403 2001-04-24 2002-04-24 Use of hypoxia inducible factor 2alpha for curing neonatal respiratory distress syndrome and as a target for the treatment of pulmonary hypertension Abandoned US20050043227A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/226,771 US7307060B2 (en) 2001-04-24 2005-09-14 Hypoxia inducible factor 2α for curing neonatal respiratory distress syndrome and as a target for the treatment of pulmonary hypertension

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
EP01201481 2001-04-24
EP01201481.7 2001-04-24
EP01203073.0 2001-08-10
EP01203073 2001-08-10
EP02075077 2002-01-11
EP02075077.4 2002-01-11
PCT/EP2002/004479 WO2002086497A2 (fr) 2001-04-24 2002-04-24 Utilisation du facteur 2alpha induit par l'hypoxie pour le traitement du syndrome de detresse respiratoire du nouveau-ne et comme cible pour le traitement de l'hypertension pulmonaire

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/226,771 Division US7307060B2 (en) 2001-04-24 2005-09-14 Hypoxia inducible factor 2α for curing neonatal respiratory distress syndrome and as a target for the treatment of pulmonary hypertension

Publications (1)

Publication Number Publication Date
US20050043227A1 true US20050043227A1 (en) 2005-02-24

Family

ID=27224281

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/475,403 Abandoned US20050043227A1 (en) 2001-04-24 2002-04-24 Use of hypoxia inducible factor 2alpha for curing neonatal respiratory distress syndrome and as a target for the treatment of pulmonary hypertension
US11/226,771 Expired - Fee Related US7307060B2 (en) 2001-04-24 2005-09-14 Hypoxia inducible factor 2α for curing neonatal respiratory distress syndrome and as a target for the treatment of pulmonary hypertension

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/226,771 Expired - Fee Related US7307060B2 (en) 2001-04-24 2005-09-14 Hypoxia inducible factor 2α for curing neonatal respiratory distress syndrome and as a target for the treatment of pulmonary hypertension

Country Status (8)

Country Link
US (2) US20050043227A1 (fr)
EP (1) EP1407276B1 (fr)
AT (1) ATE446514T1 (fr)
AU (1) AU2002257794B8 (fr)
CA (1) CA2444578C (fr)
DE (1) DE60234100D1 (fr)
DK (1) DK1407276T3 (fr)
WO (1) WO2002086497A2 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060067937A1 (en) * 2004-09-24 2006-03-30 Karumanchi S A Methods of diagnosing and treating complications of pregnancy
US20070104707A1 (en) * 2004-09-24 2007-05-10 Beth Israel Deaconess Medical Center Methods of diagnosing and treating complications of pregnancy
WO2008030283A1 (fr) * 2006-05-31 2008-03-13 Beth Israel Deaconess Medical Center Méthodes pour le diagnostic et le traitement de complications associées à une grossesse
US20090286271A1 (en) * 2006-05-31 2009-11-19 Karumanchi Ananth S Methods of Diagnosing and Treating Complications of Pregnancy
US20130323318A1 (en) * 2010-12-10 2013-12-05 Indian Institute Of Technology Protein free surfactant composition for pulmonary diseases and a process for preparing the same
US20220015730A1 (en) * 2018-11-28 2022-01-20 Koninklijke Philips N.V. Most relevant x-ray image selection for hemodynamic simulation

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6737809B2 (en) * 2000-07-31 2004-05-18 Luxim Corporation Plasma lamp with dielectric waveguide
US7144999B2 (en) 2002-11-23 2006-12-05 Isis Pharmaceuticals, Inc. Modulation of hypoxia-inducible factor 1 alpha expression
US8124582B2 (en) * 2002-12-06 2012-02-28 Fibrogen, Inc. Treatment of diabetes
WO2005034929A2 (fr) * 2003-10-10 2005-04-21 Fibrogen, Inc. Remodelage et vascularisation de tissus
CN101534865A (zh) * 2005-10-19 2009-09-16 Ibc药品公司 生物活性装配体的制备方法及其用途
US8030446B2 (en) * 2006-02-10 2011-10-04 Trustees Of Dartmouth College Mutant proline-and-arginine rich peptides and methods for using the same
LT3043784T (lt) 2013-09-09 2019-08-12 Peloton Therapeutics, Inc. Arilo eteriai ir jų panaudojimo būdai
WO2016144825A1 (fr) 2015-03-11 2016-09-15 Peloton Therapeutics, Inc. Composés aromatiques, leurs et utilisations
US10512626B2 (en) 2015-03-11 2019-12-24 Peloton Therapeautics, Inc. Compositions for use in treating glioblastoma
EP3267792A4 (fr) * 2015-03-11 2018-09-26 Peloton Therapeutics, Inc. Compositions utilisées pour traiter l'hypertension artérielle pulmonaire
US10335388B2 (en) 2015-04-17 2019-07-02 Peloton Therapeutics, Inc. Combination therapy of a HIF-2-alpha inhibitor and an immunotherapeutic agent and uses thereof
WO2016198121A1 (fr) 2015-06-12 2016-12-15 Vib Vzw Moyens et méthodes de traitement de l'hypoplasie pulmonaire
WO2017143131A1 (fr) * 2016-02-19 2017-08-24 Cornell University Stabilisation de hif et prévention de maladie pulmonaire néonatale induite par hyperoxie
WO2019006155A1 (fr) 2017-06-28 2019-01-03 Children's Medical Center Corporation Promotion de la croissance pulmonaire
US20210015899A1 (en) * 2018-03-22 2021-01-21 The Children's Medical Center Corporation Methods and compositions relating to lung repair
CN112611877B (zh) * 2020-12-31 2022-09-16 四川大学华西医院 一款利用angptl4预测急性呼吸窘迫综合征患病及预后的试剂盒

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5695963A (en) * 1997-01-17 1997-12-09 Board Of Regents, The University Of Texas System Endothelial PAS domain protein
US5840693A (en) * 1995-03-01 1998-11-24 Ludwig Institute For Cancer Research Vascular endothelial growth factor-B

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU742365B2 (en) * 1997-03-14 2002-01-03 Selective Genetics, Inc. Adenoviral vectors with modified tropism
WO2000009657A2 (fr) * 1998-08-14 2000-02-24 President And Fellows Of Harvard College Methodes de modulation d'une angiogenese

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5840693A (en) * 1995-03-01 1998-11-24 Ludwig Institute For Cancer Research Vascular endothelial growth factor-B
US5695963A (en) * 1997-01-17 1997-12-09 Board Of Regents, The University Of Texas System Endothelial PAS domain protein

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060067937A1 (en) * 2004-09-24 2006-03-30 Karumanchi S A Methods of diagnosing and treating complications of pregnancy
US20070104707A1 (en) * 2004-09-24 2007-05-10 Beth Israel Deaconess Medical Center Methods of diagnosing and treating complications of pregnancy
US7740849B2 (en) 2004-09-24 2010-06-22 Beth Israel Deaconess Medical Center Use of compounds that bind soluble endoglin and SFLT-1 for the treatment of pregnancy related hypertensive disorders
US10413591B2 (en) 2004-09-24 2019-09-17 Beth Israel Deaconess Medical Center, Inc. Methods of diagnosing and treating complications of pregnancy
WO2008030283A1 (fr) * 2006-05-31 2008-03-13 Beth Israel Deaconess Medical Center Méthodes pour le diagnostic et le traitement de complications associées à une grossesse
US20090286271A1 (en) * 2006-05-31 2009-11-19 Karumanchi Ananth S Methods of Diagnosing and Treating Complications of Pregnancy
US20130323318A1 (en) * 2010-12-10 2013-12-05 Indian Institute Of Technology Protein free surfactant composition for pulmonary diseases and a process for preparing the same
US9345716B2 (en) * 2010-12-10 2016-05-24 Indian Institute Of Technology, Bombay Protein free surfactant composition for pulmonary diseases and a process for preparing the same
US20220015730A1 (en) * 2018-11-28 2022-01-20 Koninklijke Philips N.V. Most relevant x-ray image selection for hemodynamic simulation

Also Published As

Publication number Publication date
CA2444578C (fr) 2010-12-14
EP1407276B1 (fr) 2009-10-21
AU2002257794B2 (en) 2007-01-25
WO2002086497A3 (fr) 2004-01-08
EP1407276A2 (fr) 2004-04-14
AU2002257794A1 (en) 2002-11-05
US20060205645A1 (en) 2006-09-14
US7307060B2 (en) 2007-12-11
WO2002086497A2 (fr) 2002-10-31
CA2444578A1 (fr) 2002-10-31
AU2002257794B8 (en) 2007-02-15
ATE446514T1 (de) 2009-11-15
DK1407276T3 (da) 2009-12-21
DE60234100D1 (de) 2009-12-03

Similar Documents

Publication Publication Date Title
US7307060B2 (en) Hypoxia inducible factor 2α for curing neonatal respiratory distress syndrome and as a target for the treatment of pulmonary hypertension
Compernolle et al. Loss of HIF-2α and inhibition of VEGF impair fetal lung maturation, whereas treatment with VEGF prevents fatal respiratory distress in premature mice
Geraci et al. Pulmonary prostacyclin synthase overexpression in transgenic mice protects against development of hypoxic pulmonary hypertension
Muglia et al. Proliferation and differentiation defects during lung development in corticotropin-releasing hormone–deficient mice
US7108982B1 (en) Diagnostics and the therapeutics for macular degeneration
US7255857B2 (en) Lysyl oxidase-like 1 (LOXL1) and elastogenesis
Mura et al. Functions of type II pneumocyte-derived vascular endothelial growth factor in alveolar structure, acute inflammation, and vascular permeability
Garrett et al. Pravastatin therapy during preeclampsia prevents long-term adverse health effects in mice
JP2003507076A (ja) 心臓の遺伝子発現の調節におけるhdac4およびhdac5
Benjamin et al. Epithelial-derived inflammation disrupts elastin assembly and alters saccular stage lung development
US20130109738A1 (en) Control of Cardiac Growth, Differentiation and Hypertrophy
Coulombe et al. Endothelial Sash1 is required for lung maturation through nitric oxide signaling
Qu et al. Tie2 regulates endocardial sprouting and myocardial trabeculation
Yim et al. Disruption of the Arnt gene in endothelial cells causes hepatic vascular defects and partial embryonic lethality in mice
Floro et al. Gene–environment interaction demonstrates the vulnerability of the embryonic heart
Jung et al. Cardiac-specific overexpression of oxytocin receptor leads to cardiomyopathy in mice
Yi et al. Tyrosyl phosphorylation of PZR promotes hypertrophic cardiomyopathy in PTPN11-associated Noonan syndrome with multiple lentigines
WO2012096436A2 (fr) Animal à modèle de lésion pulmonaire provoquée par une exposition à l'hyperoxie, et trousse pour diagnostiquer une lésion pulmonaire provoquée par une exposition à l'hyperoxie par utilisation d'un marqueur taz
EP2434288B1 (fr) Moyens et procédés permettant de diagnostiquer et/ou de traiter un sujet courant le risque de développer une insuffisance cardiaque
Meng et al. Cardiac fibroblasts regulate cardiomyocyte hypertrophy through dynamic regulation of type I collagen
US20100055732A1 (en) Cardiac Hypertrophy
Wan et al. Disruption of Cdyl gene impairs mouse lung epithelium differentiation and maturation
US7304203B2 (en) Transgenic TIEG non-human animals
US20100280104A1 (en) Methods and kits for diagnosis and treatment of cell-cell junction related disorders
US20050235373A1 (en) IEX-1 knockout animals

Legal Events

Date Code Title Description
AS Assignment

Owner name: VLAAMS INTERUNIVERSITAIR INSTITUUT VOOR BIOTECHNOL

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:COMPERNOLLE, VEERLE;CARMELIET, PETER;REEL/FRAME:014868/0667;SIGNING DATES FROM 20031201 TO 20031217

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION