US20080199483A1 - Long lasting fusion peptide inhibitors of viral infection - Google Patents

Long lasting fusion peptide inhibitors of viral infection Download PDF

Info

Publication number
US20080199483A1
US20080199483A1 US11/877,221 US87722107A US2008199483A1 US 20080199483 A1 US20080199483 A1 US 20080199483A1 US 87722107 A US87722107 A US 87722107A US 2008199483 A1 US2008199483 A1 US 2008199483A1
Authority
US
United States
Prior art keywords
fmoc
peptide
group
seq
modified
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
US11/877,221
Inventor
Dominique P. Bridon
Robert S. Dufresne
Nissab Boudjellab
Martin Robitaille
Peter G. Milner
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.)
Conjuchem LLC
Original Assignee
ConjuChem Biotechnologies Inc
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=26832297&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20080199483(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by ConjuChem Biotechnologies Inc filed Critical ConjuChem Biotechnologies Inc
Priority to US11/877,221 priority Critical patent/US20080199483A1/en
Publication of US20080199483A1 publication Critical patent/US20080199483A1/en
Assigned to 4523482 CANADA INC. reassignment 4523482 CANADA INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CONJUCHEM BIOTECHNOLOGIES INC.
Assigned to CONJUCHEM BIOTECHNOLOGIES, INC. reassignment CONJUCHEM BIOTECHNOLOGIES, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: 4523482 CANADA INC.
Assigned to ADVANCED DIAGNOSTICS AND DISCOVERY reassignment ADVANCED DIAGNOSTICS AND DISCOVERY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CONJUCHEM BIOTECHNOLOGIES INC.
Assigned to CONJUCHEM, LLC reassignment CONJUCHEM, LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ADVANCED DIAGNOSTICS AND DISCOVERY
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Abstract

Peptides exhibiting anti-viral and anti-fusogenic activity are modified to provide greater stability and improved half-life in vivo. The selected peptides include fusion inhibitors DP178 and DP107 and related peptides and analogs thereof. The modified peptides are capable of forming covalent bonds with one or more blood components, preferably a mobile blood component.

Description

    FIELD OF THE INVENTION
  • This invention relates to modified peptides that are inhibitors of viral activity and/or exhibit antifusogenic properties. In particular, this invention relates to modified peptide inhibitors of human immunodeficiency virus (HIV), respiratory syncytial virus (RSV), human parainfluenza virus (HPV), measles virus (MeV), and simian immunodeficiency virus (SIV) with long duration of action for the treatment of the respective viral infections. The invention also relates to conjugates of the modified peptides and endogenous carriers, particularly conjugates of the modified peptides and various mobile blood components, particularly mobile endogenous proteins.
  • BACKGROUND OF THE INVENTION
  • Membrane fusion events, while commonplace in normal cell biological processes, are also involved in a variety of disease states, including, for example the entry of enveloped viruses into cells. Peptides are known that inhibit or otherwise disrupt membrane fusion-associated events, including, for example, inhibiting retroviral transmission to uninfected cells. As an example, the synthetic peptides DP-107 and DP-178 derived from separate domains within the human immunodeficiency virus type 1 (“HIV-1”) transmembrane (“TM”) glycoprotein gp41, are potent inhibitors of HIV-1 infection and HIV induced cell-cell fusion.
  • Lambert, et al., “Peptides from Conserved Regions of Paramyxovirus Fusion (F) Proteins are Potent Inhibitors of Viral Fusion,” Proc. Natl. Acad. Science U.S.A., Mar. 5, 1996, Vol. 93 (5), pp. 2186-91, discloses that the synthetic peptides DP-107 and DP-178 (T-20), derived from separate domains within the human immunodeficiency virus type 1 (HIV-1) transmembrane (TM) protein, gp41, are potent inhibitors of HIV-1 infection and fusion. Using a computer searching strategy (computerized antiviral searching technology, C.A.S.T.) based on the predicted secondary structure of DP-107 and DP-178 (T-20), Lambert, et al. identified conserved heptad repeat domains analogous to the DP-107 and DP-178 regions of HIV-1 gp41 within the glycoproteins of other fusogenic viruses. Antiviral peptides derived from three representative paramyxoviruses, respiratory syncytial virus (RSV), human parainfluenza virus type 3 (HPIV-3), and measles virus (MV) blocked homologous virus-mediated syncytium formation and exhibited EC50 values in the range 0.015-0.250 μM. Moreover, these peptides were highly selective for the virus of origin.
  • U.S. Pat. Nos. 6,013,263, 6,017,536 and 6,020,459 incorporated herein in their entirety, likewise disclose that the 36 amino acid peptide DP178 corresponding to amino acids 638 to 673 of gp41 from the HIV-1 isolate LAI (HIV-1LAI), and the 38 amino acid peptide DP107 corresponding to amino acids 558-595 of gp41 from the HIV-1LAI, both exhibit potent anti-HIV-1 activity.
  • While many of the anti-viral or anti-fusogenic peptides described in the art exhibit potent anti-viral and/or anti-fusogenic activity, these peptides suffer from short plasma half-lifes in vivo, primarily due to rapid serum clearance and peptidase and protease activity. This in turn greatly reduces the effective anti-viral activity of the peptides. There is therefore a need for a method of prolonging the half-life of existing anti-viral and/or anti-fusogenic peptides and providing for longer duration of action of these peptides in vivo.
  • SUMMARY OF THE INVENTION
  • The present invention meets these and other needs and is directed to modified peptides having anti-viral activity and/or anti-fusogenic activity. These modified peptides provide for an increased stability in vivo and a reduced susceptibility to peptidase or protease degradation. These modified peptides thereby minimize, e.g., the need for more frequent, or even continual, administration of the peptides. The products of varying embodiments of the present invention can be used, e.g., as a prophylactic against and/or treatment for infection of a number of viruses, including human immunodeficiency virus (HIV), human respiratory syncytial virus (RSV), human parainfluenza virus (HPV), measles virus (MeV) and simian immunodeficiency virus (SIV). Modification of other peptides involved in viral transfection (e.g., Hepatitis, Epstein Barr and other related viruses) is also within the scope of the invention.
  • This invention relates to chemically reactive modifications of peptides exhibiting anti-viral and/or anti-fusogenic activity such that the modified peptides can react with available functionalities on blood components to form stable covalent bonds. In one embodiment of the invention, the modified peptides comprise a reactive group which is reactive with amino groups, hydroxyl groups, or thiol groups on blood components to form stable covalent bonds. In another embodiment of the invention, the reactive group can be a maleimide which is reactive with a thiol group on a blood protein, including a mobile blood protein such as albumin.
  • In particular, the invention relates to such chemically reactive modifications of DP107 and DP178 peptides and analogs thereof, including peptides comprised of amino acid sequences from other (non-HIV) viruses that correspond to the gp41 region of HIV from which DP107 and DP178 are derived and that exhibit anti-viral or anti-fusogenic activity. More particularly, these peptides can exhibit anti-viral activity against, among others, human respiratory syncytial virus (RSV), human parainfluenza virus (HPV), measles virus (MeV) and simian immunodeficiency virus (SIV). The invention also relates to such chemically reactive modifications of the peptides of SEQ ID NO:1 to SEQ ID NO:86.
  • The invention also relates to compositions for use in the prevention and/or treatment of viral infection comprising a peptide that exhibits anti-viral activity modified with a reactive group as described. More particularly, the invention relates to such compositions for use in the prevention and/or treatment of AIDS, human respiratory syncytial virus (RSV), human parainfluenza virus (HPV), measles virus (MeV) and simian immunodeficiency virus (SIV).
  • BRIEF DESCRIPTION OF THE TABLES
  • The invention will be better understood by reference to the Tables, in which:
  • Table 1 lists the commonly occurring amino acids together with their one letter and three letter abbreviations, and common protecting groups.
  • Table 2 shows DP178 carboxy truncations.
  • Table 3 shows DP178 amino truncations.
  • Table 4 shows DP107 carboxy truncations.
  • Table 5 shows DP107 amino truncations.
  • Table 6 shows HIV-2NIHZ DP178 analog carboxy truncations.
  • Table 7 shows HIV-2NIHZ DP178 analog amino truncations.
  • Table 8 shows RSV F2 region DP107 analog carboxy truncations.
  • Table 9 shows RSV F2 region DP107 analog amino truncations.
  • Table 10 shows RSV F1 region DP178 analog carboxy truncations.
  • Table 11 shows RSV F1 region DP178 analog amino truncations.
  • Table 12 shows HPV3 F1 region DP 178 analog carboxy truncations.
  • Table 13 shows HPV3 F1 region DP 178 analog amino truncations.
  • Table 14 shows HPV3 F1 region DP 107 analog carboxy truncations.
  • Table 15 shows HPV3 F1 region DP 107 analog amino truncations.
  • Table 16 shows representative anti-RSV peptides.
  • Table 17 shows representative anti-HPV3 peptides.
  • Table 18 shows representative anti-SIV peptides.
  • Table 19 shows representative anti-MeV peptides.
  • BRIEF DESCRIPTION OF SEQUENCE LISTING
  • The invention will be better understood by reference to the Sequence Listing, in which:
  • SEQ ID NO:1 shows the peptide sequence of DP 178.
  • SEQ ID NO:2 shows the peptide sequence of DP 107
  • SEQ ID NO:3-9 show peptide sequences of certain DP178 analogs.
  • SEQ ID NO:10-30 show the peptide sequences of RSV F1 region and F2 region corresponding to DP178 and DP107, and representative anti-RSV peptides;
  • SEQ ID NO:31-62 show the peptide sequences of HPIV3 F1 region corresponding to DP178 and DP107, and representative anti-HPIV3 peptides;
  • SEQ ID NO:63-73 show peptide sequences of SIV corresponding to DP178 and representative anti-SIV peptides; and
  • SEQ ID NO:74-78 show peptide sequences of MeV corresponding to DP178 and representative anti-MeV peptides.
  • DETAILED DESCRIPTION OF THE INVENTION
  • To ensure a complete understanding of the invention the following definitions are provided:
  • Anti-viral peptides: As used herein, anti-viral peptides shall refer to peptides that inhibit viral infection of cells, by, for example, inhibiting cell-cell fusion or free virus infection. The route of infection may involve membrane fusion, as occurs in the case of enveloped viruses, or some other fusion event involving viral and cellular structures. Peptides that inhibit viral infection by a particular virus may be referenced with respect to that particular virus, e.g., anti-HIV peptide, anti-RSV peptide, etc.
  • Antifusogenic peptides: Antifusogenic peptides are peptides demonstrating an ability to inhibit or reduce the level of membrane fusion events between two or more entities, e.g., virus-cell or cell-cell, relative to the level of membrane fusion that occurs in the absence of the peptide.
  • HIV and anti-HIV peptides: The human immunodeficiency virus (HIV), which is responsible for acquired immune deficiency syndrome (AIDS), is a member of the lentivirus family of retroviruses. There are two prevalent types of HIV, HIV-1 and HIV-2, with various strain of each having been identified. HIV targets CD-4+ cells, and viral entry depends on binding of the HIV protein gp41 to CD4+ cell surface receptors. Anti-HIV peptides refer to peptides that exhibit anti-viral activity against HIV, including inhibiting CD-4+ cell infection by free virus and/or inhibiting HIV-induced syncytia formation between infected and uninfected CD-4+ cells.
  • SIV and anti-SIV peptides: Simian immunodeficiency viruses (SIV) are lentiviruses that cause acquired immunodeficiency syndrome (AIDS)-like illnesses in susceptible monkeys. Anti-SIV peptides are peptides that exhibit anti-viral activity against SIV, including inhibiting of infection of cells by the SIV virus and inhibiting syncytia formation between infected and uninfected cells.
  • RSV and anti-RSV peptides: Respiratory syncytial virus (RSV) is a respiratory pathogen, especially dangerous in infants and small children where it can cause bronchiolitis (inflammation of the small air passages) and pneumonia. RSVs are negative sense, single stranded RNA viruses and are members of the Paramyxoviridae family of viruses. The route of infection of RSV is typically through the mucous membranes by the respiratory tract, i.e., nose, throat, windpipe and bronchi and bronchioles. Anti-RSV peptides are peptides that exhibit anti-viral activity against RSV, including inhibiting mucous membrane cell infection by free RSV virus and syncytia formation between infection and uninfected cells.
  • HPV and anti-HPV peptides: Human parainfluenza virus (HPIV or HPV), like RSV, is another leading cause of respiratory tract disease, and like RSVs, are negative sense, single stranded RNA viruses that are members of the Paramyxoviridae family of viruses. There are four recognized serotypes of HPIV—HPIV-1, HPIV-2, HPIV-3 and HPIV-4. HPIV-1 is the leading cause of croup in children, and both HPIV-1 and HPIV-2 cause upper and lower respiratory tract illnesses. HPIV-3 is more often associated with bronchiolitis and pneumonia. Anti-HPV peptides are peptides that exhibit anti-viral activity against HPV, including inhibiting infection by free HPV virus and syncytia formation between infected and uninfected cells.
  • MeV and anti-Mev peptides: Measles virus (VM or MeV) is an enveloped negative, single-stranded RNA virus belonging to the Paramyxoviridae family of viruses. Like RSV and HPV, MeV causes respiratory disease, and also produces an immuno-suppression responsible for additional, opportunistic infections. In some cases, MeV can establish infection of the brain leading to severe neurlogical complications. Anti-MeV peptides are peptides that exhibit anti-viral activity against MeV, including inhibiting infection by free MeV virus and syncytia formation between infected and uninfected cells.
  • DP-178 and DP178 analogs: Unless otherwise indicated explicitly or by context, DP-178 means the 36 amino acid DP-178 peptide corresponding to amino acid residues 638-673 of the gp41 glycoprotein of HIV-1 isolate LAI (HIVLAI) and having the sequence:
  • YTSLIHSLIEESQNQQEKNEQELLELDKWASLWNWF (SEQ ID NO: 1)

    as well as truncations, deletions and/or insertions thereof. Truncations of the DP178 peptide may comprise peptides of between 3-36 amino acids. Deletions consist of the removal of one or more amino acid residues from the DP178 peptide, and may involve the removal of a single contiguous portion of the peptide sequence or multiple portions. Insertions may comprise single amino acid residues or stretches of residues and may be made at the carboxy or amino terminal end of the DP178 peptide or at a position internal to the peptide.
  • DP178 peptide analogs are peptides whose amino acid sequences are comprised of the amino acid sequences of peptide regions of viruses other than HIV-1LAI that correspond to the gp41 region from which DP178 was derived, as well as an truncations, deletions or insertions thereof. Such other viruses may include, but are not limited to, other HIV isolates such as HIV-2NIHZ, respiratory syncytial virus (RSV), human parainfluenza virus (HPV), simian immunodeficiency virus (SIV), and measles virus (MeV). DP178 analogs also refer to those peptide sequences identified or recognized by the ALLMOTI5, 107×178×4 and PLZIP search motifs described in U.S. Pat. Nos. 6,013,263, 6,017,536 and 6,020,459 and incorporated herein, having structural and/or amino acid motif similarity to DP178. DP178 analogs further refer to peptides described as “DP178-like” as that term is defined in U.S. Pat. Nos. 6,013,263, 6,017,536 and 6,020,459.
  • DP-107 and DP107 analogs: Unless otherwise indicated explicitly or by context, DP-107 means the 38 amino acid DP-107 peptide corresponding to amino acid residues 558-595 of the gp41 protein of HIV-1 isolate LAI (HIVLAI) and having the sequence:
  • (SEQ ID NO: 2)
    NNLLRAIEAQQHLLQLTVWQIKQLQARILAVERYLKDQ

    as well as truncations, deletions and/or insertions thereof. Truncations of the DP107 peptide may comprise peptides of between 3-38 amino acids. Deletions consist of the removal of one or more amino acid residues from the DP107 peptide, and may involve the removal of a single contiguous portion of the peptide sequence or multiple portions. Insertions may comprise single amino acid residues or stretches of residues and may be made at the carboxy or amino terminal end of the DP107 peptide or at a position internal to the peptide.
  • DP107 peptide analogs are peptides whose amino acid sequences are comprised of the amino acid sequences of peptide regions of viruses other than HIV-1LAI that correspond to the gp41 region from which DP107 was derived, as well as truncations, deletions and/or insertions thereof. Such other viruses may include, but are not limited to, other HIV isolates such as HIV-2NIHZ, respiratory syncytial virus (RSV), human parainfluenza virus (HPV), simian immunodeficiency virus (SIV), and measles virus (MeV). DP107 analogs also refer to those peptide sequences identified or recognized by the ALLMOTI5, 107×178×4 and PLZIP search motifs described in U.S. Pat. Nos. 6,013,263, 6,017,536 and 6,020,459 and incorporated herein, having structural and/or amino acid motif similarity to DP107. DP107 analogs further refer to peptides described as “DP107-like” as that term is defined in U.S. Pat. Nos. 6,013,263, 6,017,536 and 6,020,459.
  • Reactive Groups: Reactive groups are chemical groups capable of forming a covalent bond. Such reactive groups are coupled or bonded to a DP-107 or DP-178 peptide or analogs thereof or other anti-viral or anti-fusogenic peptide of interest. Reactive groups will generally be stable in an aqueous environment and will usually be carboxy, phosphoryl, or convenient acyl group, either as an ester or a mixed anhydride, or an imidate, thereby capable of forming a covalent bond with functionalities such as an amino group, a hydroxy or a thiol at the target site on mobile blood components. For the most part, the esters will involve phenolic compounds, or be thiol esters, alkyl esters, phosphate esters, or the like.
  • Functionalities: Functionalities are groups on blood components to which reactive groups on modified anti-viral peptides react to form covalent bonds. Functionalities include hydroxyl groups for bonding to ester reactive entities; thiol groups for bonding to maleimides, imidates and thioester groups; amino groups for bonding to carboxy, phosphoryl or acyl groups and carboxyl groups for bonding to amino groups.
  • Blood Components: Blood components may be either fixed or mobile. Fixed blood components are non-mobile blood components and include tissues, membrane receptors, interstitial proteins, fibrin proteins, collagens, platelets, endothelial cells, epithelial cells and their associated membrane and membraneous receptors, somatic body cells, skeletal and smooth muscle cells, neuronal components, osteocytes and osteoclasts and all body tissues especially those associated with the circulatory and lymphatic systems. Mobile blood components are blood components that do not have a fixed situs for any extended period of time, generally not exceeding 5, more usually one minute. These blood components are not membrane-associated and are present in the blood for extended periods of time and are present in a minimum concentration of at least 0.1 μg/ml. Mobile blood components include serum albumin, transferrin, ferritin and immunoglobulins such as IgM and IgG. The half-life of mobile blood components is at least about 12 hours.
  • Protective Groups: Protective groups are chemical moieties utilized to protect peptide derivatives from reacting with themselves. Various protective groups are disclosed herein and in U.S. Pat. No. 5,493,007, which is hereby incorporated by reference. Such protective groups include acetyl, fluorenylmethyloxycarbonyl (Fmoc), t-butyloxycarbonyl (Boc), benzyloxycarbonyl (CBZ), and the like. The specific protected amino acids are depicted in Table 1.
  • TABLE 1
    NATURAL AMINO ACIDS AND THEIR ABBREVIATIONS
    3-Letter 1-Letter
    Name Abbreviation Abbreviation Modified Amino Acids
    Alanine Ala A Fmoc-Ala-OH
    Arginine Arg R Fmoc-Arg(Pbf)-OH
    Asparagine Asn N Fmoc-Asn(Trt)-OH
    Aspartic acid Asp D Asp(tBu)-OH
    Cysteine Cys C Fmoc-Cys(Trt)
    Glutamic acid Glu E Fmoc-Glu(tBu)-OH
    Glutamine Gln Q Fmoc-Gln(Trt)-OH
    Glycine Gly G Fmoc-Gly-OH
    Histidine His H Fmoc-His(Trt)-OH
    Isoleucine Ile I Fmoc-Ile-OH
    Leucine Leu L Fmoc-Leu-OH
    Lysine Lys Z Boc-Lys(Aloc)-OH
    Lysine Lys X Fmoc-Lys(Aloc)-OH
    Lysine Lys K Fmoc-Lys(Mtt)-OH
    Methionine Met M Fmoc-Met-OH
    Phenylalanine Phe F Fmoc-Phe-OH
    Proline Pro P Fmoc-Pro-OH
    Serine Ser S Fmoc-Ser(tBu)-OH
    Threonine Thr T Fmoc-Thr(tBu)-OH
    Tryptophan Trp W Fmoc-Trp(Boc)-OH
    Tyrosine Tyr Y Boc-Tyr(tBu)-OH
    Valine Val V Fmoc-Val-OH
  • Linking Groups: Linking (spacer) groups are chemical moieties that link or connect reactive entities to antiviral or antifusogenic peptides. Linking groups may comprise one or more alkyl moeities, alkoxy moeity, alkenyl moeity, alkynyl moeity or amino moeity substituted by alkyl moeities, cycloalkyl moeity, polycyclic moeity, aryl moeity, polyaryl moeities, substituted aryl moeities, heterocyclic moeities, and substituted heterocyclic moeities. Linking groups may also comprise poly ethoxy amino acids, such as AEA ((2-amino) ethoxy acetic acid) or a preferred linking group AEEA ([2-(2-amino)ethoxy)]ethoxy acetic acid.
  • Sensitive Functional Groups—A sensitive functional group is a group of atoms that represents a potential reaction site on an antiviral and/or antifusogenic peptide. If present, a sensitive functional group may be chosen as the attachment point for the linker-reactive group modification. Sensitive functional groups include but are not limited to carboxyl, amino, thiol, and hydroxyl groups.
  • Modified Peptides—A modified peptide is an antiviral and/or antifusogenic peptide that has been modified by attaching a reactive group. The reactive group may be attached to the peptide either via a linking group, or optionally without using a linking group. It is also contemplated that one or more additional amino acids may be added to the peptide to facilitate the attachment of the reactive entity. Modified peptides may be administered in vivo such that conjugation with blood components occurs in vivo, or they may be first conjugated to blood components in vitro and the resulting conjugated peptide (as defined below) administered in vivo.
  • Conjugated Peptides—A conjugated peptide is a modified peptide that has been conjugated to a blood component via a covalent bond formed between the reactive group of the modified peptide and the functionalities of the blood component, with or without a linking group. As used throughout this application, the term “conjugated peptide” can be made more specific to refer to particular conjugated peptides, for example “conjugated DP178” or “conjugated DP107.”
  • Taking into account these definitions, the present invention takes advantage of the properties of existing anti-viral and antifusogenic peptides. The viruses that may be inhibited by the peptides include, but are not limited to all strains of viruses listed, e.g., in U.S. Pat. Nos. 6,013,263, 6,017,536 and 6,020,459 at Tables V-VII and IX-XIV therein. These viruses include, e.g., human retroviruses, including HIV-1, HIV-2, and human T-lympocyte viruses (HTLV-I and HTLV-II), and non-human retroviruses, including bovine leukosis virus, feline sarcoma virus, feline leukemia virus, simian immunodeficiency virus (SIV), simian sarcoma virus, simian leukemia, and sheep progress pneumonia virus. Non-retroviral viruses may also be inhibited by the peptides of the present invention, including human respiratory syncytial virus (RSV), canine distemper virus, Newcastle Disease virus, human parainfluenza virus (HPIV), influenza viruses, measles viruses (MeV), Epstein-Barr viruses, hepatitis B viruses, and simian Mason-Pfizer viruses. Non-enveloped viruses may also be inhibited by the peptides of the present invention, and include, but are not limited to, picornaviruses such as polio viruses, hepatitis A virus, enteroviruses, echoviruses, coxsackie viruses, papovaviruses such as papilloma virus, parvoviruses, adenoviruses, and reoviruses.
  • As an example, the mechanism of action of HIV fusion peptides has been described as discussed in the background section of this application and antiviral and antifusogenic properties of the peptides have been well established. A synthetic peptide corresponding to the carboxyl-terminal ectodomain sequence (for instance, amino acid residues 643-678 of HIV-1 class B, of the LAI strain or residues 638-673 from similar strain as well as residues 558-595) has been shown to inhibit virus-mediated cell-cell fusion completely at low concentration. The fusion peptide competes with the leucine zipper region of the native viral gp41 thus resulting in the interference of the fusion/infection of the virus into the cell.
  • The focus of the present invention is to modify a selected anti-viral and/or antifusogenic peptide with the DAC (Drug Activity Complex) technology to confer to this peptide improved bio-availability, extended half-life and better distribution through selective conjugation of the peptide onto a protein carrier but without modifying the peptide's anti-viral properties. The carrier of choice (but not limited to) for this invention would be albumin conjugated through its free thiol by an anti-viral and/or antifusogenic peptide modified with a maleimide moiety.
  • Several peptide sequences have been described in the literature as highly potent for the prevention of HIV-1 fusion/infection. As examples, peptide DP 178 binds to a conformation of gp41 that is relevant for fusion. Thus in one embodiment of the invention, DP178 and DP178-like peptides are modified. Likewise, other embodiments of the invention include modification of DP107 and DP107-like peptide for use against HIV, as well as peptides analagous to DP107 and DP178 that are found in RSV, HPV, MeV and SIV viruses.
  • 1. DP178 and DP107
  • A. DP178 Peptides
  • The DP178 peptide corresponds to amino acid residues 638 to 673 of the transmembrane protein gp41 from the HIV-1LAI isolate, and has the 36 amino acid sequence (reading from amino to carboxy terminus):
  • (SEQ ID NO: 1)
    NH2-YTSLIHSLIEESQNQQEKNEQELLELDKWASLWNWF-COOH
  • In addition to the full-length DP178 36-mer, the peptides of this invention include truncations of the DP178 peptide comprising peptides of between 3 and 36 amino acid residues (i.e., peptides ranging in size from a tripeptide to a 36-mer polypeptide), These truncated peptides are shown in Tables 2 and 3.
  • In addition amino acid substitutions of the DP178 peptide are also within the scope of the invention. HIV-1 and HIV-2 enveloped proteins are structurally distinct, but there exists a striking amino acid conservation within the DP178-corresponding regions of HIV-1 and HIV-2. The amino acid conservation is of a periodic nature, suggesting some conservation of structure and/or function. Therefore, one possible class of amino acid substitutions would include those amino acid changes which are predicted to stabilize the structure of the DP178 peptides of the invention. Utilizing the DP178 and DP178 analog sequences described herein, the skilled artisan can readily compile DP178 consensus sequences and ascertain from these, conserved amino acid residues which would represent preferred amino acid substitutions.
  • The amino acid substitutions may be of a conserved or non-conserved nature. Conserved amino acid substitutions consist of replacing one or more amino acids of the DP178 peptide sequence with amino acids of similar charge, size, and/or hydrophobicity characteristics, such as, for example, a glutamic acid (E) to aspartic acid (D) amino acid substitution. Non-conserved substitutions consist of replacing one or more amino acids of the DP178 peptide sequence with amino acids possessing dissimilar charge, size, and/or hydrophobicity characteristics, such as, for example, a glutamic acid (E) to valine (V) substitution.
  • Amino acid insertions of DP178 may consist of single amino acid residues or stretches of residues. The insertions may be made at the carboxy or amino terminal end of the DP178 or DP178 truncated peptides, as well as at a position internal to the peptide.
  • Such insertions will generally range from 2 to 15 amino acids in length. It is contemplated that insertions made at either the carboxy or amino terminus of the peptide of interest may be of a broader size range, with about 2 to about 50 amino acids being preferred. One or more such insertions may be introduced into DP178 or DP178 truncations, as long as such insertions result in peptides which may still be recognized by the 107×178×4, ALLMOTI5 or PLZIP search motifs described above.
  • Preferred amino or carboxy terminal insertions are peptides ranging from about 2 to about 50 amino acid residues in length, corresponding to gp41 protein regions either amino to or carboxy to the actual DP178 gp41 amino acid sequence, respectively. Thus, a preferred amino terminal or carboxy terminal amino acid insertion would contain gp41 amino acid sequences found immediately amino to or carboxy to the DP178 region of the gp41 protein.
  • Deletions of DP178 or DP178 truncations are also within the scope of this invention. Such deletions consist of the removal of one or more amino acids from the DP178 or DP178-like peptide sequence, with the lower limit length of the resulting peptide sequence being 4 to 6 amino acids.
  • Such deletions may involve a single contiguous or greater than one discrete portion of the peptide sequences. One or more such deletions may be introduced into DP178 or DP178 truncations, as long as such deletions result in peptides which may still be recognized by the 107×178×4, ALLMOTI5 or PLZIP search motifs described above.
  • B. DP107 Peptides
  • DP107 is a 38 amino acid peptide which exhibits potent antiviral activity, and corresponds to residues 558 to 595 of HIV-1LAI isolate transmembrane (TM) gp41 glycoprotein, as shown here:
  • (SEQ ID NO: 2)
    NH2-NNLLRAIEAQQHLLQLTVWQIKQLQARILAVERYLKDQ-COOH
  • In addition to the full-length DP107 38-mer, the DP107 peptides include truncations of the DP107 peptide comprising peptides of between 3 and 38 amino acid residues (i.e., peptides ranging in size from a tripeptide to a 38-mer polypeptide), These peptides are shown in Tables 4 and 5, below.
  • In addition, amino acid substitutions of the DP178 peptide are also within the scope of the invention. As for DP178, there also exists a striking amino acid conservation within the DP107-corresponding regions of HIV-1 and HIV-2, again of a periodic nature, suggesting conservation of structure and/or function. Therefore, one possible class of amino acid substitutions includes those amino acid changes predicted to stabilize the structure of the DP107 peptides of the invention. Utilizing the DP107 and DP107 analog sequences described herein, the skilled artisan can readily compile DP107 consensus sequences and ascertain from these, conserved amino acid residues which would represent preferred amino acid substitutions.
  • The amino acid substitutions may be of a conserved or non-conserved nature. Conserved amino acid substitutions consist of replacing one or more amino acids of the DP107 peptide sequence with amino acids of similar charge, size, and/or hydrophobicity characteristics, such as, for example, a glutamic acid (E) to aspartic acid (D) amino acid substitution. Non-conserved substitutions consist of replacing one or more amino acids of the DP107 peptide sequence with amino acids possessing dissimilar charge, size, and/or hydrophobicity characteristics, such as, for example, a glutamic acid (E) to valine (V) substitution.
  • Amino acid insertions may consist of single amino acid residues or stretches of residues. The insertions may be made at the carboxy or amino terminal end of the DP107 or DP107 truncated peptides, as well as at a position internal to the peptide.
  • Such insertions will generally range from 2 to 15 amino acids in length. It is contemplated that insertions made at either the carboxy or amino terminus of the peptide of interest may be of a broader size range, with about 2 to about 50 amino acids being preferred. One or more such insertions may be introduced into DP107 or DP107 truncations, as long as such insertions result in peptides which may still be recognized by the 107×178×4, ALLMOTI5 or PLZIP search motifs described above.
  • Preferred amino or carboxy terminal insertions are peptides ranging from about 2 to about 50 amino acid residues in length, corresponding to gp41 protein regions either amino to or carboxy to the actual DP107 gp41 amino acid sequence, respectively. Thus, a preferred amino terminal or carboxy terminal amino acid insertion would contain gp41 amino acid sequences found immediately amino to or carboxy to the DP107 region of the gp41 protein.
  • Deletions of DP107 or DP107 truncations are also within the scope of this invention. Such deletions consist of the removal of one or more amino acids from the DP107 or DP107-like peptide sequence, with the lower limit length of the resulting peptide sequence being 4 to 6 amino acids.
  • Such deletions may involve a single contiguous or greater than one discrete portion of the peptide sequences. One or more such deletions may be introduced into DP107 or DP107 truncations, as long as such deletions result in peptides which may still be recognized by the 107×178×4, ALLMOTI5 or PLZIP search motifs.
  • DP107 and DP107 truncations are more fully described in U.S. Pat. No. 5,656,480, which is incorporated herein by reference in its entirety
  • 2. DP107 and DP178 Analogs
  • Peptides corresponding to analogs of the DP178, DP178 truncations, DP107 and DP107 truncation sequences of the invention, described, above, may be found in other viruses, including, for example, non-HIV-1 enveloped viruses, non-enveloped viruses and other non-viral organisms.
  • Such DP178 and DP107 analogs may, for example, correspond to peptide sequences present in transmembrane (“TM”) proteins of enveloped viruses and may, correspond to peptide sequences present in non enveloped and nonviral organisms. Such peptides may exhibit antifusogenic activity, antiviral activity, most particularly antiviral activity which is specific to the virus in which their native sequences are found, or may exhibit an ability to modulate intracellular processes involving coiled-coil peptide structures.
  • A. DP178 Analogs
  • DP178 analogs are peptides whose amino acid sequences are comprised of the amino acid sequences of peptide regions of, for example, other (i.e., other than HIV-1) viruses that correspond to the gp41 peptide region from which DP178 was derived. Such viruses may include, but are not limited to, other HIV-1 isolates and HIV-2 isolates.
  • DP178 analogs derived from the corresponding gp41 peptide region of other (i.e., non HIV-1LAI) HIV-1 isolates may include, for example, peptide sequences as shown below.
  • (SEQ ID NO: 3)
    NH2-YTNTIYTLLEESQNQQEKNEQELLELDKWASLWNWF-COOH
    (SEQ ID NO: 4)
    NH2-YTGIIYNLLEESQNQQEKNEQELLELDKWANLWNWF-COOH
    (SEQ ID NO: 5)
    NH2-YTSLIYSLLEKSQIQQEKNEQELLELDKWASLWNWF-COOH
  • The peptides of SEQ ID NO:3, SEQ ID NO:4 and SEQ ID NO:5 are derived from HIV-1SF2, HIV-1RF, and HIV-1MN, respectively. Other DP178 analogs include those derived from HIV-2, including the peptides of SEQ ID NO:6 and SEQ ID NO:7, which are derived from HIV-2ROD and HIV-2NIHZ, respectively. Still other useful analogs include the peptides of SEQ ID NO:8 and SEQ ID NO:9, which have been demonstrated to exhibit anti-viral activity.
  • In the present invention, it is preferred that the DP178 analogs represent peptides whose amino acid sequences correspond to the DP178 region of the gp41 protein, it is also contemplated that the peptides disclosed herein may, additionally, include amino sequences, ranging from about 2 to about 50 amino acid residues in length, corresponding to gp41 protein regions either amino to or carboxy to the actual DP178 amino acid sequence.
  • Table 6 and Table 7 show some possible truncations of the HIV-2NIHZ DP178 analog, which may comprise peptides of between 3 and 36 amino acid residues (i.e., peptides ranging in size from a tripeptide to a 36-mer polypeptide). Peptide sequences in these tables are listed from amino (left) to carboxy (right) terminus.
  • B. Additional DP178 Analogs and DP107 Analogs
  • DP178 and DP107 analogs are recognized or identified, for example, by utilizing one or more of the 107×178×4, ALLMOTI5 or PLZIP computer-assisted search strategies described above. The search strategy identifies additional peptide regions which are predicted to have structural and/or amino acid sequence features similar to those of DP107 and/or DP178.
  • The search strategies are described fully in the example presented in Section 9 of U.S. Pat. Nos. 6,013,263, 6,017,536 and 6,020,459. While this search strategy is based, in part, on a primary amino acid motif deduced from DP107 and DP178, it is not based solely on searching for primary amino acid sequence homologies, as such protein sequence homologies exist within, but not between major groups of viruses. For example, primary amino acid sequence homology is high within the TM protein of different strains of HIV-1 or within the TM protein of different isolates of simian immunodeficiency virus (SIV).
  • The computer search strategy disclosed in U.S. Pat. Nos. 6,013,263, 6,017,536 and 6,020,459 successfully identified regions of proteins similar to DP107 or DP178. This search strategy was designed to be used with a commercially-available sequence database package, preferably PC/Gene.
  • In U.S. Pat. Nos. 6,013,263, 6,017,536 and 6,020,459, a series of search motifs, the 107×178×4, ALLMOTI5 and PLZIP motifs, were designed and engineered to range in stringency from strict to broad, with 107×178×4 being preferred. The sequences identified via such search motifs, such as those listed in Tables V-XIV, of U.S. Pat. Nos. 6,013,263, 6,017,536 and 6,020,459 and included in this application by incorporation by reference, potentially exhibit antifusogenic, such as antiviral, activity, may additionally be useful in the identification of antifusogenic, such as antiviral, compounds.
  • 3. Other Anti-Viral Peptides
  • A. Anti-RSV Peptides
  • Anti-RSV peptides include DP178 and/or DP107 analogs identified from corresponding peptide sequences in RSV which have further been identified to inhibit viral infection by RSV. Such peptides of interest include the peptides of Table 16 and peptides of SEQ ID NO:10 to SEQ ID NO:30. Of particular interest are the following peptides:
  • YTSVITIELSNIKENKCNGAKVKLIKQELDKYK (SEQ ID NO: 14)
    TSVITIELSNIKENKCNGAKVKLIKQELDKYKN (SEQ ID NO: 15)
    VITIELSNIKENKCNGAKVKLIKQELDKYKNAV (SEQ ID NO: 16)
    IALLSTNKAVVSLSNGVSVLTSKVLDLKNYIDK (SEQ ID NO: 29)

    The peptide of SEQ ID NO:10 is derived from the F2 region of RSV and was identified in U.S. Pat. Nos. 6,103,236 and 6,020,459 using the search motifs described as corresponding to DP107 and DP178 peptides (i.e., “DP107/178 like”). The peptides of SEQ ID NO:14 to SEQ ID NO:16 each have amino acid sequences contained within the peptide of SEQ ID NO:10 and each has been shown to exhibit anti-RSV activity, in particular, inhibiting fusion and syncytia formation between RSV-infected and uninfected Hep-2 cells at concentrations of less than 50 μg/ml.
  • The peptide of SEQ ID NO:11 is derived from the F1 region of RSV and was identified in U.S. Pat. Nos. 6,103,236 and 6,020,459 using the search motifs described as corresponding to DP107 (i.e., “DP107-like”). The peptide of SEQ ID NO:29 contains amino acid sequences contained within the peptide of SEQ ID NO:10 and likewise has been shown to exhibit anti-RSV activity, in particular, inhibiting fusion and syncytia formation between RSV-infected and uninfected Hep-2 cells at concentrations of less than 50 μg/ml.
  • B. Anti-HPIV Peptides
  • Anti-HPIV peptides include DP178 and/or DP107 analogs identified from corresponding peptide sequences in HPIV and which have further been identified to inhibit viral infection by HPIV. Such peptides of interest include the peptides of Table 17 and SEQ ID NO:31 to SEQ ID NO:62. Of particular interest are the following peptides:
  • (SEQ ID NO: 52)
    VEAKQARSDIEKLKEAIRDTNKAVQSVQSSIGNLI
    (SEQ ID NO: 58)
    RSDIEKLKEAIRDTNKAVQSVQSSIGNLIVAIKSV
    (SEQ ID NO: 35)
    NSVALDPIDISIELNKAKSDLEESKEWIRRSNQKL
    (SEQ ID NO: 38)
    ALDPIDISIELNKAKSDLEESKEWIRRSNQKLDSI
    (SEQ ID NO: 39)
    LDPIDISIELNKAKSDLEESKEWIRRSNQKLDSIG
    (SEQ ID NO: 40)
    DPIDISIELNKAKSDLEESKEWIRRSNQKLDSIGN
    (SEQ ID NO: 41)
    PIDISIELNKAKSDLEESKEWIRRSNQKLDSIGNW
    (SEQ ID NO: 42)
    IDISIELNKAKSDLEESKEWIRRSNQKLDSIGNWH
  • The peptide of SEQ ID NO:31 is derived from the F1 region of HPIV-3 and was identified in U.S. Pat. Nos. 6,103,236 and 6,020,459 using the search motifs described as corresponding to DP107 (i.e., “DP107-like”). The peptides of SEQ ID NO:52 and SEQ ID NO:58 each have amino acid sequences contained within the peptide of SEQ ID NO:30 and each has been shown to exhibit anti-HPIV-3 activity, in particular, inhibiting fusion and syncytia formation between HPIV-3-infected Hep2 cells and uninfected CV-1W cells at concentrations of less than 1 μg/ml.
  • The peptide of SEQ ID NO:32 is also derived from the F1 region of HPIV-3 and was identified in U.S. Pat. Nos. 6,103,236 and 6,020,459 using the search motifs described as corresponding to DP178 (i.e., “DP178-like”). The peptides of SEQ ID NO:35 and SEQ ID NO:38 to SEQ ID NO:42 each have amino acid sequences contained within the peptide of SEQ ID NO:32 and each also has been shown to exhibit anti-HPIV-3 activity, in particular, inhibiting fusion and syncytia formation between HPIV-3-infected Hep2 cells and uninfected CV-1W cells at concentrations of less than 1 μg/ml.
  • C. Anti-MeV Peptides
  • Anti-MeV peptides are DP178 and/or DP107 analogs identified from corresponding peptide sequences in measles virus (MeV) which have further been identified to inhibit viral infection by the measles virus. Such peptides of particular interest include the peptides of Table 19 and peptides of SEQ ID NO:74 to SEQ ID NO:86. Of particular interest are the peptides listed below.
  • HRIDLGPPISLERLDVGTNLGNAIAKLEAKELLE (SEQ ID NO: 77)
    IDLGPPISLERLDVGTNLGNAIAKLEAKELLESS (SEQ ID NO: 79)
    LGPPISLERLDVGTNLGNAIAKLEAKELLESSDQ (SEQ ID NO: 81)
    PISLERLDVGTNLGNAIAKLEAKELLESSDQILR (SEQ ID NO: 84)

    Sequences derived from measles virus were identified in U.S. Pat. Nos. 6,103,236 and 6,020,459 using the search motifs described as corresponding to DP178 (i.e., “DP178-like”). The peptides of SEQ ID NO:77, SEQ ID NO:79, SEQ ID NO:81 and SEQ ID NO:83 each have amino acid sequences so identified, and each has been shown to exhibit anti-MeV activity, in particular, inhibiting fusion and syncytia formation between MeV-infected Hep2 and uninfected Vero cells at concentrations of less than 1 μg/ml.
  • D. Anti-SIV Peptides
  • Anti-SIV peptides are DP178 and/or DP107 analogs identified from corresponding peptide sequences in SIV which have further been identified to inhibit viral infection by SIV. Such peptides of interest include the peptides of Table 18 and peptides of SEQ ID NO:63 to SEQ ID NO:73. Of particular interest are the following peptides:
  • (SEQ ID NO: 64)
    WQEWERKVDFLEENITALLEEAQIQQEKNMYELQK
    (SEQ ID NO: 65)
    QEWERKVDFLEENITALLEEAQIQQEKNMYELQKL
    (SEQ ID NO: 66)
    EWERKVDFLEENITALLEEAQIQQEKNMYELQKLN
    (SEQ ID NO: 67)
    WERKVDFLEENITALLEEAQIQQEKNMYELQKLNS
    (SEQ ID NO: 68)
    ERKVDFLEENITALLEEAQIQQEKNMYELQKLNSW
    (SEQ ID NO: 69)
    RKVDFLEENITALLEEAQIQQEKNMYELQKLNSWD
    (SEQ ID NO: 70)
    KVDFLEENITALLEEAQIQQEKNMYELQKLNSWDV
    (SEQ ID NO: 71)
    VDFLEENITALLEEAQIQQEKNMYELQKLNSWDVF
    (SEQ ID NO: 72)
    DFLEENITALLEEAQIQQEKNMYELQKLNSWDVFG
    (SEQ ID NO: 73)
    FLEENITALLEEAQIQQEKNMYELQKLNSWDVFGN

    Sequences derived from SIV transmembrane fusion protein were identified in U.S. Pat. Nos. 6,103,236 and 6,020,459 using the search motifs described as corresponding to DP178 (i.e., “DP178-like”). The peptides of SEQ ID NO:64 to SEQ ID NO:73 each have amino acid sequences so identified, and each has been shown to exhibit potent anti-SIV activity as crude peptides.
  • 4. Modification of Anti-Viral and Antifusogenic Peptides
  • The invention contemplates modifying peptides that exhibit anti-viral and/or antifusogenic activity, including such modifications of DP-107 and DP-178 and analogs thereof. Such modified peptides can react with the available reactive functionalities on blood components via covalent linkages. The invention also relates to such modifications, such combinations with blood components, and methods for their use. These methods include extending the effective therapeutic life of the conjugated anti-viral peptides derivatives as compared to administration of the unconjugated peptides to a patient. The modified peptides are of a type designated as a DAC (Drug Affinity Complex) which comprises the anti-viral peptide molecule and a linking group together with a chemically reactive group capable of reaction with a reactive functionality of a mobile blood protein. By reaction with the blood component or protein the modified peptide, or DAC, may be delivered via the blood to appropriate sites or receptors.
  • To form covalent bonds with functionalities on the protein, one may use as a reactive group a wide variety of active carboxyl groups, particularly esters, where the hydroxyl moiety is physiologically acceptable at the levels required to modify the peptide. While a number of different hydroxyl groups may be employed in these reactive groups, the most convenient would be N-hydroxysuccinimide or (NHS), N-hydroxy-sulfosuccinimide (sulfo-NHS). In preferred embodiments of this invention, the functionality on the protein will be a thiol group and the reactive group will be a maleimido-containing group such as gamma-maleimide-butyralamide (GMBA) or maleimidopropionic acid (MPA)
  • Primary amines are the principal targets for NHS esters. Accessible α-amine groups present on the N-termini of proteins react with NHS esters. However, α-amino groups on a protein may not be desirable or available for the NHS coupling. While five amino acids have nitrogen in their side chains, only the ε-amine of lysine reacts significantly with NHS esters. An amide bond is formed when the NHS ester conjugation reaction reacts with primary amines releasing N-hydroxysuccinimide as demonstrated in the schematic below.
  • Figure US20080199483A1-20080821-C00001
  • In the preferred embodiments of this invention, the functional group on this protein will be a thiol group and the chemically reactive group will be a maleimido-containing group such as MPA or GMBA (gamma-maleimide-butyralamide). The maleimido group is most selective for sulfhydryl groups on peptides when the pH of the reaction mixture is kept between 6.5 and 7.4. At pH 7.0, the rate of reaction of maleimido groups with sulfhydryls is 1000-fold faster than with amines. A stable thioether linkage between the maleimido group and the sulfhydryl is formed which cannot be cleaved under physiological conditions, as demonstrated in the following schematic.
  • Figure US20080199483A1-20080821-C00002
  • A. Specific Labeling.
  • Preferably, the modified peptides of this invention are designed to specifically react with thiol groups on mobile blood proteins. Such reaction is preferably established by covalent bonding of the peptide modified with a maleimide link (e.g. prepared from GMBS, MPA or other maleimides) to a thiol group on a mobile blood protein such as serum albumin or IgG.
  • Under certain circumstances, specific labeling with maleimides offers several advantages over non-specific labeling of mobile proteins with groups such as NHS and sulfo-NHS. Thiol groups are less abundant in vivo than amino groups. Therefore, the maleimide-modified peptides of this invention, i.e., maleimide peptides, will covalently bond to fewer proteins. For example, in albumin (the most abundant blood protein) there is only a single thiol group. Thus, peptide-maleimide-albumin conjugates will tend to comprise approximately a 1:1 molar ratio of peptide to albumin. In addition to albumin, IgG molecules (class II) also have free thiols. Since IgG molecules and serum albumin make up the majority of the soluble protein in blood they also make up the majority of the free thiol groups in blood that are available to covalently bond to maleimide-modified peptides.
  • Further, even among free thiol-containing blood proteins, including IgGs, specific labeling with maleimides leads to the preferential formation of peptide-maleimide-albumin conjugates, due to the unique characteristics of albumin itself. The single free thiol group of albumin, highly conserved among species, is located at amino acid residue 34 (Cys34). It has been demonstrated recently that the Cys34 of albumin has increased reactivity relative to free thiols on other free thiol-containing proteins. This is due in part to the very low pK value of 5.5 for the Cys34 of albumin. This is much lower than typical pK values for cysteine residues in general, which are typically about 8. Due to this low pK, under normal physiological conditions Cys34 of albumin is predominantly in the ionized form, which dramatically increases its reactivity. In addition to the low pK value of Cys34, another factor which enhances the reactivity of Cys34 is its location, which is in a crevice close to the surface of one loop of region V of albumin. This location makes Cys34 very available to ligands of all kinds, and is an important factor in Cys34's biological role as free radical trap and free thiol scavenger. These properties make Cys34 highly reactive with maleimide-peptides, and the reaction rate acceleration can be as much as 1000-fold relative to rates of reaction of maleimide-peptides with other free-thiol containing proteins.
  • Another advantage of peptide-maleimide-albumin conjugates is the reproducibility associated with the 1:1 loading of peptide to albumin specifically at Cys34. Other techniques, such as glutaraldehyde, DCC, EDC and other chemical activations of, e.g, free amines, lack this selectivity. For example, albumin contains 52 lysine residues, 25-30 of which are located on the surface of albumin and therefore accessible for conjugation. Activating these lysine residues, or alternatively modifying peptides to couple through these lysine residues, results in a heterogenous population of conjugates. Even if 1:1 molar ratios of peptide to albumin are employed, the yield will consist of multiple conjugation products, some containing 0, 1, 2 or more peptides per albumin, and each having peptides randomly coupled at any one or more of the 25-30 available lysine sites. Given the numerous possible combinations, characterization of the exact composition and nature of each conjugate batch becomes difficult, and batch-to-batch reproducibility is all but impossible, making such conjugates less desirable as a therapeutic. Additionally, while it would seem that conjugation through lysine residues of albumin would at least have the advantage of delivering more therapeutic agent per albumin molecule, studies have shown that a 1:1 ratio of therapeutic agent to albumin is preferred. In an article by Stehle, et al., “The Loading Rate Determines Tumor Targeting properties of Methotrexate-Albumin Conjugates in Rats,” Anti-Cancer Drugs, Vol. 8, pp. 677-685 (1988), incorporated herein in its entirety, the authors report that a 1:1 ratio of the anti-cancer methotrexate to albumin conjugated via glutaraldehyde gave the most promising results. These conjugates were preferentially taken up by tumor cells, whereas conjugates bearing 5:1 to 20:1 methotrexate molecules had altered HPLC profiles and were quickly taken up by the liver in vivo. It is postulated that at these higher ratios, conformational changes to albumin diminish its effectiveness as a therapeutic carrier.
  • Through controlled administration of maleimide-peptides in vivo, one can control the specific labeling of albumin and IgG in vivo. In typical administrations, 80-90% of the administered maleimide-peptides will label albumin and less than 5% will label IgG. Trace labeling of free thiols such as glutathione will also occur. Such specific labeling is preferred for in vivo use as it permits an accurate calculation of the estimated half-life of the administered agent.
  • In addition to providing controlled specific in vivo labeling, maleimide-peptides can provide specific labeling of serum albumin and IgG ex vivo. Such ex vivo labeling involves the addition of maleimide-peptides to blood, serum or saline solution containing serum albumin and/or IgG. Once conjugation has occurred ex vivo with the maleimide-peptides, the blood, serum or saline solution can be readministered to the patient's blood for in vivo treatment.
  • In contrast to NHS-peptides, maleimide-peptides are generally quite stable in the presence of aqueous solutions and in the presence of free amines. Since maleimide-peptides will only react with free thiols, protective groups are generally not necessary to prevent the maleimide-peptides from reacting with itself. In addition, the increased stability of the modified peptide permits the use of further purification steps such as HPLC to prepare highly purified products suitable for in vivo use. Lastly, the increased chemical stability provides a product with a longer shelf life.
  • B. Non-Specific Labeling.
  • The anti-viral peptides of the invention may also be modified for non-specific labeling of blood components. Bonds to amino groups will also be employed, particularly with the formation of amide bonds for non-specific labeling. To form such bonds, one may use as a chemically reactive group a wide variety of active carboxyl groups, particularly esters, where the hydroxyl moiety is physiologically acceptable at the levels required. While a number of different hydroxyl groups may be employed in these linking agents, the most convenient would be N-hydroxysuccinimide (NHS) and N-hydroxy-sulfosuccinimide (sulfo-NHS).
  • Other linking agents which may be utilized are described in U.S. Pat. No. 5,612,034, which is hereby incorporated herein.
  • The various sites with which the chemically reactive group of the modified peptides may react in vivo include cells, particularly red blood cells (erythrocytes) and platelets, and proteins, such as immunoglobulins, including IgG and IgM, serum albumin, ferritin, steroid binding proteins, transferrin, thyroxin binding protein, α-2-macroglobulin, and the like. Those receptors with which the modified peptides react, which are not long-lived, will generally be eliminated from the human host within about three days. The proteins indicated above (including the proteins of the cells) will remain at least three days, and may remain five days or more (usually not exceeding 60 days, more usually not exceeding 30 days) particularly as to the half life, based on the concentration in the blood.
  • For the most part, reaction will be with mobile components in the blood, particularly blood proteins and cells, more particularly blood proteins and erythrocytes. By “mobile” is intended that the component does not have a fixed situs for any extended period of time, generally not exceeding 5 minutes, more usually one minute, although some of the blood component may be relatively stationary for extended periods of time. Initially, there will be a relatively heterogeneous population of functionalized proteins and cells. However, for the most part, the population within a few days will vary substantially from the initial population, depending upon the half-life of the functionalized proteins in the blood stream. Therefore, usually within about three days or more, IgG will become the predominant functionalized protein in the blood stream.
  • Usually, by day 5 post-administration, IgG, serum albumin and erythrocytes will be at least about 60 mole %, usually at least about 75 mole %, of the conjugated components in blood, with IgG, IgM (to a substantially lesser extent) and serum albumin being at least about 50 mole %, usually at least about 75 mole %, more usually at least about 80 mole %, of the non-cellular conjugated components.
  • The desired conjugates of non-specific modified peptides to blood components may be prepared in vivo by administration of the modified peptides to the patient, which may be a human or other mammal. The administration may be done in the form of a bolus or introduced slowly over time by infusion using metered flow or the like.
  • If desired, the subject conjugates may also be prepared ex vivo by combining blood with modified peptides of the present invention, allowing covalent bonding of the modified peptides to reactive functionalities on blood components and then returning or administering the conjugated blood to the host. Moreover, the above may also be accomplished by first purifying an individual blood component or limited number of components, such as red blood cells, immunoglobulins, serum albumin, or the like, and combining the component or components ex vivo with the chemically reactive modified peptides. The functionalized blood or blood component may then be returned to the host to provide in vivo the subject therapeutically effective conjugates. The blood also may be treated to prevent coagulation during handling ex vivo.
  • 5. Synthesis of Modified Anti-Viral and Anti-Fusogenic Peptides
  • A. Peptide Synthesis
  • Anti-viral and/or anti-fusogenic peptides according to the present invention may be synthesized by standard methods of solid phase peptide chemistry known to those of ordinary skill in the art. For example, peptides may be synthesized by solid phase chemistry techniques following the procedures described by Steward and Young (Steward, J. M. and Young, J. D., Solid Phase Peptide Synthesis, 2nd Ed., Pierce Chemical Company, Rockford, Ill., (1984) using an Applied Biosystem synthesizer. Similarly, multiple peptide fragments may be synthesized then linked together to form larger peptides. These synthetic peptides can also be made with amino acid substitutions at specific locations.
  • For solid phase peptide synthesis, a summary of the many techniques may be found in J. M. Stewart and J. D. Young, Solid Phase Peptide Synthesis, W. H. Freeman Co. (San Francisco), 1963 and J. Meienhofer, Hormonal Proteins and Peptides, vol. 2, p. 46, Academic Press (New York), 1973. For classical solution synthesis see G. Schroder and K. Lupke, The Peptides, Vol. 1, Acacemic Press (New York). In general, these methods comprise the sequential addition of one or more amino acids or suitably protected amino acids to a growing peptide chain. Normally, either the amino or carboxyl group of the first amino acid is protected by a suitable protecting group. The protected or derivatized amino acid is then either attached to an inert solid support or utilized in solution by adding the next amino acid in the sequence having the complimentary (amino or carboxyl) group suitably protected and under conditions suitable for forming the amide linkage. The protecting group is then removed from this newly added amino acid residue and the next amino acid (suitably protected) is added, and so forth.
  • After all the desired amino acids have been linked in the proper sequence, any remaining protecting groups (and any solid support) are removed sequentially or concurrently to afford the final polypeptide. By simple modification of this general procedure, it is possible to add more than one amino acid at a time to a growing chain, for example, by coupling (under conditions which do not racemize chiral centers) a protected tripeptide with a properly protected dipeptide to form, after deprotection, a pentapeptide.
  • A particularly preferred method of preparing compounds of the present invention involves solid phase peptide synthesis wherein the amino acid α-N-terminal is protected by an acid or base sensitive group. Such protecting groups should have the properties of being stable to the conditions of peptide linkage formation while being readily removable without destruction of the growing peptide chain or racemization of any of the chiral centers contained therein. Suitable protecting groups are 9-fluorenylmethyloxycarbonyl (Fmoc), t-butyloxycarbonyl (Boc), benzyloxycarbonyl (Cbz), biphenylisopropyloxycarbonyl, t-amyloxycarbonyl, isobornyloxycarbonyl, α,α-dimethyl-3,5-dimethoxybenzyloxycarbonyl, o-nitrophenylsulfenyl, 2-cyano-t-butyloxycarbonyl, and the like. The 9-fluorenyl-methyloxycarbonyl (Fmoc) protecting group is particularly preferred for the synthesis of the peptides of the present invention. Other preferred side chain protecting groups are, for side chain amino groups like lysine and arginine, 2,2,5,7,8-pentamethylchroman-6-sulfonyl (pmc), nitro, p-toluenesulfonyl, 4-methoxybenzene-sulfonyl, Cbz, Boc, and adamantyloxycarbonyl; for tyrosine, benzyl, o-bromobenzyloxycarbonyl, 2,6-dichlorobenzyl, isopropyl, t-butyl (t-Bu), cyclohexyl, cyclopenyl and acetyl (Ac); for serine, t-butyl, benzyl and tetrahydropyranyl; for histidine, trityl, benzyl, Cbz, p-toluenesulfonyl and 2,4-dinitrophenyl; for tryptophan, formyl; for asparticacid and glutamic acid, benzyl and t-butyl and for cysteine, triphenylmethyl (trityl).
  • In the solid phase peptide synthesis method, the α-C-terminal amino acid is attached to a suitable solid support or resin. Suitable solid supports useful for the above synthesis are those materials which are inert to the reagents and reaction conditions of the stepwise condensation-deprotection reactions, as well as being insoluble in the media used. The preferred solid support for synthesis of α-C-terminal carboxy peptides is 4-hydroxymethylphenoxymethyl-copoly(styrene-1% divinylbenzene). The preferred solid support for α-C-terminal amide peptides is the 4-(2′,4′-dimethoxyphenyl-Fmoc-aminomethyl)phenoxyacetamidoethyl resin available from Applied Biosystems (Foster City, Calif.). The α-C-terminal amino acid is coupled to the resin by means of N,N′-dicyclohexylcarbodiimide (DCC), N,N′-diisopropylcarbodiimide (DIC) or O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluronium-hexafluorophosphate (HBTU), with or without 4-dimethylaminopyridine (DMAP), 1-hydroxybenzotriazole (HOBT), benzotriazol-1-yloxy-tris(dimethylamino)phosphonium-hexafluorophosphate (BOP) or bis(2-oxo-3-oxazolidinyl)phosphine chloride (BOPCl), mediated coupling for from about 1 to about 24 hours at a temperature of between 10° and 50° C. in a solvent such as dichloromethane or DMF.
  • When the solid support is 4-(2′,4′-dimethoxyphenyl-Fmoc-aminomethyl)phenoxy-acetamidoethyl resin, the Fmoc group is cleaved with a secondary amine, preferably piperidine, prior to coupling with the α-C-terminal amino acid as described above. The preferred method for coupling to the deprotected 4-(2′,4′-dimethoxyphenyl-Fmoc-aminomethyl)phenoxy-acetamidoethyl resin is O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluroniumhexafluoro-phosphate (HBTU, 1 equiv.) and 1-hydroxybenzotriazole (HOBT, 1 equiv.) in DMF. The coupling of successive protected amino acids can be carried out in an automatic polypeptide synthesizer as is well known in the art. In a preferred embodiment, the α-N-terminal amino acids of the growing peptide chain are protected with Fmoc. The removal of the Fmoc protecting group from the α-N-terminal side of the growing peptide is accomplished by treatment with a secondary amine, preferably piperidine. Each protected amino acid is then introduced in about 3-fold molar excess, and the coupling is preferably carried out in DMF. The coupling agent is normally O-benzotriazol-1-yl-N,N,N′,N′-tetramethyluroniumhexafluorophosphate (HBTU, 1 equiv.) and 1-hydroxybenzotriazole (HOBT, 1 equiv.).
  • At the end of the solid phase synthesis, the polypeptide is removed from the resin and deprotected, either in successively or in a single operation. Removal of the polypeptide and deprotection can be accomplished in a single operation by treating the resin-bound polypeptide with a cleavage reagent comprising thioanisole, water, ethanedithiol and trifluoroacetic acid. In cases wherein the α-C-terminal of the polypeptide is an alkylamide, the resin is cleaved by aminolysis with an alkylamine. Alternatively, the peptide may be removed by transesterification, e.g. with methanol, followed by aminolysis or by direct transamidation. The protected peptide may be purified at this point or taken to the next step directly. The removal of the side chain protecting groups is accomplished using the cleavage cocktail described above. The fully deprotected peptide is purified by a sequence of chromatographic steps employing any or all of the following types: ion exchange on a weakly basic resin (acetate form); hydrophobic adsorption chromatography on underivitized polystyrene-divinylbenzene (for example, Amberlite XAD); silica gel adsorption chromatography; ion exchange chromatography on carboxymethylcellulose; partition chromatography, e.g. on Sephadex G-25, LH-20 or countercurrent distribution; high performance liquid chromatography (HPLC), especially reverse-phase HPLC on octyl- or octadecylsilyl-silica bonded phase column packing.
  • Molecular weights of these ITPs are determined using Fast Atom Bombardment (FAB) Mass Spectroscopy.
  • (1) N-Terminal Protective Groups
  • As discussed above, the term “N-protecting group” refers to those groups intended to protect the α-N-terminal of an amino acid or peptide or to otherwise protect the amino group of an amino acid or peptide against undesirable reactions during synthetic procedures. Commonly used N-protecting groups are disclosed in Greene, “Protective Groups In Organic Synthesis,” (John Wiley & Sons, New York (1981)), which is hereby incorporated by reference. Additionally, protecting groups can be used as pro-drugs which are readily cleaved in vivo, for example, by enzymatic hydrolysis, to release the biologically active parent. α-N-protecting groups comprise loweralkanoyl groups such as formyl, acetyl (“Ac”), propionyl, pivaloyl, t-butylacetyl and the like; other acyl groups include 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o-nitrophenoxyacetyl, -chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4-bromobenzoyl, 4-nitrobenzoyl and the like; sulfonyl groups such as benzenesulfonyl, p-toluenesulfonyl and the like; carbamate forming groups such as benzyloxycarbonyl, p-chlorobenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p-nitrobenzyloxycarbonyl, 2-nitrobenzyloxycarbonyl, p-bromobenzyloxycarbonyl, 3,4-dimethoxybenzyloxycarbonyl, 3,5-dimethoxybenzyloxycarbonyl, 2,4-dimethoxybenzyloxycarbonyl, 4-ethoxybenzyloxycarbonyl, 2-nitro-4,5-dimethoxybenzyloxycarbonyl, 3,4,5-trimethoxybenzyloxycarbonyl, 1-(p-biphenylyl)-1-methylethoxycarbonyl, α,α-dimethyl-3,5-dimethoxybenzyloxycarbonyl, benzhydryloxycarbonyl, t-butyloxycarbonyl (Boc), diisopropylmethoxycarbonyl, isopropyloxycarbonyl, ethoxycarbonyl, methoxycarbonyl, allyloxycarbonyl, 2,2,2,-trichloroethoxycarbonyl, phenoxycarbonyl, 4-nitrophenoxycarbonyl, fluorenyl-9-methoxycarbonyl, cyclopentyloxycarbonyl, adamantyloxycarbonyl, cyclohexyloxycarbonyl, phenylthiocarbonyl and the like; arylalkyl groups such as benzyl, triphenylmethyl, benzyloxymethyl, 9-fluorenylmethyloxycarbonyl (Fmoc) and the like and silyl groups such as trimethylsilyl and the like.
  • (2) Carboxy Protective Groups
  • As discussed above, the term “carboxy protecting group” refers to a carboxylic acid protecting ester or amide group employed to block or protect the carboxylic acid functionality while the reactions involving other functional sites of the compound are performed. Carboxy protecting groups are disclosed in Greene, “Protective Groups in Organic Synthesis” pp. 152-186 (1981), which is hereby incorporated by reference. Additionally, a carboxy protecting group can be used as a pro-drug whereby the carboxy protecting group can be readily cleaved in vivo, for example by enzymatic hydrolysis, to release the biologically active parent. Such carboxy protecting groups are well known to those skilled in the art, having been extensively used in the protection of carboxyl groups in the penicillin and cephalosporin fields as described in U.S. Pat. Nos. 3,840,556 and 3,719,667, the disclosures of which are hereby incorporated herein by reference. Representative carboxy protecting groups are C1-C8 loweralkyl (e.g., methyl, ethyl or t-butyl and the like); arylalkyl such as phenethyl or benzyl and substituted derivatives thereof such as alkoxybenzyl or nitrobenzyl groups and the like; arylalkenyl such as phenylethenyl and the like; aryl and substituted derivatives thereofsuch as 5-indanyl and the like; dialkylaminoalkyl such as dimethylaminoethyl and the like); alkanoyloxyalkyl groups such as acetoxymethyl, butyryloxymethyl, valeryloxymethyl, isobutyryloxymethyl, isovaleryloxymethyl, 1-(propionyloxy)-1-ethyl, 1-(pivaloyloxyl)-1-ethyl, 1-methyl-1-(propionyloxy)-1-ethyl, pivaloyloxymethyl, propionyloxymethyl and the like; cycloalkanoyloxyalkyl groups such as cyclopropylcarbonyloxymethyl, cyclobutylcarbonyloxymethyl, cyclopentylcarbonyloxymethyl, cyclohexylcarbonyloxymethyl and the like; aroyloxyalkyl such as benzoyloxymethyl, benzoyloxyethyl and the like; arylalkylcarbonyloxyalkyl such as benzylcarbonyloxymethyl, 2-benzylcarbonyloxyethyl and the like; alkoxycarbonylalkyl or cycloalkyloxycarbonylalkyl such as methoxycarbonylmethyl, cyclohexyloxycarbonylmethyl, 1-methoxycarbonyl-1-ethyl and the like; alkoxycarbonyloxyalkyl or cycloalkyloxycarbonyloxyalkyl such as methoxycarbonyloxymethyl, t-butyloxycarbonyloxymethyl, 1-ethoxycarbonyloxy-1-ethyl, 1-cyclohexyloxycarbonyloxy-1-ethyl and the like; aryloxycarbonyloxyalkyl such as 2-(phenoxycarbonyloxy)ethyl, 2-(5-indanyloxycarbonyloxy)ethyl and the like; alkoxyalkylcarbonyloxyalkyl such as 2-(1-methoxy-2-methylpropan-2-oyloxy)ethyl and like; arylalkyloxycarbonyloxyalkyl such as 2-(benzyloxycarbonyloxy)ethyl and the like; arylalkenyloxycarbonyloxyalkyl such as 2-(3-phenylpropen-2-yloxycarbonyloxy)ethyl and the like; alkoxycarbonylaminoalkyl such as t-butyloxycarbonylaminomethyl and the like; alkylaminocarbonylaminoalkyl such as methylaminocarbonylaminomethyl and the like; alkanoylaminoalkyl such as acetylaminomethyl and the like; heterocycliccarbonyloxyalkyl such as 4-methylpiperazinylcarbonyloxymethyl and the like; dialkylaminocarbonylalkyl such as dimethylaminocarbonylmethyl, diethylaminocarbonylmethyl and the like; (5-(loweralkyl)-2-oxo-1,3-dioxolen-4-yl)alkyl such as (5-t-butyl-2-oxo-1,3-dioxolen-4-yl)methyl and the like; and (5-phenyl-2-oxo-1,3-dioxolen-4-yl)alkyl such as (5-phenyl-2-oxo-1,3-dioxolen-4-yl)methyl and the like.
  • Representative amide carboxy protecting groups are aminocarbonyl and loweralkylaminocarbonyl groups.
  • Preferred carboxy-protected compounds of the invention are compounds wherein the protected carboxy group is a loweralkyl, cycloalkyl or arylalkyl ester, for example, methyl ester, ethyl ester, propyl ester, isopropyl ester, butyl ester, sec-butyl ester, isobutyl ester, amyl ester, isoamyl ester, octyl ester, cyclohexyl ester, phenylethyl ester and the like or an alkanoyloxyalkyl, cycloalkanoyloxyalkyl, aroyloxyalkyl or an arylalkylcarbonyloxyalkyl ester. Preferred amide carboxy protecting groups are loweralkylaminocarbonyl groups. For example, aspartic acid may be protected at the α-C-terminal by an acid labile group (e.g. t-butyl) and protected at the β-C-terminal by a hydrogenation labile group (e.g. benzyl) then deprotected selectively during synthesis.
  • B. Peptide Modification
  • The manner of producing the modified peptides of the present invention will vary widely, depending upon the nature of the various elements comprising the peptide. The synthetic procedures will be selected so as to be simple, provide for high yields, and allow for a highly purified stable product. Normally, the chemically reactive group will be created at the last stage of the synthesis, for example, with a carboxyl group, esterification to form an active ester. Specific methods for the production of modified peptides of the present invention are described below.
  • Specifically, the selected peptide is first assayed for anti-viral activity, and then is modified with the linking group only at either the N-terminus, C-terminus or interior of the peptide. The anti-viral activity of this modified peptide-linking group is then assayed. If the anti-viral activity is not reduced dramatically (i.e., reduced less than 10-fold), then the stability of the modified peptide-linking group is measured by its in vivo lifetime. If the stability is not improved to a desired level, then the peptide is modified at an alternative site, and the procedure is repeated until a desired level of anti-viral and stability is achieved.
  • More specifically, each peptide selected to undergo modification with a linker and a reactive entity group will be modified according to the following criteria: if a terminal carboxylic group is available on the peptide and is not critical for the retention of anti-viral activity, and no other sensitive functional group is present on the peptide, then the carboxylic acid will be chosen as attachment point for the linker-reactive group modification. If the terminal carboxylic group is involved in anti-viral activity, or if no carboxylic acids are available, then any other sensitive functional group not critical for the retention of anti-viral activity will be selected as the attachment point for the linker-reactive entity modification. If several sensitive functional groups are available on a a peptide, a combination of protecting groups will be used in such a way that after addition of the linker/reactive entity and deprotection of all the protected sensitive functional groups, retention of anti-viral activity is still obtained. If no sensitive functional groups are available on the peptide, or if a simpler modification route is desired, synthetic efforts will allow for a modification of the original peptide in such a way that retention of anti-viral is maintained. In this case the modification will occur at the opposite end of the peptide
  • An NHS derivative may be synthesized from a carboxylic acid in absence of other sensitive functional groups in the peptide. Specifically, such a peptide is reacted with N-hydroxysuccinimide in anhydrous CH2Cl2 and EDC, and the product is purified by chromatography or recrystallized from the appropriate solvent system to give the NHS derivative.
  • Alternatively, an NHS derivative may be synthesized from a peptide that contains an amino and/or thiol group and a carboxylic acid. When a free amino or thiol group is present in the molecule, it is preferable to protect these sensitive functional groups prior to perform the addition of the NHS derivative. For instance, if the molecule contains a free amino group, a transformation of the amine into a Fmoc or preferably into a tBoc protected amine is necessary prior to perform the chemistry described above. The amine functionality will not be deprotected after preparation of the NHS derivative. Therefore this method applies only to a compound whose amine group is not required to be freed to induce the desired anti-viral effect. If the amino group needs to be freed to retain the original properties of the molecule, then another type of chemistry described below has to be performed.
  • In addition, an NHS derivative may be synthesized from a peptide containing an amino or a thiol group and no carboxylic acid. When the selected molecule contains no carboxylic acid, an array of bifunctional linkers can be used to convert the molecule into a reactive NHS derivative. For instance, ethylene glycol-bis(succinimydylsuccinate) (EGS) and triethylamine dissolved in DMF and added to the free amino containing molecule (with a ratio of 10:1 in favor of EGS) will produce the mono NHS derivative. To produce an NHS derivative from a thiol derivatized molecule, one can use N-[-maleimidobutyryloxy]succinimide ester (GMBS) and triethylamine in DMF. The maleimido group will react with the free thiol and the NHS derivative will be purified from the reaction mixture by chromatography on silica or by HPLC.
  • An NHS derivative may also be synthesized from a peptide containing multiple sensitive functional groups. Each case will have to be analyzed and solved in a different manner. However, thanks to the large array of protecting groups and bifunctional linkers that are commercially available, this invention is applicable to any peptide with preferably one chemical step only to modify the peptide (as described above) or two steps (as described above involving prior protection of a sensitive group) or three steps (protection, activation and deprotection). Under exceptional circumstances only, would multiple steps (beyond three steps) synthesis be required to transform a peptide into an active NHS or maleimide derivative.
  • A maleimide derivative may also be synthesized from a peptide containing a free amino group and a free carboxylic acid. To produce a maleimide derivative from a amino derivatized molecule, one can use N-[γ-maleimidobutyryloxy]succinimide ester (GMBS) and triethylamine in DMF. The succinimide ester group will react with the free amino and the maleimide derivative will be purified from the reaction mixture by crystallization or by chromatography on silica or by HPLC.
  • Finally, a maleimide derivative may be synthesized from a peptide containing multiple other sensitive functional groups and no free carboxylic acids. When the selected molecule contains no carboxylic acid, an array of bifunctional crosslinking reagents can be used to convert the molecule into a reactive NHS derivative. For instance maleimidopropionic acid (MPA) can be coupled to the free amine to produce a maleimide derivative through reaction of the free amine with the carboxylic group of MPA using HBTU/HOBt/DIEA activation in DMF.
  • Many other commercially available heterobifunctional crosslinking reagents can alternatively be used when needed. A large number of bifunctional compounds are available for linking to entities. Illustrative reagents include: azidobenzoyl hydrazide, N-[4-(p-azidosalicylamino)butyl]-3′-[2′-pyridyldithio)propionamide), bis-sulfosuccinimidyl suberate, dimethyl adipimidate, disuccinimidyl tartrate, N-y-maleimidobutyryloxysuccinimide ester, N-hydroxy sulfosuccinimidyl-4-azidobenzoate, N-succinimidyl [4-azidophenyl]-1,3′-dithiopropionate, N-succinimidyl [4-iodoacetyl]aminobenzoate, glutaraldehyde, and succinimidyl 4-[N-maleimidomethyl]cyclohexane-1-carboxylate.
  • 6. Uses of Modified Anti-Viral Peptides
  • Modified anti-viral peptides of the invention may be used as a therapeutic agent in the treatment of patients who are suffering from viral infection, and can be administered to patients according to the methods described below and other methods known in the art. Effective therapeutic dosages of the modified peptides may be determined through procedures well known by those in the art and will take into consideration any concerns over potential toxicity of the peptide.
  • The modified peptides can also be administered prophylactically to previously uninfected individuals. This can be advantageous in cases where an individual has been subjected to a high risk of exposure to a virus, as can occur when individual has been in contact with an infected individual where there is a high risk of viral transmission. This can be expecially advantageous where there is known cure for the virus, such as the HIV virus. As a example, prophylactic administration of a modified anti-HIV peptide would be advantageous in a situation where a health care worker has been exposed to blood from an HIV-infected individual, or in other situations where an individual engaged in high-risk activities that potentially expose that individual to the HIV virus.
  • 7. Administration of Modified Anti-Viral and Anti-Fusogenic Peptides
  • Generally, the modified peptides will be administered in a physiologically acceptable medium, e.g. deionized water, phosphate buffered saline (PBS), saline, aqueous ethanol or other alcohol, plasma, proteinaceous solutions, mannitol, aqueous glucose, alcohol, vegetable oil, or the like. Other additives which may be included include buffers, where the media are generally buffered at a pH in the range of about 5 to 10, where the buffer will generally range in concentration from about 50 to 250 mM, salt, where the concentration of salt will generally range from about 5 to 500 mM, physiologically acceptable stabilizers, and the like. The compositions may be lyophilized for convenient storage and transport.
  • The subject modified peptides will for the most part be administered parenterally, such as intravenously (IV), intraarterially (IA), intramuscularly (IM), subcutaneously (SC), or the like. Administration may in appropriate situations be by transfusion. In some instances, where reaction of the functional group is relatively slow, administration may be oral, nasal, rectal, transdermal or aerosol, where the nature of the conjugate allows for transfer to the vascular system. Usually a single injection will be employed although more than one injection may be used, if desired. The modified peptides may be administered by any convenient means, including syringe, trocar, catheter, or the like.
  • The particular manner of administration will vary depending upon the amount to be administered, whether a single bolus or continuous administration, or the like. Preferably, the administration will be intravascularly, where the site of introduction is not critical to this invention, preferably at a site where there is rapid blood flow, e.g., intravenously, peripheral or central vein. Other routes may find use where the administration is coupled with slow release techniques or a protective matrix. The intent is that the modified peptide be effectively distributed in the blood, so as to be able to react with the blood components. The concentration of the conjugate will vary widely, generally ranging from about 1 pg/ml to 50 mg/ml. The total administered intravascularly will generally be in the range of about 0.1 mg/ml to about 10 mg/ml, more usually about 1 mg/ml to about 5 mg/ml.
  • By bonding to long-lived components of the blood, such as immunoglobulin, serum albumin, red blood cells and platelets, a number of advantages ensue. The activity of the peptide is extended for days to weeks. Only one administration need be given during this period of time. Greater specificity can be achieved, since the active compound will be primarily bound to large molecules, where it is less likely to be taken up intracellularly to interfere with other physiological processes.
  • 8. Monitoring the Presence of Modified Peptides
  • The blood of the mammalian host may be monitored for the presence of the modified peptide compound one or more times. By taking a portion or sample of the blood of the host, one may determine whether the peptide has become bound to the long-lived blood components in sufficient amount to be therapeutically active and, thereafter, the level of the peptide compound in the blood. If desired, one may also determine to which of the blood components the peptide is bound. This is particularly important when using non-specific modified peptides. For specific maleimide-modified peptides, it is much simpler to calculate the half life of serum albumin and IgG.
  • A. Immuno Assays
  • Another aspect of this invention relates to methods for determining the concentration of the anti-viral peptides and/or analogs, or their derivatives and conjugates in biological samples (such as blood) using antibodies specific for the peptides, peptide analogs or their derivatives and conjugates, and to the use of such antibodies as a treatment for toxicity potentially associated with such peptides, analogs, and/or their derivatives or conjugates. This is advantageous because the increased stability and life of the peptides in vivo in the patient might lead to novel problems during treatment, including increased possibility for toxicity.
  • The use of anti-therapeutic agent antibodies, either monoclonal or polyclonal, having specificity for a particular peptide, peptide analog or derivative thereof, can assist in mediating any such problem. The antibody may be generated or derived from a host immunized with the particular peptide, analog or derivative thereof, or with an immunogenic fragment of the agent, or a synthesized immunogen corresponding to an antigenic determinant of the agent. Preferred antibodies will have high specificity and affinity for native, modified and conjugated forms of the peptide, peptide analog or derivative. Such antibodies can also be labeled with enzymes, fluorochromes, or radiolables.
  • Antibodies specific for modified peptides may be produced by using purified peptides for the induction of peptide-specific antibodies. By induction of antibodies, it is intended not only the stimulation of an immune response by injection into animals, but analogous steps in the production of synthetic antibodies or other specific binding molecules such as screening of recombinant immunoglobulin libraries. Both monoclonal and polyclonal antibodies can be produced by procedures well known in the art.
  • The anti-peptide antibodies may be used to treat toxicity induced by administration of the modified peptide, analog or derivative thereof, and may be used ex vivo or in vivo. Ex vivo methods would include immuno-dialysis treatment for toxicity employing anti-therapeutic agent antibodies fixed to solid supports. In vivo methods include administration of anti-therapeutic agent antibodies in amounts effective to induce clearance of antibody-agent complexes.
  • The antibodies may be used to remove the modified peptides, analogs or derivatives thereof, and conjugates thereof, from a patient's blood ex vivo by contacting the blood with the antibodies under sterile conditions. For example, the antibodies can be fixed or otherwise immobilized on a column matrix and the patient's blood can be removed from the patient and passed over the matrix. The modified peptide, peptide analogs, derivatives or conjugates will bind to the antibodies and the blood containing a low concentration of peptide, analog, derivative or conjugate, then may be returned to the patient's circulatory system. The amount of peptide compound removed can be controlled by adjusting the pressure and flow rate.
  • Preferential removal of the peptides, analogs, derivatives and conjugates from the plasma component of a patient's blood can be effected, for example, by the use of a semipermeable membrane, or by otherwise first separating the plasma component from the cellular component by ways known in the art prior to passing the plasma component over a matrix containing the anti-therapeutic antibodies. Alternatively the preferential removal of peptide-conjugated blood cells, including red blood cells, can be effected by collecting and concentrating the blood cells in the patient's blood and contacting those cells with fixed anti-therapeutic antibodies to the exclusion of the serum component of the patient's blood.
  • The anti-therapeutic antibodies can be administered in vivo, parenterally, to a patient that has received the peptide, analogs, derivatives or conjugates for treatment. The antibodies will bind peptide compounds and conjugates. Once bound the peptide activity will be hindered if not completely blocked thereby reducing the biologically effective concentration of peptide compound in the patient's bloodstream and minimizing harmful side effects. In addition, the bound antibody—peptide complex will facilitate clearance of the peptide compounds and conjugates from the patient's blood stream.
  • The invention having been fully described can be further appreciated and understood with reference to the following non-limiting examples.
  • EXAMPLE 1 Preparation of a Modified DP178—Synthesis of YTSLIHSLIEESQNQQEKNEQELLELDKWASLWNWFK(MPA)-NH2
  • In this example, DP178 (SEQ ID NO:1) is synthesized and modified to include a linker and maleimide group according to the following synthesis scheme. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, DP178 is a potent inhibitor of HIV-1, and inhibits both cell-induced syncytia formation between HIV-1 infected and uninfected cells and infection of uninfected cells be cell-free HIV-1 virus.
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Phe-OH, Fmoc-Trp(Boc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Trp(Boc)-OH, Fmoc-Leu-OH, Fmoc-Ser(tBu)-OH, Fmoc-Ala-OH, Fmoc-Trp(Boc)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Met-OH, Fmoc-Lys(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Glu(tBu)-OH; Fmoc-Gln(Trt)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Ile-OH, Fmoc-Leu-OH, Fmoc-Ser(tBu)-OH, Fmoc-His(Boc)-OH, Fmoc-Ile-OH, Fmoc-Leu-OH, Fmoc-Ser(tBu)-OH, Fmoc-Thr(tBu)-OH, Fmoc-Tyr(tBu)-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). At the end of the synthesis. The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 run to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00003
  • EXAMPLE 2 Preparation of a Modified DP107—Synthesis of
  • NNLLRAIEAQQHLLQLTVWQIKQLQARILAVERYLKDQK(MPA)NH2
  • In this example, DP107 (SEQ ID NO:2) is synthesized and modified to include a linker and maleimide group according to the following synthesis scheme. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, DP107 exhibits potent antiviral activity against HIV.
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Asp(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Leu-OH, Fmoc-Tyr(tBu)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Val-OH, Fmoc-Ala-OH, Fmoc-Leu-OH, Fmoc-Ile-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Ala-OH, Fmoc-Gln(Trt)-OH, Fmoc-Leu-OH, Fmoc-Gln(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ile-OH, Fmoc-Gln(Trt)-OH, Fmoc-Trp(Boc)-OH, Fmoc-Val-OH, Fmoc-Thr(tBu)-OH, Fmoc-Leu-OH, Fmoc-Gln(Trt)-OH, Fmoc-Leu-OH, Fmoc-Leu-OH, Fmoc-His(Boc)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ala-OH, Fmoc-Glu(tBu)-OH, Fmoc-Ile-OH, Fmoc-Ala-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Leu-OH, Fmoc-Leu-OH, Fmoc-Asn(Trt)-OH, Fmoc-Asn(Trt)-OH,They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). At the end of the synthesis. The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00004
  • EXAMPLE 3
  • Preparation of a Modified anti-RSV Peptide (C Terminal)
  • In this example, the peptide VITIELSNIKENKCNGAKVKLIKQELDKYKNAV (SEQ ID NO:16) is modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, the native sequence (SEQ ID NO.) inhibits viral infection of respiratory syncytial virus (RSV), including inhibiting fusion and syncytia formation between RSV-infected and uninfected Hep-2 cells.
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Val-OH, Fmoc-Ala-OH, Fmoc-Asn(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Tyr(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ile-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Val-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Gly-OH, Fmoc-Asn(Trt)-OH, Fmoc-Cys(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ile-OH, Fmoc-Asn(Trt)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Ile-OH, Fmoc-Thr(tBu)-OH, Fmoc-Ile-OH, Fmoc-Val-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00005
  • EXAMPLE 4 Preparation of a Modified Anti-RSV Peptide (T-N Terminal)
  • In this example, the peptide VITIELSNIKENKCNGAKVKLIKQELDKYKNAV (SEQ ID NO:17), which corresponds to the peptide of SEQ ID NO:16 but where a Cysteine (C) has been substituted for the Methionine (M), residue is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, the native sequence (SEQ ID NO:16) inhibits viral infection of respiratory syncytial virus (RSV), including inhibiting fusion and syncytia formation between RSV-infected and uninfected Hep-2 cells.
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Val-OH, Fmoc-Ala-OH, Fmoc-Asn(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Tyr(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ile-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Val-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Gly-OH, Fmoc-Asn(Trt)-OH, Fmoc-Met-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ile-OH, Fmoc-Asn(Trt)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Ile-OH, Fmoc-Thr(tBu)-OH, Fmoc-Ile-OH, Fmoc-Val-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00006
  • EXAMPLE 5 Preparation of a Modified Anti-RSV Peptide
  • In this example, the peptide SEQ ID NO:14 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:14 inhibits viral infection of respiratory syncytial virus (RSV), including inhibiting fusion and syncytia formation between RSV-infected and uninfected Hep-2 cells.
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Tyr(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ile-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Val-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Gly-OH, Fmoc-Asn(Trt)-OH, Fmoc-Cys(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ile-OH, Fmoc-Asn(Trt)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Ile-OH, Fmoc-Thr(tBu)-OH, Fmoc-Ile-OH, Fmoc-Val-OH, Fmoc-Ser(tBu)-OH, Fmoc-Thr(tBu)-OH, Fmoc-Tyr(tBu)-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at 214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00007
  • EXAMPLE 6 (T-143) Preparation of a Modified Anti-RSV Peptide
  • In this example, the peptide SEQ ID NO:15 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:15 inhibits viral infection of respiratory syncytial virus (RSV), including inhibiting fusion and syncytia formation between RSV-infected and uninfected Hep-2 cells.
  • Solid phase peptide synthesis of the modified peptide analog on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Tyr(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ile-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Val-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Gly-OH, Fmoc-Asn(Trt)-OH, Fmoc-Cys(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ile-OH, Fmoc-Asn(Trt)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Ile-OH, Fmoc-Thr(tBu)-OH, Fmoc-Ile-OH, Fmoc-Val-OH, Fmoc-Ser(tBu)-OH, Fmoc-Thr(tBu)-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00008
  • EXAMPLE 7 Preparation of a Modified Anti-RSV Peptide (C Terminal)
  • In this example, the peptide SEQ ID NO:17), which corresponds to SEQ ID NO:16 with a cysteine (C) substituted for the Methionine (M), is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, the native sequence SEQ ID NO:16. inhibits viral infection of respiratory syncytial virus (RSV), including inhibiting fusion and syncytia formation between RSV-infected and uninfected Hep-2 cells.
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Val-OH, Fmoc-Ala-OH, Fmoc-Asn(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Tyr(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ile-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Val-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Gly-OH, Fmoc-Asn(Trt)-OH, Fmoc-Met-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ile-OH, Fmoc-Asn(Trt)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Ile-OH, Fmoc-Thr(tBu)-OH, Fmoc-Ile-OH, Fmoc-Val-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00009
  • EXAMPLE 8 Preparation of a Modified Anti-RSV Peptide
  • In this example, the peptide SEQ ID NO:29. is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:29 inhibits viral infection of respiratory syncytial virus (RSV), including inhibiting fusion and syncytia formation between RSV-infected and uninfected Hep-2 cells.
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ile-OH, Fmoc-Tyr(tBu)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Leu-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH, Fmoc-Val-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Thr(tBu)-OH, Fmoc-Leu-OH, Fmoc-Val-OH, Fmoc-Ser(tBu)-OH, Fmoc-Val-OH, Fmoc-Gly-OH, Fmoc-Asn(Trt)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Leu-OH, Fmoc-Ser(tBu)-OH, Fmoc-Val-OH, Fmoc-Val-OH, Fmoc-Ala-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Thr(tBu)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Leu-OH, Fmoc-Leu-OH, Fmoc-Ala-OH, Fmoc-Ile-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3. times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian 30 Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide
  • Figure US20080199483A1-20080821-C00010
  • EXAMPLE 9 (T-173) Preparation of a Modified Anti-HPIV Peptide
  • In this example, the peptide SEQ ID NO:52. is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:52 inhibits viral infection of human parainfluenza virus 3 (HPIV3), including inhibiting fusion and syncytia formation between HPIV3-infected Hep2 cells and uninfected CV-1W cells.
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Ile-OH, Fmoc-Leu-OH, Fmoc-Asn(Trt)-OH, Fmoc-Gly-OH, Fmoc-Ile-OH, Fmoc-Ser(tBu)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Val-OH, Fmoc-Ser(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Val-OH, Fmoc-Ala-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Thr(tBu)-OH, Fmoc-Asp(tBu)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Ile-OH, Fmoc-Ala-OH, Fmoc-Glu(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Ile-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Ala-OH, Fmoc-Gln(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Glu(tBu)-OH, Fmoc-Val-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00011
  • EXAMPLE 10 Preparation of a Modified Anti-HPIV Peptide
  • In this example, the peptide SEQ ID NO:58 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:58 inhibits viral infection of human parainfluenza virus 3 (HPIV3), including inhibiting fusion and syncytia formation between HPIV3-infected Hep2 cells and uninfected CV-1W cells.
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Val-OH, Fmoc-Ser(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ile-OH, Fmoc-Ala-OH, Fmoc-Val-OH, Fmoc-Ile-OH, Fmoc-Leu-OH, Fmoc-Asn(Trt)-OH, Fmoc-Gly-OH, Fmoc-Ile-OH, Fmoc-Ser(tBu)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Val-OH, Fmoc-Ser(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Val-OH, Fmoc-Ala-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Thr(tBu)-OH, Fmoc-Asp(tBu)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Ile-OH, Fmoc-Ala-OH, Fmoc-Glu(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Ile-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Arg(Pbf)-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (1 8:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00012
  • EXAMPLE 11 Preparation of a Modified Anti-HPIV Peptide
  • In this example, the peptide SEQ ID NO:35 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:35 inhibits viral infection of human parainfluenza virus 3 (HPIV3), including inhibiting fusion and syncytia formation between HPIV3-infected Hep2 cells and uninfected CV-1W cells.
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Ile-OH, Fmoc-Trp(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Leu-OH, Fmoc-Ile-OH, Fmoc-Glu(tBu)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Ile-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ile-OH, Fmoc-Pro-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH, Fmoc-Gln(Trt)-OH Fmoc-Ala-OH, Fmoc-Val-OH, Fmoc-Ser(tBu)-OH, Fmoc-Asn(Trt)-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00013
  • EXAMPLE 12 Preparation of a Modified Anti-HPIV Peptide
  • In this example, the peptide SEQ ID NO:38 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO: 38 inhibits viral infection of human parainfluenza virus 3 (HPIV3), including inhibiting fusion and syncytia formation between HPIV3-infected Hep2 cells and uninfected CV-1W cells.
  • Solid phase peptide synthesis of the modified peptide analog on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Ile-OH, Fmoc-Ser(tBu)-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Ile-OH, Fmoc-Trp(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Ile-OH, Fmoc-Ser(tBu)-OH, Fmoc-Ile-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ile-OH, Fmoc-Pro-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH, Fmoc-Ala-OH BOC-Lys(Aloc)-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (VN) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00014
  • EXAMPLE 13 Preparation of a Modified Anti-HPIV Peptide
  • In this example, the peptide SEQ ID NO:39 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:39 inhibits viral infection of human parainfluenza virus 3 (HPIV3), including inhibiting fusion and syncytia formation between HPIV3-infected Hep2 cells and uninfected CV-1 W cells.
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Gly-OH, Fmoc-Ile-OH, Fmoc-Ser(tBu)-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Gly-OH, Fmoc-Asn(Trt)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Ile-OH, Fmoc-Trp(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Ile-OH, Fmoc-Ser(tBu)-OH, Fmoc-Ile-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ile-OH, Fmoc-Pro-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00015
  • EXAMPLE 14 Preparation of a Modified Anti-HPIV Peptide
  • In this example, the peptide SEQ ID NO:40 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO. inhibits viral infection of human parainfluenza virus 3 (HPIV3), including inhibiting fusion and syncytia formation between HPIV3-infected Hep2 cells and uninfected CV-1W cells.
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Gly-OH, Fmoc-Ile-OH, Fmoc-Ser(tBu)-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Gly-OH, Fmoc-Asn(Trt)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Ile-OH, Fmoc-Trp(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Ile-OH, Fmoc-Ser(tBu)-OH, Fmoc-Ile-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ile-OH, Fmoc-Pro-OH, Fmoc-Asp(tBu)-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00016
  • EXAMPLE 15 Preparation of a Modified Anti-HPIV Peptide
  • In this example, the peptide SEQ ID NO:41 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:41 inhibits viral infection of human parainfluenza virus 3 (HPIV3), including inhibiting fusion and syncytia formation between HPIV3-infected Hep2 cells and uninfected CV-1W cells.
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Trp(Boc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Gly-OH, Fmoc-Ile-OH, Fmoc-Ser(tBu)-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Gly-OH, Fmoc-Asn(Trt)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Ile-OH, Fmoc-Trp(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Ile-OH, Fmoc-Ser(tBu)-OH, Fmoc-Ile-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ile-OH, Fmoc-Pro-OH Boc-Lys(Aloc)-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00017
  • EXAMPLE 16 Preparation of a Modified Anti-HPIV Peptide
  • In this example, the peptide SEQ ID NO:42 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:42 inhibits viral infection of human parainfluenza virus 3 (HPIV3), including inhibiting fusion and syncytia formation between HPIV3-infected Hep2 cells and uninfected CV-1W cells.
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-His(Boc)-OH, Fmoc-Trp(Boc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Gly-OH, Fmoc-Ile-OH, Fmoc-Ser(tBu)-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Gly-OH, Fmoc-Asn(Trt)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Ile-OH, Fmoc-Trp(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Lys(Boc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Ile-OH, Fmoc-Ser(tBu)-OH, Fmoc-Ile-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ile-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC
  • Figure US20080199483A1-20080821-C00018
  • EXAMPLE 17 Preparation of a Modified Anti-MeV Peptide
  • In this example, the peptide SEQ ID NO:77. is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:77 inhibits viral infection of measles virus (MeV), including inhibiting fusion and syncytia formation between MeV-infected and uninfected Vero cells.
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Ile-OH, Fmoc-Ala-OH, Fmoc-Asn(Trt)-OH, Fmoc-Gly-OH, Fmoc-Leu-OH, Fmoc-Asn(Trt)-OH, Fmoc-Thr(tBu)-OH, Fmoc-Gly-OH, Fmoc-Val-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Ser(tBu)-OH, Fmoc-Ile-OH, Fmoc-Pro-OH, Fmoc-Pro-OH, Fmoc-Gly-OH, Fmoc-Leu-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ile-OH, Fmoc-Arg(Pbf)-OH, Fmoc-His(Boc)OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide. (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00019
  • EXAMPLE 18 Preparation of a Modified Anti-MeV Peptide
  • In this example, the peptide SEQ ID NO:79 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:79 inhibits viral infection of measles virus (MeV), including inhibiting fusion and syncytia formation between MeV-infected and uninfected Vero cells.
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Ile-OH, Fmoc-Ala-OH, Fmoc-Asn(Trt)-OH, Fmoc-Gly-OH, Fmoc-Leu-OH, Fmoc-Asn(Trt)-OH, Fmoc-Thr(tBu)-OH, Fmoc-Gly-OH, Fmoc-Val-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Ser(tBu)-OH, Fmoc-Ile-OH, Fmoc-Pro-OH, Fmoc-Pro-OH, Fmoc-Gly-OH, Fmoc-Leu-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ile-OH, They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC
  • Figure US20080199483A1-20080821-C00020
  • EXAMPLE 19 Preparation of a Modified Anti-MeV Peptide
  • In this example, the peptide SEQ ID NO:81 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO: 79 inhibits viral infection of measles virus (MeV), including inhibiting fusion and syncytia formation between MeV-infected and uninfected Vero cells.
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Ile-OH, Fmoc-Ala-OH, Fmoc-Asn(Trt)-OH, Fmoc-Gly-OH, Fmoc-Leu-OH, Fmoc-Asn(Trt)-OH, Fmoc-Thr(tBu)-OH, Fmoc-Gly-OH, Fmoc-Val-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Ser(tBu)-OH, Fmoc-Ile-OH, Fmoc-Pro-OH, Fmoc-Pro-OH, Fmoc-Gly-OH, Fmoc-Leu-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (VN) piperidine in N,N-dimethylformrnamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00021
  • EXAMPLE 20 Preparation of a Modified Anti-MeV Peptide
  • In this example, the peptide SEQ ID NO:84 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:84 inhibits viral infection of measles virus (MeV), including inhibiting fusion and syncytia formation between MeV-infected and uninfected Vero cells.
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Leu-OH, Fmoc-Ile-OH, Fmoc-Gln(Trt)-OH, Fmoc-Asp(tBu)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Ala-OH, Fmoc-Ile-OH, Fmoc-Ala-OH, Fmoc-Asn(Trt)-OH, Fmoc-Gly-OH, Fmoc-Leu-OH, Fmoc-Asn(Trt)-OH, Fmoc-Thr(tBu)-OH, Fmoc-Gly-OH, Fmoc-Val-OH, Fmoc-Asp(tBu)-OH, Fmoc-Leu-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Ser(tBu)-OH, Fmoc-Ile-OH, Fmoc-Pro-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00022
  • EXAMPLE 21 Preparation of a Modified Anti-SIV Peptide
  • In this example, the peptide SEQ ID NO:64 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:64. exhibits potent antiviral activity as a crude peptide against simian immunodeficiency virus (SIV).
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Tyr(tBu)-OH, Fmoc-Met-OH, Fmoc-Asn(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ile-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ala-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Leu-OH, Fmoc-Ala-OH, Fmoc-Thr(tBu)-OH, Fmoc-Ile-OH, Fmoc-Asn(Trt)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Phe-OH, Fmoc-Asp(tBu)-OH, Fmoc-Val-OH, Fmoc-Lys(Boc)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Trp(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Trp(Boc)-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00023
  • EXAMPLE 22 Preparation of a Modified Anti-SIV Peptide
  • In this example, the peptide SEQ ID NO:65 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:65 exhibits potent antiviral activity as a crude peptide against simian immunodeficiency virus (SIV).
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Tyr(tBu)-OH, Fmoc-Met-OH, Fmoc-Asn(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ile-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ala-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Leu-OH, Fmoc-Ala-OH, Fmoc-Thr(tBu)-OH, Fmoc-Ile-OH, Fmoc-Asn(Trt)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Phe-OH, Fmoc-Asp(tBu)-OH, Fmoc-Val-OH, Fmoc-Lys(Boc)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Trp(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (VN) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00024
  • EXAMPLE 23 Preparation of a Modified Anti-SIV Peptide
  • In this example, the peptide SEQ ID NO:66 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:66 exhibits potent antiviral activity as a crude peptide against simian immunodeficiency virus (SIV).
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Asn(Trt)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Tyr(tBu)-OH, Fmoc-Met-OH, Fmoc-Asn(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ile-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ala-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Leu-OH, Fmoc-Ala-OH, Fmoc-Thr(tBu)-OH, Fmoc-Ile-OH, Fmoc-Asn(Trt)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Phe-OH, Fmoc-Asp(tBu)-OH, Fmoc-Val-OH, Fmoc-Lys(Boc)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Trp(Boc)-OH, Fmoc-Glu(tBu)-OH Boc-Lys(Aloc)-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00025
  • EXAMPLE 24 Preparation of a Modified Anti-SIV Peptide
  • In this example, the peptide SEQ ID NO:67 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:67 exhibits potent antiviral activity as a crude peptide against simian immunodeficiency virus (SIV).
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Tyr(tBu)-OH, Fmoc-Met-OH, Fmoc-Asn(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ile-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ala-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Leu-OH, Fmoc-Ala-OH, Fmoc-Thr(tBu)-OH, Fmoc-Ile-OH, Fmoc-Asn(Trt)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Phe-OH, Fmoc-Asp(tBu)-OH, Fmoc-Val-OH, Fmoc-Lys(Boc)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Trp(Boc)-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylfQrmarnide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00026
  • EXAMPLE 25 Preparation of a Modified Anti-SIV Peptide
  • In this example, the peptide SEQ ID NO:68 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:68 exhibits potent antiviral activity as a crude peptide against simian immunodeficiency virus (SIV).
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Trp(Boc)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Tyr(tBu)-OH, Fmoc-Met-OH, Fmoc-Asn(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ile-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ala-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Leu-OH, Fmoc-Ala-OH, Fmoc-Thr(tBu)-OH, Fmoc-Ile-OH, Fmoc-Asn(Trt)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Phe-OH, Fmoc-Asp(tBu)-OH, Fmoc-Val-OH, Fmoc-Lys(Boc)-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Glu(tBu)-OH Boc-Lys(Aloc)-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00027
  • EXAMPLE 26 Preparation of a Modified Anti-SIV Peptide
  • In this example, the peptide SEQ ID NO:69 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:69 exhibits potent antiviral activity as a crude peptide against simian immunodeficiency virus (SIV).
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Asp(tBu)-OH, Fmoc-Trp(Boc)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Tyr(tBu)-OH, Fmoc-Met-OH, Fmoc-Asn(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ile-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ala-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Leu-OH, Fmoc-Ala-OH, Fmoc-Thr(tBu)-OH, Fmoc-Ile-OH, Fmoc-Asn(Trt)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Phe-OH, Fmoc-Asp(tBu)-OH, Fmoc-Val-OH, Fmoc-Lys(Boc)-OH, Fmoc-Arg(Pbf)-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00028
  • EXAMPLE 27 Preparation of a Modified Anti-SIV Peptide
  • In this example, the peptide SEQ ID NO:70. is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:70 exhibits potent antiviral activity as a crude peptide against simian immunodeficiency virus (SIV).
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Val-OH, Fmoc-Asp(tBu)-OH, Fmoc-Trp(Boc)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Tyr(tBu)-OH, Fmoc-Met-OH, Fmoc-Asn(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ile-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ala-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Leu-OH, Fmoc-Ala-OH, Fmoc-Thr(tBu)-OH, Fmoc-Ile-OH, Fmoc-Asn(Trt)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Phe-OH, Fmoc-Asp(tBu)-OH, Fmoc-Val-OH, Fmoc-Lys(Boc)-OH, Boc-Lys(Aloc)-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00029
  • EXAMPLE 28 Preparation of a Modified Anti-SIV Peptide
  • In this example, the peptide SEQ ID NO:71 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:71 exhibits potent antiviral activity as a crude peptide against simian immunodeficiency virus (SIV).
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Phe-OH, Fmoc-Val-OH, Fmoc-Asp(tBu)-OH, Fmoc-Trp(Boc)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Tyr(tBu)-OH, Fmoc-Met-OH, Fmoc-Asn(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ile-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ala-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)OH, Fmoc-Leu-OH, Fmoc-Leu-OH, Fmoc-Ala-OH, Fmoc-Thr(tBu)-OH, Fmoc-Ile-OH, Fmoc-Asn(Trt)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Phe-OH, Fmoc-Asp(tBu)-OH, Fmoc-Val-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00030
  • EXAMPLE 29 Preparation of a Modified Anti-SIV Peptide
  • In this example, the peptide SEQ ID NO:72 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:72 exhibits potent antiviral activity as a crude peptide against simian immunodeficiency virus (SIV).
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Gly-OH, Fmoc-Phe-OH, Fmoc-Val-OH, Fmoc-Asp(tBu)-OH, Fmoc-Trp(Boc)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Tyr(tBu)-OH, Fmoc-Met-OH, Fmoc-Asn(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ile-OH, Fmoc-Gln(Trt)OH, Fmoc-Ala-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Leu-OH, Fmoc-Ala-OH, Fmoc-Thr(tBu)-OH, Fmoc-Ile-OH, Fmoc-Asn(Trt)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Phe-OH, Fmoc-Asp(tBu)-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00031
  • EXAMPLE 30 Preparation of a Modified Anti-SIV Peptide
  • In this example, the peptide SEQ ID NO:73 is synthesized and modified to include a linker and maleimide group according to the synthesis scheme set forth below. As reported in U.S. Pat. Nos. 6,013,236 and 6,020,459, SEQ ID NO:73 exhibits potent antiviral activity as a crude peptide against simian immunodeficiency virus (SIV).
  • Solid phase peptide synthesis of the modified peptide on a 100 μmole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA. The following protected amino acids are sequentially added to resin: Fmoc-Lys(Aloc)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Gly-OH, Fmoc-Phe-OH, Fmoc-Val-OH, Fmoc-Asp(tBu)-OH, Fmoc-Trp(Boc)-OH, Fmoc-Ser(tBu)-OH, Fmoc-Asn(Trt)-OH, Fmoc-Leu-OH, Fmoc-Lys(Boc)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Leu-OH, Fmoc-Glu(tBu)-OH, Fmoc-Tyr(tBu)-OH, Fmoc-Met-OH, Fmoc-Asn(Trt)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ile-OH, Fmoc-Gln(Trt)-OH, Fmoc-Ala-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Leu-OH, Fmoc-Ala-OH, Fmoc-Thr(tBu)-OH, Fmoc-Ile-OH, Fmoc-Asn(Trt)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Glu(tBu)-OH, Fmoc-Leu-OH, Fmoc-Phe-OH. They are dissolved in N,N-dimethylformamide (DMF) and, according to the sequence, activated using O-benzotriazol-1-yl-N,N,N′,N′-tetramethyl-uronium hexafluorophosphate (HBTU) and Diisopropylethylamine (DIEA). Removal of the Fmoc protecting group is achieved using a solution of 20% (V/V) piperidine in N,N-dimethylformamide (DMF) for 20 minutes (step 1). The selective deprotection of the Lys (Aloc) group is performed manually and accomplished by treating the resin with a solution of 3 eq of Pd(PPh3)4 dissolved in 5 mL of CHCl3:NMM:HOAc (18:1:0.5) for 2 h (Step 2). The resin is then washed with CHCl3 (6×5 mL), 20% HOAc in DCM (6×5 mL), DCM (6×5 mL), and DMF (6×5 mL). The synthesis is then re-automated for the addition of the 3-maleimidopropionic acid (Step 3). Between every coupling, the resin is washed 3 times with N,N-dimethylformamide (DMF) and 3 times with isopropanol. The peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et2O (Step 4). The product is purified by preparative reversed phased HPLC using a Varian (Rainin) preparative binary HPLC system: gradient elution of 30-55% B (0.045% TFA in H2O (A) and 0.045% TFA in CH3CN (B)) over 180 min at 9.5 mL/min using a Phenomenex Luna 10μ phenyl-hexyl, 21 mm×25 cm column and UV detector (Varian Dynamax UVD II) at λ214 and 254 nm to afford the desired modified peptide (i.e., DAC) in >95% purity, as determined by RP-HPLC.
  • Figure US20080199483A1-20080821-C00032
  • While certain embodiments of the invention have been described and exemplified, those having ordinary skill in the art will understand that the invention is not intended to be limited to the specifics of any of these embodiments, but is rather defined by the accompanying claims.
  • TABLE 2
    DP178 CARBOXY TRUNCATIONS
    YTS
    YTSL
    YTSLI
    YTSLIH
    YTSLIHS
    YTSLIHSL
    YTSLIHSLI
    YTSLIHSLIE
    YTSLIHSLIEE
    YTSLIHSLIEES
    YTSLIHSLIEESQ
    YTSLIHSLIEESQN
    YTSLIHSLIEESQNQ
    YTSLIHSLIEESQNQQ
    YTSLIHSLIEESQNQQE
    YTSLIHSLIEESQNQQEK
    YTSLIHSLIEESQNQQEKN
    YTSLIHSLIEESQNQQEKNE
    YTSLIHSLIEESQNQQEKNEQ
    YTSLIHSLIEESQNQQEKNEQE
    YTSLIHSLIEESQNQQEKNEQEL
    YTSLIHSLIEESQNQQEKNEQELL
    YTSLIHSLIEESQNQQEKNEQELLE
    YTSLIHSLIEESQNQQEKNEQELLEL
    YTSLIHSLIEESQNQQEKNEQELLELD
    YTSLIHSLIEESQNQQEKNEQELLELDK
    YTSLIHSLIEESQNQQEKNEQELLELDKW
    YTSLIHSLIEESQNQQEKNEQELLELDKWA
    YTSLIHSLIEESQNQQEKNEQELLELDKWAS
    YTSLIHSLIEESQNQQEKNEQELLELDKWASL
    YTSLIHSLIEESQNQQEKNEQELLELDKWASLW
    YTSLIHSLIEESQNQQEKNEQELLELDKWASLWN
    YTSLIHSLIEESQNQQEKNEQELLELDKWASLWNW
    YTSLIHSLIEESQNQQEKNEQELLELDKWASLWNWF
  • The one letter amino acid code of Table 1 is used.
  • TABLE 3
    DP178 AMINO TRUNCATIONS
    NWF
    WNWF
    LWNWF
    SLWNWF
    ASLWNWF
    WASLWNWF
    KWASLWNWF
    DKWASLWNWF
    LDKWASLWNWF
    ELDKWASLWNWF
    LELDKWASLWNWF
    LLELDKWASLWNWF
    ELLELDKWASLWNWF
    QELLELDKWASLWNWF
    EQELLELDKWASLWNWF
    NEQELLELDKWASLWNWF
    KNEQELLELDKWASLWNWF
    EKNEQELLELDKWASLWNWF
    QEKNEQELLELDKWASLWNWF
    QQEKNEQELLELDKWASLWNWF
    NQQEKNEQELLELDKWASLWNWF
    QNQQEKNEQELLELDKWASLWNWF
    SQNQQEKNEQELLELDKWASLWNWF
    ESQNQQEKNEQELLELDKWASLWNWF
    EESQNQQEKNEQELLELDKWASLWNWF
    IEESQNQQEKNEQELLELDKWASLWNWF
    LIEESQNQQEKNEQELLELDKWASLWNWF
    SLIEESQNQQEKNEQELLELDKWASLWNWF
    HSLIEESQNQQEKNEQELLELDKWASLWNWF
    IHSLIEESQNQQEKNEQELLELDKWASLWNWF
    LIHSLIEESQNQQEKNEQELLELDKWASLWNWF
    SLIHSLIEESQNQQEKNEQELLELDKWASLWNWF
    TSLIHSLIEESQNQQEKNEQELLELDKWASLWNWF
    YTSLIHSLIEESQNQQEKNEQELLELDKWASLWNWF
  • The one letter amino acid code of Table 1 is used.
  • TABLE 4
    DP107 CARBOXY TRUNCATIONS
    NNL
    NNLL
    NNLLR
    NNLLRA
    NNLLRAI
    NNLLRAIE
    NNLLRAIEA
    NNLLRAIEAQ
    NNLLRAIEAQQ
    NNLLRAIEAQQH
    NNLLRAIEAQQHL
    NNLLRAIEAQQHLL
    NNLLRAIEAQQHLLQ
    NNLLRAIEAQQHLLQL
    NNLLRAIEAQQHLLQLT
    NNLLRAIEAQQHLLQLTV
    NNLLRAIEAQQHLLQLTVW
    NNLLRAIEAQQHLLQLTVWQ
    NNLLRAIEAQQHLLQLTVWQI
    NNLLRAIEAQQHLLQLTVWQIK
    NNLLRAIEAQQHLLQLTVWQIKQ
    NNLLRAIEAQQHLLQLTVWQIKQL
    NNLLRAIEAQQHLLQLTVWQIKQLQ
    NNLLRAIEAQQHLLQLTVWQIKQLQA
    NNLLRAIEAQQHLLQLTVWQIKQLQAR
    NNLLRAIEAQQHLLQLTVWQIKQLQARI
    NNLLRAIEAQQHLLQLTVWQIKQLQARIL
    NNLLRAIEAQQHLLQLTVWQIKQLQARILA
    NNLLRAIEAQQHLLQLTVWQIKQLQARILAV
    NNLLRAIEAQQHLLQLTVWQIKQLQARILAVE
    NNLLRAIEAQQHLLQLTVWQIKQLQARILAVER
    NNLLRAIEAQQHLLQLTVWQIKQLQARILAVERY
    NNLLRAIEAQQHLLQLTVWQIKQLQARILAVERYL
    NNLLRAIEAQQHLLQLTVWQIKQLQARILAVERYLK
    NNLLRAIEAQQHLLQLTVWQIKQLQARILAVERYLKD
    NNLLRAIEAQQHLLQLTVWQIKQLQARILAVERYLKDQ
  • The one letter amino acid code of Table 1 is used.
  • TABLE 5
    DP107 AMINO TRUNCATIONS
    KDQ
    LKDQ
    YLKDQ
    RYLKDQ
    ERYLKDQ
    VERYLKDQ
    AVERYLKDQ
    LAVERYLKDQ
    ILAVERYLKDQ
    RILAVERYLKDQ
    ARILAVERYLKDQ
    QARILAVERYLKDQ
    LQARILAVERYLKDQ
    QLQARILAVERYLKDQ
    KQLQARILAVERYLKDQ
    IKQLQARILAVERYLKDQ
    QIKQLQARILAVERYLKDQ
    WQIKQLQARILAVERYLKDQ
    VWQIKQLQARILAVERYLKDQ
    TVWQIKQLQARILAVERYLKDQ
    LTVWQIKQLQARILAVERYLKDQ
    QLTVWQIKQLQARILAVERYLKDQ
    LQLTVWQIKQLQARILAVERYLKDQ
    LLQLTVWQIKQLQARILAVERYLKDQ
    HLLQLTVWQIKQLQARILAVERYLKDQ
    QHLLQLTVWQIKQLQARILAVERYLKDQ
    QQHLLQLTVWQIKQLQARILAVERYLKDQ
    AQQHLLQLTVWQIKQLQARILAVERYLKDQ
    EAQQHLLQLTVWQIKQLQARILAVERYLKDQ
    IEAQQHLLQLTVWQIKQLQARILAVERYLKDQ
    AIEAQQHLLQLTVWQIKQLQARILAVERYLKDQ
    RAIEAQQHLLQLTVWQIKQLQARILAVERYLKDQ
    LRAIEAQQHLLQLTVWQIKQLQARILAVERYLKDQ
    LLRAIEAQQHLLQLTVWQIKQLQARILAVERYLKDQ
    NLLRAIEAQQHLLQLTVWQIKQLQARILAVERYLKDQ
    NNLLRAIEAQQHLLQLTVWQIKQLQARILAVERYLKDQ
  • The one letter amino acid code of Table 1 is used.
  • TABLE 6
    HIV-2NIHZDP178 analog carboxy truncations.
    LEA
    LEAN
    LEANI
    LEANIS
    LEANISQ
    LEANISQS
    LEANISQSL
    LEANISQSLE
    LEANISQSLEQ
    LEANISQSLEQA
    LEANISQSLEQAQ
    LEANISQSLEQAQI
    LEANISQSLEQAQIQ
    LEANISQSLEQAQIQQ
    LEANISQSLEQAQIQQE
    LEANISQSLEQAQIQQEK
    LEANISQSLEQAQIQQEKN
    LEANISQSLEQAQIQQEKNM
    LEANISQSLEQAQIQQEKNMY
    LEANISQSLEQAQIQQEKNMYE
    LEANISQSLEQAQIQQEKNMYEL
    LEANISQSLEQAQIQQEKNMYELQ
    LEANISQSLEQAQIQQEKNMYELQK
    LEANISQSLEQAQIQQEKNMYELQKL
    LEANISQSLEQAQIQQEKNMYELQKLN
    LEANISQSLEQAQIQQEKNMYELQKLNS
    LEANISQSLEQAQIQQEKNMYELQKLNSW
    LEANISQSLEQAQIQQEKNMYELQKLNSWD
    LEANISQSLEQAQIQQEKNMYELQKLNSWDV
    LEANISQSLEQAQIQQEKNMYELQKLNSWDVF
    LEANISQSLEQAQIQQEKNMYELQKLNSWDVFT
    LEANISQSLEQAQIQQEKNMYELQKLNSWDVFTN
    LEANISQSLEQAQIQQEKNMYELQKLNSWDVFTNW
    LEANISQSLEQAQIQQEKNMYELQKLNSWDVFTNWL
  • The one letter amino acid code of Table 1 is used.
  • TABLE 7
    HIV-2NIHZ DP178 analog amino truncations.
    NWL
    TNWL
    FTNWL
    VFTNWL
    DVFTNWL
    WDVFTNWL
    SWDVFTNWL
    NSWDVFTNWL
    LNSWDVFTNWL
    KLNSWDVFTNWL
    QKLNSWDVFTNWL
    LQKLNSWDVFTNWL
    ELQKLNSWDVFTNWL
    YELQKLNSWDVFTNWL
    MYELQKLNSWDVFTNWL
    NMYELQKLNSWDVFTNWL
    KNMYELQKLNSWDVFTNWL
    EKNMYELQKLNSWDVFTNWL
    QEKNMYELQKLNSWDVFTNWL
    QQEKNMYELQKLNSWDVFTNWL
    IQQEKNMYELQKLNSWDVFTNWL
    QIQQEKNMYELQKLNSWDVFTNWL
    AQIQQEKNMYELQKLNSWDVFTNWL
    QAQIQQEKNMYELQKLNSWDVFTNWL
    EQAQIQQEKNMYELQKLNSWDVFTNWL
    LEQAQIQQEKNMYELQKLNSWDVFTNWL
    SLEQAQIQQEKNMYELQKLNSWDVFTNWL
    QSLEQAQIQQEKNMYELQKLNSWDVFTNWL
    SQSLEQAQIQQEKNMYELQKLNSWDVFTNWL
    ISQSLEQAQIQQEKNMYELQKLNSWDVFTNWL
    NISQSLEQAQIQQEKNMYELQKLNSWDVFTNWL
    ANISQSLEQAQIQQEKNMYELQKLNSWDVFTNWL
    EANISQSLEQAQIQQEKNMYELQKLNSWDVFTNWL
    LEANISQSLEQAQIQQEKNMYELQKLNSWDVFTNWL
  • The one letter amino acid code of Table 1 is used.
  • TABLE 8
    RESPIRATORY SYNCYTIAL VIRUS (RSV) DP107 F2
    REGION ANALOG CARBOXY TRUNCATIONS
    YTS
    YTSV
    YTSVI
    YTSVIT
    YTSVITI
    YTSVITIE
    YTSVITIEL
    YTSVITIELS
    YTSVITIELSN
    YTSVITIELSNI
    YTSVITIELSNIK
    YTSVITIELSNIKE
    YTSVITIELSNIKEN
    YTSVITIELSNIKENK
    YTSVITIELSNIKENKC
    YTSVITIELSNIKENKCN
    YTSVITIELSNIKENKCNG
    YTSVITIELSNIKENKCNGT
    YTSVITIELSNIKENKCNGTD
    YTSVITIELSNIKENKCNGTDA
    YTSVITIELSNIKENKCNGTDAK
    YTSVITIELSNIKENKCNGTDAKV
    YTSVITIELSNIKENKCNGTDAKVK
    YTSVITIELSNIKENKCNGTDAKVKL
    YTSVITIELSNIKENKCNGTDAKVKLI
    YTSVITIELSNIKENKCNGTDAKVKLIK
    YTSVITIELSNIKENKCNGTDAKVKLIKQ
    YTSVITIELSNIKENKCNGTDAKVKLIKQE
    YTSVITIELSNIKENKCNGTDAKVKLIKQEL
    YTSVITIELSNIKENKCNGTDAKVKLIKQELD
    YTSVITIELSNIKENKCNGTDAKVKLIKQELDK
    YTSVITIELSNIKENKCNGTDAKVKLIKQELDKY
    YTSVITIELSNIKENKCNGTDAKVKLIKQELDKYK
    YTSVITIELSNIKENKCNGTDAKVKLIKQELDKYKN
    YTSVITIELSNIKENKCNGTDAKVKLIKQELDKYKNA
    YTSVITIELSNIKENKCNGTDAKVKLIKQELDKYKNAV
    YTSVITIELSNIKENKCNGTDAKVKLIKQELDKYKNAVT
    YTSVITIELSNIKENKCNGTDAKVKLIKQELDKYKNAVTE
    YTSVITIELSNIKENKCNGTDAKVKLIKQELDKYKNAVTEL
    YTSVITIELSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQ
    YTSVITIELSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQL
    YTSVITIELSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQLL
    YTSVITIELSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQLLM
    YTSVITIELSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQLLMQ
    YTSVITIELSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQLLMQS
    YTSVITIELSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQLLMQST
  • The one letter amino acid code of Table 1 is used.
  • TABLE 9
    RESPIRATORY SYNCYTIAL VIRUS (RSV) DP107 F2
    REGION ANALOG AMINO TRUNCATIONS
    QST
    MQST
    LMQST
    LLMQST
    QLLMQST
    LQLLMQST
    ELQLLMQST
    TELQLLMQST
    VTELQLLMQST
    AVTELQLLMQST
    NAVTELQLLMQST
    KNAVTELQLLMQST
    YKNAVTELQLLMQST
    KYKNAVTELQLLMQST
    DKYKNAVTELQLLMQST
    LDKYKNAVTELQLLMQST
    ELDKYKNAVTELQLLMQST
    QELDKYKNAVTELQLLMQST
    KQELDKYKNAVTELQLLMQST
    IKQELDKYKNAVTELQLLMQST
    LIKQELDKYKNAVTELQLLMQST
    KLIKQELDKYKNAVTELQLLMQST
    VKLIKQELDKYKNAVTELQLLMQST
    KVKLIKQELDKYKNAVTELQLLMQST
    AKVKLIKQELDKYKNAVTELQLLMQST
    DAKVKLIKQELDKYKNAVTELQLLMQST
    TDAKVKLIKQELDKYKNAVTELQLLMQST
    GTDAKVKLIKQELDKYKNAVTELQLLMQST
    NGTDAKVKLIKQELDKYKNAVTELQLLMQST
    CNGTDAKVKLIKQELDKYKNAVTELQLLMQST
    KCNGTDAKVKLIKQELDKYKNAVTELQLLMQST
    NKCNGTDAKVKLIKQELDKYKNAVTELQLLMQST
    KENKCNGTDAKVKLIKQELDKYKNAVTELQLLMQST
    IKENKCNGTDAKVKLIKQELDKYKNAVTELQLLMQST
    NIKENKCNGTDAKVKLIKQELDKYKNAVTELQLLMQST
    SNIKENKCNGTDAKVKLIKQELDKYKNAVTELQLLMQST
    LSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQLLMQST
    ELSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQLLMQST
    IELSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQLLMQST
    TIELSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQLLMQST
    ITIELSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQLLMQST
    VITIELSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQLLMQST
    SVITIELSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQLLMQST
    TSVITIELSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQLLMQST
  • The one letter amino acid code of Table 1 is used.
  • TABLE 10
    RESPIRATORY SYNCYTIAL VIRUS (RSV) F1 DP178
    REGION ANALOG CARBOXY TRUNCATIONS
    FYD
    FYDP
    FYDPL
    FYDPLV
    FYDPLVF
    FYDPLVFP
    FYDPLVFPS
    FYDPLVFPSD
    FYDPLVFPSDE
    FYDPLVFPSDEF
    FYDPLVFPSDEFD
    FYDPLVFPSDEFDA
    FYDPLVFPSDEFDAS
    FYDPLVFPSDEFDASI
    FYDPLVFPSDEFDASIS
    FYDPLVFPSDEFDASISQ
    FYDPLVFPSDEFDASISQV
    FYDPLVFPSDEFDASISQVN
    FYDPLVFPSDEFDASISQVNE
    FYDPLVFPSDEFDASISQVNEK
    FYDPLVFPSDEFDASISQVNEKI
    FYDPLVFPSDEFDASISQVNEKIN
    FYDPLVFPSDEFDASISQVNEKINQ
    FYDPLVFPSDEFDASISQVNEKINQS
    FYDPLVFPSDEFDASISQVNEKINQSL
    FYDPLVFPSDEFDASISQVNEKINQSLA
    FYDPLVFPSDEFDASISQVNEKINQSLAF
    FYDPLVFPSDEFDASISQVNEKINQSLAFI
    FYDPLVFPSDEFDASISQVNEKINQSLAFIR
    FYDPLVFPSDEFDASISQVNEKINQSLAFIRK
    FYDPLVFPSDEFDASISQVNEKINQSLAFIRKS
    FYDPLVFPSDEFDASISQVNEKINQSLAFIRKSD
    FYDPLVFPSDEFDASISQVNEKINQSLAFIRKSDE
    FYDPLVFPSDEFDASISQVNEKINQSLAFIRKSDEL
    FYDPLVFPSDEFDASISQVNEKINQSLAFIRKSDELL
  • The one letter amino acid code of Table 1 is used.
  • TABLE 11
    RESPIRATORY SYNCYTIAL VIRUS (RSV) F1 DP178
    REGION ANALOG AMINO TRUNCATIONS
    DELL
    SDELL
    KSDELL
    RKSDELL
    IRKSDELL
    FIRKSDELL
    AFIRKSDELL
    LAFIRKSDELL
    SLAFIRKSDELL
    QSLAFIRKSDELL
    NQSLAFIRKSDELL
    INQSLAFIRKSDELL
    KINQSLAFIRKSDELL
    EKINQSLAFIRKSDELL
    NEKINQSLAFIRKSDELL
    VNEKINQSLAFIRKSDELL
    QVNEKINQSLAFIRKSDELL
    SQVNEKINQSLAFIRKSDELL
    ISQVNEKINQSLAFIRKSDELL
    SISQVNEKINQSLAFIRKSDELL
    ASISQVNEKINQSLAFIRKSDELL
    DASISQVNEKINQSLAFIRKSDELL
    FDASISQVNEKINQSLAFIRKSDELL
    EFDASISQVNEKINQSLAFIRKSDELL
    DEFDASISQVNEKINQSLAFIRKSDELL
    SDEFDASISQVNEKINQSLAFIRKSDELL
    PSDEFDASISQVNEKINQSLAFIRKSDELL
    FPSDEFDASISQVNEKINQSLAFIRKSDELL
    VFPSDEFDASISQVNEKINQSLAFIRKSDELL
    LVFPSDEFDASISQVNEKINQSLAFIRKSDELL
    PLVFPSDEFDASISQVNEKINQSLAFIRKSDELL
    DPLVFPSDEFDASISQVNEKINQSLAFIRKSDELL
    YDPLVFPSDEFDASISQVNEKINQSLAFIRKSDELL
  • The one letter amino acid code of Table 1 is used.
  • TABLE 12
    HUMAN PARAINFLUENZA VIRUS 3 (HPV3) F1 REGION DP178
    ANALOG CARBOXY TRUNCATIONS
    ITL
    ITLN
    ITLNN
    ITLNNS
    ITLNNSV
    ITLNNSVA
    ITLNNSVAL
    ITLNNSVALD
    ITLNNSVALDP
    ITLNNSVALDPI
    ITLNNSVALDPID
    ITLNNSVALDPIDI
    ITLNNSVALDPIDIS
    ITLNNSVALDPIDISI
    ITLNNSVALDPIDISIE
    ITLNNSVALDPIDISIEL
    ITLNNSVALDPIDISIELN
    ITLNNSVALDPIDISIELNK
    ITLNNSVALDPIDISIELNKA
    ITLNNSVALDPIDISIELNKAK
    ITLNNSVALDPIDISIELNKAKS
    ITLNNSVALDPIDISIELNKAKSD
    ITLNNSVALDPIDISIELNKAKSDL
    ITLNNSVALDPIDISIELNKAKSDLE
    ITLNNSVALDPIDISIELNKAKSDLEE
    ITLNNSVALDPIDISIELNKAKSDLEES
    ITLNNSVALDPIDISIELNKAKSDLEESK
    ITLNNSVALDPIDISIELNKAKSDLEESKE
    ITLNNSVALDPIDISIELNKAKSDLEESKEW
    ITLNNSVALDPIDISIELNKAKSDLEESKEWI
    ITLNNSVALDPIDISIELNKAKSDLEESKEWIR
    ITLNNSVALDPIDISIELNKAKSDLEESKEWIRR
    ITLNNSVALDPIDISIELNKAKSDLEESKEWIRRS
  • The one letter amino acid code of Table 1 is used.
  • TABLE 13
    HUMAN PARAINFLUENZA VIRUS 3 (HPV3) F1 REGION DP178
    ANALOG AMINO TRUNCATIONS
    RRS
    IRRS
    WIRRS
    EWIRRS
    KEWIRRS
    SKEWIRRS
    ESKEWIRRS
    EESKEWIRRS
    LEESKEWIRRS
    DLEESKEWIRRS
    SDLEESKEWIRRS
    KSDLEESKEWIRRS
    AKSDLEESKEWIRRS
    KAKSDLEESKEWIRRS
    NKAKSDLEESKEWIRRS
    LNKAKSDLEESKEWIRRS
    ELNKAKSDLEESKEWIRRS
    IELNKAKSDLEESKEWIRRS
    SIELNKAKSDLEESKEWIRRS
    ISIELNKAKSDLEESKEWIRRS
    DISIELNKAKSDLEESKEWIRRS
    IDISIELNKAKSDLEESKEWIRRS
    PIDISIELNKAKSDLEESKEWIRRS
    DPIDISIELNKAKSDLEESKEWIRRS
    LDPIDISIELNKAKSDLEESKEWIRRS
    ALDPIDISLELNKAKSDLEESKEWIRRS
    VALDPIDISIELNKAKSDLEESKEWIRRS
    SVALDPIDISIELNKAKSDLEESKEWIRRS
    NSVALDPIDISIELNKAKSDLEESKEWIRRS
    NNSVALDPIDISIELNKAKSDLEESKEWIRRS
    LNNSVALDPIDISIELNKAKSDLEESKEWIRRS
    TLNNSVALDPIDISIELNKAKSDLEESKEWIRRS
  • The one letter amino acid code of Table 1 is used.
  • TABLE 14
    HUMAN PARAINFLUENZA VIRUS 3 (HPV3) F1 REGION
    DP107 ANALOG CARBOXY TRUNCATIONS
    ALG
    ALGV
    ALGVA
    ALGVAT
    ALGVATS
    ALGVATSA
    ALGVATSAQ
    ALGVATSAQI
    ALGVATSAQIT
    ALGVATSAQITA
    ALGVATSAQITAA
    ALGVATSAQITAAV
    ALGVATSAQITAAVA
    ALGVATSAQITAAVAL
    ALGVATSAQITAAVALV
    ALGVATSAQITAAVALVE
    ALGVATSAQITAAVALVEA
    ALGVATSAQITAAVALVEAK
    ALGVATSAQITAAVALVEAKQ
    ALGVATSAQITAAVALVEAKQA
    ALGVATSAQITAAVALVEAKQAR
    ALGVATSAQITAAVALVEAKQARS
    ALGVATSAQITAAVALVEAKQARSD
    ALGVATSAQITAAVALVEAKQARSDI
    ALGVATSAQITAAVALVEAKQARSDIE
    ALGVATSAQITAAVALVEAKQARSDIEK
    ALGVATSAQITAAVALVEAKQARSDIEKL
    ALGVATSAQITAAVALVEAKQARSDIEKLK
    ALGVATSAQITAAVALVEAKQARSDIEKLKE
    ALGVATSAQITAAVALVEAKQARSDIEKLKEA
    ALGVATSAQITAAVALVEAKQARSDIEKLKEAI
    ALGVATSAQITAAVALVEAKQARSDIEKLKEAIR
  • The one letter amino acid code of Table 1 is used.
  • TABLE 15
    HUMAN PARAINFLUENZA VIRUS 3 (HPV3) F1 REGION
    DP107 ANALOG AMINO TRUNCATIONS
    IRD
    AIRD
    EAIRD
    KEAIRD
    LKEAIRD
    KLKEAIRD
    EKLKEAIRD
    IEKLKEAIRD
    DIEKLKEAIRD
    SDIEKLKEAIRD
    RSDIEKLKEAIRD
    ARSDIEKLKEAIRD
    QARSDIEKLKEAIRD
    KQARSDIEKLKEAIRD
    AKQARSDIEKLKEAIRD
    EAKQARSDIEKLKEAIRD
    VEAKQARSDIEKLKEAIRD
    LVEAKQARSDIEKIKEAIRD
    ALVEAKQARSDIEKLKEAIRD
    VALVEAKQARSDIEKLKEAIRD
    AVALVEAKQARSDIEKLKEAIRD
    AAVALVEAKQARSDIEKLKEAIRD
    TAAVALVEAKQARSDIEKLKEAIRD
    ITAAVALVEAKQARSDIEKLKEAIRD
    QITAAVALVEAKQARSDIEKLKEAIRD
    AQITAAVALVEAKQARSDIEKLKEAIRD
    SAQITAAVALVEAKQARSDIEKLKEAIRD
    TSAQITAAVALVEAKQARSDIEKLKEAIRD
    ATSAQITAAVALVEAKQARSDLEKLKEAIRD
    VATSAQITAAVALVEAKQARSDIEKLKEAIRD
    GVATSAQITAAVALVEAKQARSDIEKLKEAIRD
    LGVATSAQITAAVALVEAKQARSDIEKLKEAIRD
  • The one letter amino acid code of Table 1 is used.
  • TABLE 16
    ANTI-RESPIRATORY SYNCYTIAL VIRUS (RSV) PEPTIDES
    TSVITIELSNIKENKCNGTDAKVKLIKQELDKYKN
    SVITIELSNIKENKCNGTDAKVKLIKQELDKYKNA
    VITIELSNIKENKCNGTDAKVKLIKQELDKYKNAV
    VAVSKVLHLEGEVNKIALLSTNKAVVSLSNGVS
    AVSKVLHLEGEVNKIALLSTNKAVVSLSNGVSV
    VSKVLHLEGEVNKIALLSTNKAVVSLSNGVSVL
    SKVLHLEGEVNKIALLSTNKAVVSLSNGVSVLT
    KVLHLEGEVNKIALLSTNKAVVSLSNGVSVLTS
    LEGEVNKIALLSTNKAVVSLSNGVSVLTSKVLD
    GEVNKIALLSTNKAVVSLSNGVSVLTSKVLDLK
    EVNKIALLSTNKAVVSLSNGVSVLTSKVLDLKN
    VNKIALLSTNKAVVSLSNGVSVLTSKVLDLKNY
    NKIALLSTNKAVVSLSNGVSVLTSKVLDLKNYI
    KIALLSTNKAVVSLSNGVSVLTSKVLDLKNYID
    IALLSTNKAVVSLSNGVSVLTSKVLDLKNYIDK
    ALLSTNKAVVSLSNGVSVLTSKVLDLKNYIDKQ
    VAVSKVLHLEGEVNKIALLSTNKAVVSLSNGVS
    AVSKVLHLEGEVNKIALLSTNKAVVSLSNGVSV
    VSKVLHLEGEVNKIALLSTNKAVVSLSNGVSVL
    SKVLHLEGEVNKIALLSTNKAVVSLSNGVSVLT
    KVLHLEGEVNKIALLSTNKAVVSLSNGVSVLTS
    LEGEVNKIALLSTNKAVVSLSNGVSVLTSKVLD
    GEVNKIALLSTNKAVVSLSNGVSVLTSKVLDLK
    EVNKIALLSTNKAVVSLSNGVSVLTSKVLDLKN
    VNKIALLSTNKAVVSLSNGVSVLTSKVLDLKNY
    NKIALLSTNKAVVSLSNGVSVLTSKVLDLKNYI
    KIALLSTNKAVVSLSNGVSVLTSKVLDLKNYID
    IALLSTNKAVVSLSNGVSVLTSKVLDLKNYIDK
    ALLSTNKAVVSLSNGVSVLTSKVLDLKNYIDKQ
  • The one letter amino acid code of Table 1 is used.
  • TABLE 17
    ANTI-HUMAN PARAINFLUENZA VIRUS 3 (HPV3) PEPTIDES
    TLNNSVALDPIDISIELNKAKSDLEESKEWIRRSN
    LNNSVALDPIDISIELNKAKSDLEESKEWIRRSNQ
    NNSVALDPIDISIELNKAKSDLEESKEWIRRSNQK
    NSVALDPIDISIELNKAKSDLEESKEWIRRSNQKL
    SVALDPIDISIELNKAKSDLEESKEWIRRSNQKLD
    VALDPIDISIELNKAKSDLEESKEWIRRSNQKLDS
    ALDPIDISIELNKAKSDLEESKEWIRRSNQKLDSI
    LDPIDISIELNKAKSDLEESKEWIRRSNQKLDSIG
    DPIDISIELNKAKSDLEESKEWIRRSNQKLDSIGN
    PIDISIELNKAKSDLEESKEWIRRSNQKLDSIGNW
    IDISIELNKAKSDLEESKEWIRRSNQKLDSIGNWH
    DISIELNKAKSDLEESKEWIRRSNQKLDSIGNWHQ
    ISIELNKAKSDLEESKEWIRRSNQKLDSIGNWHQS
    SIELNKAKSDLEESKEWIRRSNQKLDSIGNWHQSS
    IELNKAKSDLEESKEWIRRSNQKLDSIGNWHQSST
    ELNKAKSDLEESKEWIRRSNQKLDSIGNWHQSSTT
    TAAVALVEAKQARSDIEKLKEAIRDTNKAVQSVQS
    AVALVEAKQARSDIEKLKEAIRDTNKAVQSVQSSI
    LVEAKQARSDIEKLKEAIRDTNKAVQSVQSSIGNL
    VEAKQARSDIEKLKEAIRDTNKAVQSVQSSIGNLI
    EAKQARSDIEKLKEAIRDTNKAVQSVQSSIGNLIV
    AKQARSDIEKLKEAIRDTNKAVQSVQSSIGNLIVA
    KQARSDIEKLKEAIRDTNKAVQSVQSSIGNLIVAI
    QARSDIEKLKEAIRDTNKAVQSVQSSIGNLIVAIK
    ARSDIEKLKEAIRDTNKAVQSVQSSIGNLIVAIKS
    RSDIEKLKEAIRDTNKAVQSVQSSIGNLIVAIKSV
    SDIEKLKEAIRDTNKAVQSVQSSIGNLIVAIKSVQ
    KLKEAIRDTNKAVQSVQSSIGNLIVAIKSVQDYVN
    LKEAIRDTNKAVQSVQSSIGNLIVAIKSVQDYVNK
    AIRDTNKAVQSVQSSIGNLIVAIKSVQDYVNKEIV
  • The one letter amino acid code of Table 1 is used.
  • TABLE 18
    ANTI-SIMIAN IMMUNODEFICIENCY VIRUS (SIV) PEPTIDES
    WQEWERKVDFLEENITALLEEAQIQQEKNMYELQK
    QEWERKVDFLEENITALLEEAQIQQEKNMYELQKL
    EWERKVDFLEENITALLEEAQIQQEKNMYELQKLN
    WERKVDFLEENITALLEEAQIQQEKNMYELQKLNS
    ERKVDFLEENITALLEEAQIQQEKNMYELQKLNSW
    RKVDFLEENITALLEEAQIQQEKNMYELQKLNSWD
    KVDFLEENITALLEEAQIQQEKNMYELQKLNSWDV
    VDFLEENITALLEEAQIQQEKNMYELQKLNSWDVF
    DFLEENITALLEEAQIQQEKNMYELQKLNSWDVFG
    FLEENITALLEEAQIQQEKNMYELQKLNSWDVFGN
  • The one letter amino acid code of Table 1 is used.
  • TABLE 19
    ANTI-MEASLES VIRUS (MEV) PEPTIDES
    LHRIDLGPPISLERLDVGTNLGNAIAKLEAKELL
    HRIDLGPPISLERLDVGTNLGNAIAKLEAKELLE
    RIDLGPPISLERLDVGTNLGNAIAKLEAKELLES
    IDLGPPISLERLDVGTNLGNAIAKLEAKELLESS
    DLGPPISLERLDVGTNLGNAIAKLEAKELLESSD
    LGPPISLERLDVGTNLGNAIAKLEAKELLESSDQ
    GPPISLERLDVGTNLGNAIAKLEAKELLESSDQI
    PPISLERLDVGTNLGNAIAKLEAKELLESSDQIL
    PISLERLDVGTNLGNAIAKLEAKELLESSDQILR
    SLERLDVGTNLGNAIAKLEAKELLESSDQILRSM
    LERLDVGTNLGNAIAKLEAKELLESSDQILRSMK
  • The one letter amino acid code of Table 1 is used.

Claims (91)

1. A modified anti-viral peptide comprising:
a peptide that exhibits anti-viral activity against respiratory syncytial virus (RSV), and
a reactive group which is reactive with amino groups, hydroxyl groups, or thiol groups on blood components to form stable covalent bonds.
2. The modified peptide of claim 1, wherein said reactive group is a succinimidyl or a maleimido group.
3. The modified peptide of claim 1, wherein said reactive group is a maleimido group which is reactive with a thiol group on a blood protein.
4.-8. (canceled)
9. The modified peptide of claim 1, wherein said peptide is selected from the group consisting of SEQ ID NO: 10 to SEQ ID NO: 30.
10. The modified peptide of claim 1, wherein said peptide is selected from the group consisting of SEQ ID N0: 14 to SEQ ID N0: 17 and SEQ ID NO: 29.
11. A modified anti-viral peptide comprising:
a peptide exhibits antiviral activity against human parainfluenza virus (HPIV), and
a reactive group which is reactive with amino groups, hydroxyl groups, or thiol groups on blood components to form stable covalent bonds.
12. The modified peptide of claim 11, wherein said peptide is selected from the group consisting of SEQ ID N0: 31 to SEQ ID N0: 62.
13. The modified peptide of claim 11, wherein said peptide is selected from the group consisting of SEQ ID NO: 35, SEQ ID N0: 38 to SEQ ID N0: 42, SEQ ID N0: 52 and SEQ ID N0: 58.
14. A modified anti-viral peptide comprising:
a peptide exhibits antiviral activity against measles virus (MeV), and
a reactive group which is reactive with amino groups, hydroxyl groups, or thiol groups on blood components to form stable covalent bonds.
15. The modified peptide of claim 14, wherein said peptide is selected from the group consisting of SEQ ID N0: 74 to SEQ ID N0: 86.
16. The modified peptide of claim 14, wherein said peptide is selected from the group consisting of SEQ ID N0: 77, SEQ ID N0: 79, SEQ ID N0: 81 and SEQ ID N0: 84.
17. A modified anti-viral peptide comprising:
a peptide exhibits antiviral activity against simian immunodeficiency virus (SIV), and
a reactive group which is reactive with amino groups, hydroxyl groups, or thiol groups on blood components to form stable covalent bonds.
18. The modified peptide of claim 17, wherein said peptide is selected from the group consisting of SEQ ID N0: 63 to SEQ ID N0: 73.
19. (canceled)
20. The modified peptide of claim 11, wherein said reactive group is a maleimido group which is reactive with a thiol group on a blood protein.
21.-22. (canceled)
23. The modified peptide of claim 14, wherein said reactive group is a maleimido group which is reactive with a thiol group on a blood protein.
24.-25. (canceled)
26. The modified peptide of claim 17, wherein said reactive group is a maleimido group which is reactive with a thiol group on a blood protein.
27.-30. (canceled)
31. The modified peptide of claim 11, wherein said reactive group is a succinimidyl or a maleimido group.
32. The modified peptide of claim 14, wherein said reactive group is a succinimidyl or a maleimido group.
33. The modified peptide of claim 17, wherein said reactive group is a succinimidyl or a maleimido group.
34. An anti-viral peptide-albumin conjugate comprising:
a peptide that exhibits anti-viral activity against respiratory syncytial virus (RSV) that comprises an amino acid sequence and a reactive group which is reactive with amino groups, hydroxyl groups, or thiol groups on blood components to form stable covalent bonds; and
albumin, wherein the peptide is covalently bonded to the albumin through the reactive group.
35. The conjugate of claim 34, wherein said reactive group is a maleimido group which is reactive with a thiol group on the albumin.
36. The conjugate of claim 34, wherein said peptide is selected from the group consisting of SEQ ID NO: 10 to SEQ ID NO: 30.
37. The conjugate of claim 34, wherein said peptide is selected from the group consisting of SEQ ID N0: 14 to SEQ ID N0: 17 and SEQ ID NO: 29.
38. An anti-viral peptide-albumin conjugate comprising:
a peptide that exhibits anti-viral activity against human parainfluenza virus (HPIV), that comprises an amino acid sequence and a reactive group which is reactive with amino groups, hydroxyl groups, or thiol groups on blood components to form stable covalent bonds; and
albumin, wherein the peptide is covalently bonded to the albumin through the reactive group.
39. The conjugate of claim 38, wherein said reactive group is a maleimido group which is reactive with a thiol group on the albumin.
40. The conjugate of claim 38, wherein said peptide is selected from the group consisting of SEQ ID N0: 31 to SEQ ID N0: 62.
41. The conjugate of claim 38, wherein said peptide is selected from the group consisting of SEQ ID N0: 35, SEQ ID N0: 38 to 42, SEQ ID N0: 52 and SEQ ID N0: 58.
42. An anti-viral peptide-albumin conjugate comprising:
a peptide that exhibits anti-viral activity against measles virus (MeV), that comprises an amino acid sequence and a reactive group which is reactive with amino groups, hydroxyl groups, or thiol groups on blood components to form stable covalent bonds; and
albumin, wherein the peptide is covalently bonded to the albumin through the reactive group.
43. The conjugate of claim 42, wherein said reactive group is a maleimido group which is reactive with a thiol group on the albumin.
44. The conjugate of claim 42, wherein said peptide is selected from the group consisting of SEQ ID N0: 74 to SEQ ID N0: 86.
45. The conjugate of claim 42 wherein said peptide is selected from the group consisting of SEQ ID N0: 77, SEQ ID N0: 79, SEQ ID N0: 81 and SEQ ID N0: 84.
46. An anti-viral peptide-albumin conjugate comprising:
a peptide that exhibits anti-viral activity against simian immunodeficiency virus (SIV), that comprises an amino acid sequence and a reactive group which is reactive with amino groups, hydroxyl groups, or thiol groups on blood components to form stable covalent bonds; and
albumin, wherein the peptide is covalently bonded to the albumin through the reactive group.
47. The conjugate of claim 46, wherein said reactive group is a maleimido group which is reactive with a thiol group on the albumin.
48. The conjugate of claim 46, wherein said peptide is selected from the group consisting of SEQ ID N0: 63 to SEQ ID N0: 73.
49. A composition comprising the modified anti-viral peptide of claim 1 and a physiologically acceptable medium.
50. A composition comprising the modified anti-viral peptide of claim 11 and a physiologically acceptable medium.
51. A composition comprising the modified anti-viral peptide of claim 14 and a physiologically acceptable medium.
52. A composition comprising the modified anti-viral peptide of claim 17 and a physiologically acceptable medium.
53. A composition comprising the conjugate of claim 34 and a physiologically acceptable medium.
54. A composition comprising the conjugate of claim 38 and a physiologically acceptable medium.
55. A composition comprising the conjugate of claim 42 and a physiologically acceptable medium.
56. A composition comprising the conjugate of claim 46 and a physiologically acceptable medium.
57. A method of inhibiting or reducing membrane fusion between respiratory syncytial virus (RSV) and a cell, comprising contacting the RSV with a modified peptide of claim 1.
58. A method of inhibiting or reducing membrane fusion between human parainfluenze virus (HPIV) and a cell, comprising contacting the HPIV with a modified peptide of claim 11.
59. A method of inhibiting or reducing membrane fusion between measles virus (MeV) and a cell, comprising contacting the MeV with a modified peptide of claim 14.
60. A method of inhibiting or reducing membrane fusion between simian immunodeficiency virus (SIV) and a cell, comprising contacting the SIV with a modified peptide of claim 17.
61. A method of inhibiting or reducing membrane fusion between respiratory syncytial virus (RSV) and a cell, comprising contacting the RSV with a conjugate of claim 34.
62. A method of inhibiting or reducing membrane fusion between human parainfluenze virus (HPIV) and a cell, comprising contacting the HPIV with a conjugate of claim 38.
63. A method of inhibiting or reducing membrane fusion between measles virus (MeV) and a cell, comprising contacting the MeV with a conjugate of claim 42.
64. A method of inhibiting or reducing membrane fusion between simian immunodeficiency virus (SIV) and a cell, comprising contacting the SIV with a conjugate of claim 46.
65. A method of making a conjugate of claim 34, comprising:
contacting a modified peptide of claim 1 with albumin to form an anti-viral peptide-albumin conjugate.
66. The method of claim 65, wherein the modified anti-viral peptide is made by combining an anti-RSV peptide having a free amino group, N-[γ-maleimidobutyryloxy]succinimide ester (GMBS) and trietylamine or maleimidopropionic acid (MPA) to form a maleimide containing group at the free amino group, to form the modified anti-viral peptide.
67. A method of making a conjugate of claim 38, comprising:
contacting a modified peptide of claim 11 with albumin to form an anti-viral peptide-albumin conjugate.
68. The method of claim 67, wherein the modified anti-viral peptide is made by combining an anti-HPIV peptide having a free amino group, N-[γ-maleimidobutyryloxy]succinimide ester (GMBS) and trietylamine or maleimidopropionic acid (MPA) to form a maleimide containing group at the free amino group, to form the modified anti-viral peptide.
69. A method of making a conjugate of claim 42, comprising:
contacting a modified peptide of claim 14 with albumin to form an anti-viral peptide-albumin conjugate.
70. The method of claim 69, wherein the modified anti-viral peptide is made by combining an anti-MeV peptide having a free amino group, N-[γ-maleimidobutyryloxy]succinimide ester (GMBS) and trietylamine or maleimidopropionic acid (MPA) to form a maleimide containing group at the free amino group, to form the modified anti-viral peptide.
71. A method of making a conjugate of claim 46, comprising:
contacting a modified peptide of claim 17 with albumin to form an anti-viral peptide-albumin conjugate.
72. The method of claim 71, wherein the modified anti-viral peptide is made by combining an anti-SIV peptide having a free amino group, N-[γ-maleimidobutyryloxy]succinimide ester (GMBS) and trietylamine or maleimidopropionic acid (MPA) to form a maleimide containing group at the free amino group, to form the modified anti-viral peptide.
73. A method of treating or preventing respiratory syncytial virus (RSV) infection in a subject, comprising
administering a modified peptide of claim 1 to a subject having or at risk of RSV infection, thereby treating or preventing the infection.
74. The method of claim 73, wherein the subject has brochiolitis.
75. The method of claim 73, wherein the subject has pneumonia.
76. The method of claim 73, wherein the modified peptide is conjugated with albumin in vivo.
77. The method of claim 73, wherein the modified peptide is conjugated with albumin ex vivo.
78. A method of treating or preventing respiratory syncytial virus (RSV) infection in a subject, comprising
administering a conjugate of claim 34 to a subject having or at risk of RSV infection, thereby treating or preventing the infection.
79. The method of claim 78, wherein the subject has brochiolitis.
80. The method of claim 78, wherein the subject has pneumonia.
81. A method of treating or preventing human parainfluenza virus (HPIV) infection in a subject, comprising
administering a modified peptide of claim 11 to a subject having or at risk of HPIV infection, thereby treating or preventing the infection.
82. The method of claim 81, wherein the subject has respiratory tract disease.
83. The method of claim 81, wherein the subject has croup.
84. The method of claim 81, wherein the subject has brochiolitis.
85. The method of claim 81, wherein the subject has pneumonia.
86. The method of claim 81, wherein the modified peptide is conjugated with albumin in vivo.
87. The method of claim 81, wherein the modified peptide is conjugated with albumin ex vivo.
88. A method of treating or preventing human parainfluenza virus (HPIV) infection in a subject, comprising
administering a conjugate of claim 38 to a subject having or at risk of HPIV infection, thereby treating or preventing the infection.
89. The method of claim 88, wherein the subject has respiratory tract disease.
90. The method of claim 88, wherein the subject has croup.
91. The method of claim 88, wherein the subject has brochiolitis.
92. The method of claim 88, wherein the subject has pneumonia.
93. A method of treating or preventing measles virus (MeV) infection in a subject, comprising
administering a modified peptide of claim 14 to a subject having or at risk of MeV infection, thereby treating or preventing the infection.
94. The method of claim 93, wherein the modified peptide is conjugated with albumin in vivo.
95. The method of claim 93, wherein the modified peptide is conjugated with albumin ex vivo.
96. A method of treating or preventing measles virus (MeV) infection in a subject, comprising
administering a conjugate of claim 42 to a subject having or at risk of MeV infection, thereby treating or preventing the infection.
97. A method of treating or preventing simian immunodeficiency virus (SIV) infection in a subject, comprising
administering a modified peptide of claim 17 to a subject having or at risk of SIV infection, thereby treating or preventing the infection.
98. The method of claim 97, wherein the modified peptide is conjugated with albumin in vivo.
99. The method of claim 97, wherein the modified peptide is conjugated with albumin ex vivo.
100. A method of treating or preventing simian immunodeficiency virus (SIV) infection in a subject, comprising
administering a conjugate of claim 46 to a subject having or at risk of SIV infection, thereby treating or preventing the infection.
US11/877,221 1999-05-17 2007-10-23 Long lasting fusion peptide inhibitors of viral infection Abandoned US20080199483A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/877,221 US20080199483A1 (en) 1999-05-17 2007-10-23 Long lasting fusion peptide inhibitors of viral infection

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US13440699P 1999-05-17 1999-05-17
US15340699P 1999-09-10 1999-09-10
PCT/US2000/013651 WO2000069902A1 (en) 1999-05-17 2000-05-17 Long lasting fusion peptide inhibitors or viral infection
US09/623,533 US7582301B1 (en) 1999-05-17 2000-05-17 Long-lasting antiviral fusion inhibitor peptide conjugates comprising albumin and human immunodeficiency virus (HIV) peptides
US10/950,010 US7608271B2 (en) 1999-05-17 2004-09-24 Modified human immunodeficiency virus anti-fusogenic GP41 peptides comprising a maleimide-containing group
US11/877,221 US20080199483A1 (en) 1999-05-17 2007-10-23 Long lasting fusion peptide inhibitors of viral infection

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/950,010 Continuation US7608271B2 (en) 1999-05-17 2004-09-24 Modified human immunodeficiency virus anti-fusogenic GP41 peptides comprising a maleimide-containing group

Publications (1)

Publication Number Publication Date
US20080199483A1 true US20080199483A1 (en) 2008-08-21

Family

ID=26832297

Family Applications (4)

Application Number Title Priority Date Filing Date
US09/623,533 Expired - Fee Related US7582301B1 (en) 1999-05-17 2000-05-17 Long-lasting antiviral fusion inhibitor peptide conjugates comprising albumin and human immunodeficiency virus (HIV) peptides
US10/950,010 Expired - Fee Related US7608271B2 (en) 1999-05-17 2004-09-24 Modified human immunodeficiency virus anti-fusogenic GP41 peptides comprising a maleimide-containing group
US11/877,300 Abandoned US20080176794A1 (en) 1999-05-17 2007-10-23 Long lasting fusion peptide inhibitors of viral infection
US11/877,221 Abandoned US20080199483A1 (en) 1999-05-17 2007-10-23 Long lasting fusion peptide inhibitors of viral infection

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US09/623,533 Expired - Fee Related US7582301B1 (en) 1999-05-17 2000-05-17 Long-lasting antiviral fusion inhibitor peptide conjugates comprising albumin and human immunodeficiency virus (HIV) peptides
US10/950,010 Expired - Fee Related US7608271B2 (en) 1999-05-17 2004-09-24 Modified human immunodeficiency virus anti-fusogenic GP41 peptides comprising a maleimide-containing group
US11/877,300 Abandoned US20080176794A1 (en) 1999-05-17 2007-10-23 Long lasting fusion peptide inhibitors of viral infection

Country Status (14)

Country Link
US (4) US7582301B1 (en)
EP (3) EP1179012B2 (en)
JP (3) JP4216480B2 (en)
CN (1) CN1351611A (en)
AT (2) ATE443714T1 (en)
AU (1) AU761591B2 (en)
BR (1) BR0010757A (en)
CA (1) CA2372338A1 (en)
DE (2) DE60043021D1 (en)
DK (1) DK1264840T3 (en)
ES (2) ES2334106T3 (en)
HK (1) HK1053128A1 (en)
PT (1) PT1264840E (en)
WO (1) WO2000069902A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050065075A1 (en) * 2001-05-31 2005-03-24 Erickson John W. Long lasting fusion peptide inhibitors for hiv infection
US20050070475A1 (en) * 1999-05-17 2005-03-31 Conjuchem, Inc. Long lasting fusion peptide inhibitors of viral infection
US20080039532A1 (en) * 2004-05-06 2008-02-14 Dominique Bridon Compounds For Specific Viral Target
US20090088378A1 (en) * 2007-01-12 2009-04-02 Omar Quraishi Long lasting inhibitors of viral infection
US20090088377A1 (en) * 2007-05-16 2009-04-02 Conjuchem Biotechnologies, Inc. Cysteic acid derivatives of anti-viral peptides
WO2013150532A1 (en) 2012-04-04 2013-10-10 Yeda Research And Development Co. Ltd. Lipopeptide conjugates comprising sphingolipid and hiv gp41 derived peptides

Families Citing this family (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE252601T1 (en) 1999-05-17 2003-11-15 Conjuchem Inc LONG-ACTING INSULINOTROPE PEPTIDES
US6887470B1 (en) 1999-09-10 2005-05-03 Conjuchem, Inc. Protection of endogenous therapeutic peptides from peptidase activity through conjugation to blood components
US20090175821A1 (en) * 1999-05-17 2009-07-09 Bridon Dominique P Modified therapeutic peptides with extended half-lives in vivo
US6514500B1 (en) * 1999-10-15 2003-02-04 Conjuchem, Inc. Long lasting synthetic glucagon like peptide {GLP-!}
US7601691B2 (en) 1999-05-17 2009-10-13 Conjuchem Biotechnologies Inc. Anti-obesity agents
US20040266673A1 (en) * 2002-07-31 2004-12-30 Peter Bakis Long lasting natriuretic peptide derivatives
JP4280070B2 (en) 2001-02-16 2009-06-17 コンジュケム バイオテクノロジーズ インコーポレイテッド Long-lasting glucagon-like peptide 2 (GLP-2) for the treatment of gastrointestinal diseases and disorders
CN1255548C (en) 2001-06-15 2006-05-10 霍夫曼-拉罗奇有限公司 Acetylation of GP41 framgnets
JP5385497B2 (en) * 2002-09-24 2014-01-08 フロンティア バイオテクノロジーズ カンパニー リミテッド Peptide derivative fusion inhibitor for HIV infection
US7556813B2 (en) * 2002-09-27 2009-07-07 Trimeris, Inc. Antiviral peptide-polymer conjugate comprising a polymer covalently attached to two or more synthetic HIV gp41 HR1 and/or HR2 peptides
CA2443365C (en) * 2002-11-19 2010-01-12 F. Hoffmann-La Roche Ag Methods for the recombinant production of antifusogenic peptides
US8133733B2 (en) 2003-10-24 2012-03-13 Gencia Corporation Nonviral vectors for delivering polynucleotides to target tissues
US20090123468A1 (en) 2003-10-24 2009-05-14 Gencia Corporation Transducible polypeptides for modifying metabolism
US8062891B2 (en) 2003-10-24 2011-11-22 Gencia Corporation Nonviral vectors for delivering polynucleotides to plants
EP2418281B1 (en) 2003-10-24 2016-06-01 Gencia Corporation Methods and compositions for delivering polynucleotides
US8507277B2 (en) 2003-10-24 2013-08-13 Gencia Corporation Nonviral vectors for delivering polynucleotides
ES2347902T3 (en) * 2004-04-23 2010-11-22 Conjuchem Biotechnologies Inc. Canadian Corporation 4528590 SOLID PHASE FOR USE IN A PROCEDURE FOR PURIFICATION OF ALBUMINE CONJUGATES.
US7723063B2 (en) * 2004-04-28 2010-05-25 Intrinsic Lifesciences Methods for measuring levels of bioactive human hepcidin
US8039432B2 (en) * 2005-11-09 2011-10-18 Conjuchem, Llc Method of treatment of diabetes and/or obesity with reduced nausea side effect
AU2006329215A1 (en) * 2005-12-22 2007-06-28 Conjuchem Biotechnologies Inc. Process for the production of preformed conjugates of albumin and a therapeutic agent
CN100475270C (en) 2006-01-20 2009-04-08 清华大学 Medicine for treating tumor, and application thereof
CN101002945B (en) 2006-01-20 2012-09-05 清华大学 Novel complex used for treating tumor
WO2007097903A2 (en) 2006-02-02 2007-08-30 Trimeris, Inc. Hiv fusion inhibitor peptides with improved biological properties
CN100366633C (en) * 2006-04-18 2008-02-06 河北师范大学 Antiviral polypeptide from Zongdian Tuanwa frog and its application in pharmacy
TW200817438A (en) 2006-08-17 2008-04-16 Hoffmann La Roche A conjugate of an antibody against CCR5 and an antifusogenic peptide
BRPI0715794A2 (en) 2006-08-17 2013-07-23 Hoffmann La Roche ccr5 antibody conjugate and antifusogenic peptide
US20090099074A1 (en) * 2007-01-10 2009-04-16 Conjuchem Biotechnologies Inc. Modulating food intake
CL2008000707A1 (en) * 2007-03-13 2008-09-22 Hoffmann La Roche CONJUGATE OF ANTIFUSOGENIC POLYPEPTIDES AND POLYPEPTIDES DERIVED FROM THE GLOBULAR HEAD OF THE COMPLEMENT FACTOR C1Q; PHARMACEUTICAL COMPOSITION THAT UNDERSTANDS IT; ITS USE TO TREAT VIRIC INFECTIONS; AND PRODUCTION METHOD.
CL2008002092A1 (en) 2007-07-20 2009-05-29 Hoffmann La Roche Conjugate containing two or more antifusogenic peptides and an anti-cd-4 antibody; Method of production; pharmaceutical composition comprising it; antifusogenic polypeptides and use of the conjugate to treat viral infections.
EP2604623A3 (en) 2007-08-08 2013-10-02 Novozymes Biopharma DK A/S Transferrin variants and conjugates
EP2036980A1 (en) * 2007-09-14 2009-03-18 Gruber, Jens Down regulation of gene expression using virus-like particles charged with nucleic acid
US20090186819A1 (en) * 2007-12-11 2009-07-23 Marieve Carrier Formulation of insulinotropic peptide conjugates
JP5936112B2 (en) 2009-02-11 2016-06-15 アルブミディクス アクティーゼルスカブ Albumin variants and complexes
KR101425404B1 (en) * 2009-07-17 2014-08-01 한림대학교 산학협력단 Immunostimulatory Compositions Comprising Liposome―Encapsulated Oligonucleotides and Epitopes
CN105567699A (en) 2009-10-30 2016-05-11 诺维信生物制药丹麦公司 Albumin variants
CN106977608A (en) 2010-04-09 2017-07-25 阿尔布麦狄克斯公司 Albumin derivant and variant
EP2616487B1 (en) 2010-09-14 2015-03-25 F.Hoffmann-La Roche Ag Serpin-finger fusion polypeptide
WO2012059486A1 (en) 2010-11-01 2012-05-10 Novozymes Biopharma Dk A/S Albumin variants
US8822417B2 (en) 2011-05-05 2014-09-02 Novozymes Biopharma DIC A/S Albumin variants
WO2013039861A2 (en) 2011-09-12 2013-03-21 modeRNA Therapeutics Engineered nucleic acids and methods of use thereof
WO2013075066A2 (en) 2011-11-18 2013-05-23 Eleven Biotherapeutics, Inc. Proteins with improved half-life and other properties
BR112014018679A2 (en) 2012-03-16 2017-07-04 Novozymes Biopharma Dk As albumin variants
AU2013243949A1 (en) 2012-04-02 2014-10-30 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
CA2868398A1 (en) 2012-04-02 2013-10-10 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
MX2015005363A (en) 2012-11-08 2015-11-06 Novozymes Biopharma Dk As Albumin variants.
EP3318124A3 (en) 2013-02-16 2018-05-30 Albumedix A/S Pharmacokinetic animal model
EP2981334A4 (en) * 2013-04-03 2016-11-23 Univ California Compositions and methods for inhibiting viral activity
JP6306700B2 (en) 2013-11-01 2018-04-04 ユニバーシティ オブ オスロUniversity of Oslo Modified albumin and use thereof
CN103864921B (en) * 2014-01-16 2017-12-26 苏州大学 Folic acid adriamycin immune formulation of dual-target treating cancer and preparation method thereof
WO2015140591A1 (en) 2014-03-21 2015-09-24 Nordlandssykehuset Hf Anti-cd14 antibodies and uses thereof
WO2015164615A1 (en) 2014-04-24 2015-10-29 University Of Oslo Anti-gluten antibodies and uses thereof
WO2017029407A1 (en) 2015-08-20 2017-02-23 Albumedix A/S Albumin variants and conjugates
CA3004790A1 (en) 2015-11-10 2017-05-18 Visterra, Inc. Lipopolysaccharide binding antibody-antimicrobial peptide conjugates and uses thereof
CN110337590A (en) 2016-11-04 2019-10-15 奥胡斯大学 The identification and treatment of tumour characterized by neonatal Fc receptor is overexpressed
WO2018096396A1 (en) 2016-11-22 2018-05-31 University Of Oslo Albumin variants and uses thereof
AU2018209940A1 (en) 2017-01-18 2019-07-11 Visterra, Inc. Antibody molecule-drug conjugates and uses thereof
CN111303245B (en) * 2020-02-21 2023-06-27 成都奥达生物科技有限公司 Anti-syncytial virus membrane fusion inhibitor
EP4126064A1 (en) 2020-04-03 2023-02-08 Visterra, Inc. Antibody molecule-drug conjugates and uses thereof
WO2021211303A1 (en) * 2020-04-14 2021-10-21 Contrafect Corporation Antiviral, bacteriophage-derived polypeptides and their use against viruses

Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5116944A (en) * 1989-12-29 1992-05-26 Neorx Corporation Conjugates having improved characteristics for in vivo administration
US5464933A (en) * 1993-06-07 1995-11-07 Duke University Synthetic peptide inhibitors of HIV transmission
US5612034A (en) * 1990-10-03 1997-03-18 Redcell, Inc. Super-globuling for in vivo extended lifetimes
US5614487A (en) * 1993-05-28 1997-03-25 Genentech, Inc. Sustained release pharmaceutical composition
US5656480A (en) * 1992-07-20 1997-08-12 Duke University Compounds which inhibit HIV replication
US5876969A (en) * 1992-01-31 1999-03-02 Fleer; Reinhard Fusion polypeptides comprising human serum albumin, nucleic acids encoding same, and recombinant expression thereof
US6013263A (en) * 1993-06-07 2000-01-11 Trimeris, Inc. Measles virus peptides with antifusogenic and antiviral activities
US6017536A (en) * 1993-06-07 2000-01-25 Trimeris, Inc. Simian immunodeficiency virus peptides with antifusogenic and antiviral activities
US6063761A (en) * 1993-08-13 2000-05-16 Kings College London Hepatoselective pharmaceutical actives
US6103236A (en) * 1995-05-10 2000-08-15 Kyowa Hakko Kogyo Co., Ltd. Toxin conjugates
US6107489A (en) * 1998-03-17 2000-08-22 Conjuchem, Inc. Extended lifetimes in vivo renin inhibitors
US6150088A (en) * 1997-04-17 2000-11-21 Whitehead Institute For Biomedical Research Core structure of gp41 from the HIV envelope glycoprotein
US6258782B1 (en) * 1998-05-20 2001-07-10 Trimeris, Inc. Hybrid polypeptides with enhanced pharmacokinetic properties
US6342225B1 (en) * 1993-08-13 2002-01-29 Deutshces Wollforschungsinstitut Pharmaceutical active conjugates
US6518013B1 (en) * 1993-06-07 2003-02-11 Trimeris, Inc. Methods for the inhibition of epstein-barr virus transmission employing anti-viral peptides capable of abrogating viral fusion and transmission
US20040106589A1 (en) * 1996-05-22 2004-06-03 Protarga Pharmaceuticals, Inc. Fatty acid-pharmaceutical agent conjugates
US6818611B1 (en) * 1998-10-13 2004-11-16 University Of Georgia Research Foundation, Inc. Stabilized bioactive peptides and methods of identification, synthesis and use
US7307148B2 (en) * 2004-04-23 2007-12-11 Conjuchem Biotechnologies Inc. Method for purification of albumin conjugates
US20080039532A1 (en) * 2004-05-06 2008-02-14 Dominique Bridon Compounds For Specific Viral Target
US7365162B2 (en) * 1998-10-13 2008-04-29 University Of Georgia Research Foundation, Inc. Stabilized bioactive peptides and methods of identification, synthesis, and use
US20080176794A1 (en) * 1999-05-17 2008-07-24 Conjuchem Biotechnologies Inc. Long lasting fusion peptide inhibitors of viral infection
US20090175821A1 (en) * 1999-05-17 2009-07-09 Bridon Dominique P Modified therapeutic peptides with extended half-lives in vivo

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3719667A (en) 1970-08-24 1973-03-06 Lilly Co Eli Epimerization of 6-acylamido and 6-imido penicillin sulfoxide esters
US3840556A (en) 1971-05-28 1974-10-08 Lilly Co Eli Penicillin conversion by halogen electrophiles and anti-bacterials derived thereby
US4652629A (en) * 1985-07-03 1987-03-24 The Salk Institute For Biological Studies Synthetic peptide-based anti-rabies compositions and methods
DK0578728T3 (en) 1991-04-05 1999-04-12 Genentech Inc Platelet aggregation inhibitors, which have high specificity for GP IIbIIIa
EP0602290B1 (en) * 1992-12-04 1999-08-25 ConjuChem, Inc. Antibody-conjugated Hepatitis B surface antigen and use thereof
JP4086936B2 (en) 1996-10-03 2008-05-14 株式会社ブリヂストン Dummy wafer
JP2001522817A (en) * 1997-11-07 2001-11-20 コンジュケム,インコーポレーテッド Novel conjugate of opioid and endogenous carrier
ATE406577T1 (en) * 1997-11-07 2008-09-15 Conjuchem Biotechnologies Inc METHODS FOR SCREENING AFFINITY MARKER LIBRARIES
WO1999048536A2 (en) * 1998-03-23 1999-09-30 Conjuchem, Inc. Delivery of long lasting therapeutic agents by forming covalent attachments in vivo
DE19926154A1 (en) 1999-06-09 2000-12-14 Ktb Tumorforschungs Gmbh Process for the preparation of an injectable pharmaceutical preparation
DE19926475A1 (en) 1999-06-10 2000-12-14 Ktb Tumorforschungs Gmbh Carrier-drug conjugates
US7090851B1 (en) 1999-09-10 2006-08-15 Conjuchem Inc. Long lasting fusion peptide inhibitors of viral infection
DE60216151T2 (en) 2001-05-31 2007-09-27 ConjuChem Biotechnologies Inc., Montreal Long acting fusion peptide inhibitors against HIV infection

Patent Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5116944A (en) * 1989-12-29 1992-05-26 Neorx Corporation Conjugates having improved characteristics for in vivo administration
US5612034A (en) * 1990-10-03 1997-03-18 Redcell, Inc. Super-globuling for in vivo extended lifetimes
US5876969A (en) * 1992-01-31 1999-03-02 Fleer; Reinhard Fusion polypeptides comprising human serum albumin, nucleic acids encoding same, and recombinant expression thereof
US5656480A (en) * 1992-07-20 1997-08-12 Duke University Compounds which inhibit HIV replication
US5614487A (en) * 1993-05-28 1997-03-25 Genentech, Inc. Sustained release pharmaceutical composition
US5464933A (en) * 1993-06-07 1995-11-07 Duke University Synthetic peptide inhibitors of HIV transmission
US6013263A (en) * 1993-06-07 2000-01-11 Trimeris, Inc. Measles virus peptides with antifusogenic and antiviral activities
US6017536A (en) * 1993-06-07 2000-01-25 Trimeris, Inc. Simian immunodeficiency virus peptides with antifusogenic and antiviral activities
US6518013B1 (en) * 1993-06-07 2003-02-11 Trimeris, Inc. Methods for the inhibition of epstein-barr virus transmission employing anti-viral peptides capable of abrogating viral fusion and transmission
US6342225B1 (en) * 1993-08-13 2002-01-29 Deutshces Wollforschungsinstitut Pharmaceutical active conjugates
US6063761A (en) * 1993-08-13 2000-05-16 Kings College London Hepatoselective pharmaceutical actives
US6103236A (en) * 1995-05-10 2000-08-15 Kyowa Hakko Kogyo Co., Ltd. Toxin conjugates
US20040106589A1 (en) * 1996-05-22 2004-06-03 Protarga Pharmaceuticals, Inc. Fatty acid-pharmaceutical agent conjugates
US6150088A (en) * 1997-04-17 2000-11-21 Whitehead Institute For Biomedical Research Core structure of gp41 from the HIV envelope glycoprotein
US6107489A (en) * 1998-03-17 2000-08-22 Conjuchem, Inc. Extended lifetimes in vivo renin inhibitors
US6258782B1 (en) * 1998-05-20 2001-07-10 Trimeris, Inc. Hybrid polypeptides with enhanced pharmacokinetic properties
US6818611B1 (en) * 1998-10-13 2004-11-16 University Of Georgia Research Foundation, Inc. Stabilized bioactive peptides and methods of identification, synthesis and use
US7365162B2 (en) * 1998-10-13 2008-04-29 University Of Georgia Research Foundation, Inc. Stabilized bioactive peptides and methods of identification, synthesis, and use
US20080176794A1 (en) * 1999-05-17 2008-07-24 Conjuchem Biotechnologies Inc. Long lasting fusion peptide inhibitors of viral infection
US20090175821A1 (en) * 1999-05-17 2009-07-09 Bridon Dominique P Modified therapeutic peptides with extended half-lives in vivo
US7582301B1 (en) * 1999-05-17 2009-09-01 Conjuchem Biotechnologies, Inc. Long-lasting antiviral fusion inhibitor peptide conjugates comprising albumin and human immunodeficiency virus (HIV) peptides
US7307148B2 (en) * 2004-04-23 2007-12-11 Conjuchem Biotechnologies Inc. Method for purification of albumin conjugates
US20080039532A1 (en) * 2004-05-06 2008-02-14 Dominique Bridon Compounds For Specific Viral Target

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050070475A1 (en) * 1999-05-17 2005-03-31 Conjuchem, Inc. Long lasting fusion peptide inhibitors of viral infection
US20080176794A1 (en) * 1999-05-17 2008-07-24 Conjuchem Biotechnologies Inc. Long lasting fusion peptide inhibitors of viral infection
US7608271B2 (en) 1999-05-17 2009-10-27 Conjuchem Biotechnologies Inc. Modified human immunodeficiency virus anti-fusogenic GP41 peptides comprising a maleimide-containing group
US20050065075A1 (en) * 2001-05-31 2005-03-24 Erickson John W. Long lasting fusion peptide inhibitors for hiv infection
US7741453B2 (en) 2001-05-31 2010-06-22 Conjuchem Biotechnologies, Inc. Long lasting fusion peptide inhibitors for HIV infection
US20080039532A1 (en) * 2004-05-06 2008-02-14 Dominique Bridon Compounds For Specific Viral Target
US20090088378A1 (en) * 2007-01-12 2009-04-02 Omar Quraishi Long lasting inhibitors of viral infection
US20090088377A1 (en) * 2007-05-16 2009-04-02 Conjuchem Biotechnologies, Inc. Cysteic acid derivatives of anti-viral peptides
WO2013150532A1 (en) 2012-04-04 2013-10-10 Yeda Research And Development Co. Ltd. Lipopeptide conjugates comprising sphingolipid and hiv gp41 derived peptides

Also Published As

Publication number Publication date
EP2110381A1 (en) 2009-10-21
US20050070475A1 (en) 2005-03-31
US7608271B2 (en) 2009-10-27
WO2000069902A1 (en) 2000-11-23
EP1179012B2 (en) 2009-07-15
ES2185595T3 (en) 2003-05-01
ATE226593T1 (en) 2002-11-15
ATE443714T1 (en) 2009-10-15
BR0010757A (en) 2002-02-19
CA2372338A1 (en) 2000-11-23
JP2002544287A (en) 2002-12-24
AU761591B2 (en) 2003-06-05
EP1179012B1 (en) 2002-10-23
US7582301B1 (en) 2009-09-01
ES2185595T5 (en) 2009-12-17
HK1053128A1 (en) 2003-10-10
DE60000665T3 (en) 2009-10-29
DK1264840T3 (en) 2009-11-16
PT1264840E (en) 2010-01-04
JP2009167208A (en) 2009-07-30
EP1264840B1 (en) 2009-09-23
CN1351611A (en) 2002-05-29
US20080176794A1 (en) 2008-07-24
JP4216480B2 (en) 2009-01-28
EP1179012A1 (en) 2002-02-13
JP2008101021A (en) 2008-05-01
EP1179012B9 (en) 2003-09-10
DE60000665T2 (en) 2003-06-26
AU5027100A (en) 2000-12-05
DE60000665D1 (en) 2002-11-28
EP1264840A1 (en) 2002-12-11
ES2334106T3 (en) 2010-03-05
DE60043021D1 (en) 2009-11-05

Similar Documents

Publication Publication Date Title
US7582301B1 (en) Long-lasting antiviral fusion inhibitor peptide conjugates comprising albumin and human immunodeficiency virus (HIV) peptides
US7741453B2 (en) Long lasting fusion peptide inhibitors for HIV infection
US7090851B1 (en) Long lasting fusion peptide inhibitors of viral infection
US20090088377A1 (en) Cysteic acid derivatives of anti-viral peptides
US20090088378A1 (en) Long lasting inhibitors of viral infection
EP1479691B1 (en) Long lasting fusion peptide inhibitors for HIV infection
EP1752469B1 (en) Long lasting fusion peptide inhibitors for HIV infection
US20080312415A1 (en) Long lasting fusion peptide inhibitors for hiv infection
WO2008144590A2 (en) Long lasting modified antifusogenic peptide for preventing hiv infection

Legal Events

Date Code Title Description
AS Assignment

Owner name: 4523482 CANADA INC., CANADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CONJUCHEM BIOTECHNOLOGIES INC.;REEL/FRAME:023594/0067

Effective date: 20090825

Owner name: 4523482 CANADA INC.,CANADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CONJUCHEM BIOTECHNOLOGIES INC.;REEL/FRAME:023594/0067

Effective date: 20090825

AS Assignment

Owner name: CONJUCHEM BIOTECHNOLOGIES, INC., CANADA

Free format text: CHANGE OF NAME;ASSIGNOR:4523482 CANADA INC.;REEL/FRAME:023639/0162

Effective date: 20090825

Owner name: CONJUCHEM BIOTECHNOLOGIES, INC.,CANADA

Free format text: CHANGE OF NAME;ASSIGNOR:4523482 CANADA INC.;REEL/FRAME:023639/0162

Effective date: 20090825

AS Assignment

Owner name: ADVANCED DIAGNOSTICS AND DISCOVERY, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CONJUCHEM BIOTECHNOLOGIES INC.;REEL/FRAME:025198/0735

Effective date: 20100823

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: CONJUCHEM, LLC, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ADVANCED DIAGNOSTICS AND DISCOVERY;REEL/FRAME:026157/0022

Effective date: 20110218