WO2011133658A1 - Compositions et procédés de ciblage et d'administration d'agents thérapeutiques dans des cellules - Google Patents

Compositions et procédés de ciblage et d'administration d'agents thérapeutiques dans des cellules Download PDF

Info

Publication number
WO2011133658A1
WO2011133658A1 PCT/US2011/033229 US2011033229W WO2011133658A1 WO 2011133658 A1 WO2011133658 A1 WO 2011133658A1 US 2011033229 W US2011033229 W US 2011033229W WO 2011133658 A1 WO2011133658 A1 WO 2011133658A1
Authority
WO
WIPO (PCT)
Prior art keywords
receptor
growth factor
compound
cell
copi
Prior art date
Application number
PCT/US2011/033229
Other languages
English (en)
Inventor
John R. Murphy
Original Assignee
Boston Medical Center Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Boston Medical Center Corporation filed Critical Boston Medical Center Corporation
Publication of WO2011133658A1 publication Critical patent/WO2011133658A1/fr

Links

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/6415Toxins or lectins, e.g. clostridial toxins or Pseudomonas exotoxins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/66Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid the modifying agent being a pre-targeting system involving a peptide or protein for targeting specific cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0002General or multifunctional contrast agents, e.g. chelated agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/34Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Corynebacterium (G)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/55Fusion polypeptide containing a fusion with a toxin, e.g. diphteria toxin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/74Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3513Protein; Peptide

Definitions

  • X is selected from a cytotoxic agent, a therapeutic agent, and a diagnostic agent and comprises at least one thiol-containing group capable of forming a disulfide bond with Y;
  • Z is a polypeptide targeting moiety that is bound to Y at its carboxy-terminal end.
  • X is selected from a siRNA, dsRNA, an RNAi molecule, a protein nucleic acid (PNA) molecule, and a polypeptide (e.g., a transcription factor or growth factor).
  • the lysine-rich domain includes one or more lysine rich motifs having a dibasic signature selected from KKXX and KXKXX, or aromatic amino acid sequences selected from FFXXBB(X) repeat,.
  • the methods of the invention include the administration of this composition to treat cytomegalovirus infections in the eye, e.g., in patients with HIV.
  • a detectable label when coupled to a conjuage compound of the invention emits a signal that can be detected by a signal transducing machine.
  • the detectable label can emit a signal spontaneously, such as when the detectable label is a radionuclide.
  • the detectable label emits a signal as a result of being stimulated by an external field such as when the detectable label is a relaxivity metal.
  • signals include, without limitation, gamma rays, X-rays, visible light, infrared energy, and radio waves.
  • Examples of signal transducing machines include, without limitation, gamma cameras including SPECT/CT devices, PET scanners, fluorimeters, and Magnetic Resonance Imaging (MRI) machines.
  • a “pharmaceutically acceptable excipient” is meant a carrier that is physiologically acceptable to the treated mammal while retaining the therapeutic properties of the compound with which it is administered.
  • One exemplary pharmaceutically acceptable excipient is physiological saline.
  • Other physiologically acceptable excipients and their formulations are known to one skilled in the art and described, for example, in “Remington: The Science and Practice of Pharmacy” (20th ed., ed. A.R. Gennaro AR., 2000, Lippincott Williams & Wilkins).
  • Figure 6 In silico prediction of 25 amino acid fragment of transmembrane domain helices 1 and 2 (DWDVIRDKTKTKISSLKEHGH) of diphtheria toxin.
  • the three dimensional model was constructed using the PEP-Fold algorithm (Maupetit et al, 2009) which is based on the structural alphabet SA letters to describe the conformation of four consecutive residues, and then couples the predicted series of SA letter to a greedy algorithm and course-grained force field.
  • the resulting structure was the visualized using the Accelrys DS Visualizer (v 2.0.1.7347).
  • EF, and botulinum neurotoxins serotypes A, C, and D on the cytoplasmic side of endosomal vesicle membranes can mimic cargo motifs and/or the KKXX and FFXX of adaptor proteins which are known to bind COPI complex proteins.
  • replacement of the native transmembrane helix 1 with the 13 amino acid COPI binding domain from the cytoplasmic tail of p23 results in a domain swap mutant whose cytotoxic potency is identical to that of the wild type toxin.
  • COPI complex binding to the N-terminal lysine- rich portion of the transmembrane domain of these toxins is an essential feature for C- domain delivery to the eukaryotic cell cytosol.
  • the COPI coatomer complex plays an essential role in the efficient delivery of toxins, such as DT and other toxins, e.g., anthrax LF and EF and botulinum neurotoxins serotypes A, C, and D, across the endosomal vesicle membrane and into the eukaryotic cell cytosol.
  • toxins such as DT and other toxins, e.g., anthrax LF and EF and botulinum neurotoxins serotypes A, C, and D
  • the requirements for COPI complex protein for both the DT C-domain and LF entry into the cytosol confirm that there is a common mechanism of entry for these highly divergent bacterial protein toxins.
  • Y is a polypeptide containing a lysine-rich domain that is capable of interacting with cellular COPI complex proteins and that includes at least one bond that is cleavable by an intracytosolic enzyme (e.g., the polypeptide may include a cysteine residue that forms a disulfide bond with the at least one thiol group (e.g., a cysteine residue) of the X moiety that is cleavable by an intracytosolic enzyme (e.g., thioredoxin reductase)), in which the lysine-rich domain includes one or more lysine rich motifs having a dibasic signature (e.g., KKXX, XKXX) and/or an aromatic amino acid sequences (e.g,.
  • the X moiety may also include a linker that connects the therapeutic agent (e.g., a siRNA, dsRNA, RNAi, a protein nucleic acid (PNA) molecule, or a protein agent (e.g., a transcription factors and other polypeptides)) to a thiol-containing group that can be used to form a disulfide bond with the thiol-containing group of the Y moiety.
  • the therapeutic agent e.g., a siRNA, dsRNA, RNAi, a protein nucleic acid (PNA) molecule, or a protein agent (e.g., a transcription factors and other polypeptides)
  • the transmembrane and receptor binding domains of the non- toxic DT mutant CRM 197 and the DT-related fusion protein toxins are used as structural platforms for the development of non-viral, cell receptor-specific siRNA cytosolic delivery systems. These systems employ their respective cell receptor- specific targeting and endosomal vesicle membrane pore forming ability and COPI binding motifs for enhanced delivery of peptide-siR As to the target cell cytosol.
  • the X moiety is prepared so that it is suitable for thiol-specific attachment via a free cysteine to the Y moiety of the conjugate compound.
  • Thiol-specific drug attachment to a peptide analog can be direct or indirect, i.e. via a chelator (e.g., MX-DTPA, which is useful in preparing the peptide analogs of the invention; the maleimido derivatives of MX-DTPA chelator is reactive with thiol groups of a peptide portion of the X moeity (i.e., SH groups of one or more free cysteines) to form a thioether linkage).
  • the thiol attachment methods of the present invention are generally applicable to the attachment of drugs/chelators to the Y/Z portion of the conjugate compound.
  • the thiol linkage can be a stable linkage, for example as a thioether linkage.
  • a drug or chelator is functionalized with a thiol reactive group (e.g., a maleimido group) that provides a stable thioether linkage.
  • a drug can comprise a cleavable site, such that the X moiety can be released from Y moiety (e.g., by reducing the disulfide bond).
  • representative cleavable sites include acid-labile and enzyme-labile sites.
  • Therapeutic agents that can be used as the "cargo" of the compounds of the invention include cytotoxic polypeptides, such as cytochrome c, caspase 1-10, granzyme A or B, tumor necrosis factor-alpha (TNF-a), TNF- ⁇ , Fas, Fas ligand, Fas-associated death doman-like IL- ⁇ ⁇ converting enzyme (FLICE), TRAIL/AP02L, TWEAK/AP03L, Bax, Bid, Bik, Bad, Bak, RICK, vascular apoptosis inducing proteins 1 and 2 (VAP1 and VAP2), pierisin, apoptosis-inducing protein (AIP), IL-l propiece polypeptide, apoptin, apoptin-associated protein 1 (AAP-1), endostatin, angiostatin, and biologically-active fragments thereof.
  • cytotoxic polypeptides such as cytochrome c, caspase 1-10, granzyme A
  • the X moiety can also be selected from nucleic acid molecules, e.g., siR As, dsRNAs, and other nucleic acid molecules that are known in the art to silence gene expression.
  • the nucleic acid molecules are those that silence genes that express polypeptides that are known to be involved in disease.
  • siRNA molecules for use in the treatment of diseases are known in the art (see, e.g., U.S. Patent Nos. 7,056,704; 7,678,896; 7,678,897; and 7691998; and U.S. Patent Application Publication Nos. 20100062051 (entitled Composition for Treatment of Cervix Cancer); 20100062436; 20100062951; 20100062967; 20100063131;
  • 20100063132 (entitled Small Interfering RNA and Pharmaceutical Composition for Treatment of Hepatitis B Comprising the Same); 20100063134 (entitled Treatment of Neurodegenerative Disease Through Intracranial Delivery of siRNA); and 20100063308; 20080249046; 20080260854; 20090318536; and 20100098664; each of which is incorporated by reference herein in their entirety).
  • Any of the siRNA molecules described in these publications can be used as the X moiety in the conjugate compounds of the invention for use in the treatment of diseases for which the siRNA molecules are known to treat or were developed to treat.
  • the X moiety may also be a transcription factor or a nucleic acid molecule that encodes a transcription factor.
  • the transcription factor can be selected from helix-turn- helix motif proteins, homeodomain proteins, zinc finger motif proteins, steroid receptor proteins, leucine zipper motif proteins, helix-loop-helix motif proteins, and ⁇ -sheet motif proteins.
  • the X moiety is a nucleic acid binding compound that binds nonspecifically to nucleic acids and is selected from the group consisting of poly- L-lysine, protamine, histone and spermine.
  • the X moiety is a nucleic acid binding domain that binds the coding region of a ribosome inactivating protein such as saporin.
  • Transcription factors for use as the X moiety in the preparation of conjugate compounds of the invention are known in the art (see, e.g., U.S. Patent No. 6,037,329, incorporated by reference herein in its entirety).
  • the X moiety may also be a growth factor polypeptide or a nucleic acid molecule that encodes the growth factor selected from leptin receptor (LPTR), granulocyte colony stimulating factor receptor (GCSFR), LIF/OSM/CNTF common beta chain (GPI30), leukemia inhibiting factor receptor (LIFR), oncostatin-M receptor beta chain (OSMR), interleukin-12 receptor beta-1 chain (IL12RB1), and interleukin-12 receptor beta-2 chain (IL 12RB2).
  • LPTR leptin receptor
  • GCSFR granulocyte colony stimulating factor receptor
  • LIF/OSM/CNTF common beta chain GPI30
  • LIF/OSM/CNTF common beta chain GPI30
  • LIF/OSM/CNTF common beta chain GPI30
  • OSMR leukemia inhibiting factor receptor
  • IL12RB1 interleukin-12 receptor beta-1 chain
  • IL 12RB2 interleukin-12 receptor beta-2 chain
  • IL12RB1 with IL12RB2 such as G-CSF, GM-CSF or M-CSF
  • SCF stem cell factor
  • SCPF stem cell proliferation factor
  • IL1 , IL4, IL5, IL6, IL11, IL12 various Interleukins
  • TGF- ⁇ , MIP-1- ⁇ , TNF-a and also many other low molecular weight factors.
  • the X moiety may also be a growth factor polypeptide or a nucleic acid molecule that encodes the growth factor (e.g., a growth factor selected from stem cell factor (SCF), FLT3, IL-3, IL-6, GSF, GM-CSF, and erythropoietin).
  • a growth factor selected from stem cell factor (SCF), FLT3, IL-3, IL-6, GSF, GM-CSF, and erythropoietin.
  • the conjugate compound can include a targeting moiety (Z) that targets the conjugate compound to a stem cell (e.g., a hemotopoeitic stem cell or a mesenchymal stem cell) and can include an X moiety, such as one of the growth factors described above, that promotes the differentiation of stem cells into a desired lineage (e.g., neuronal, hepatic, osteogenic, chondrogenic, tendonogenic, ligamentogenic, myogenic, marrow stromagenic, adipogenic or dermogenic lineage). Growth factors that stimulate the differentiation of stem cells are known in the art (see, e.g,. U.S. Patent No. 5,942,225, incorporated by reference herein).
  • Stem cells may also be differentiated using a conjugate compound of the invention into pancreatic islet cells (or primary cells of the islets of Langerhans), which may then be transplanted into a subject suffering from diabetes (e.g., diabetes mellitus, type 1), see e.g., Burns et al., (2006) Curr. Stem Cell Res. Ther., 2:255-266.
  • pancreatic beta cells derived from induced cells and differentiated using a conjugate compound of the invention may be transplanted into a subject suffering from diabetes (e.g., diabetes mellitus, type 1).
  • Coronaviridae family which includes the severe acute respiratory syndrome (SARS) virus
  • SARS severe acute respiratory syndrome
  • Rhabdoviridae family which includes the rabies virus and vesicular stomatitis virus (VSV)
  • VSV vesicular stomatitis virus
  • RSV human respiratory syncytial virus
  • RV human respiratory syncytial virus
  • NSV vesicular stomatitis virus
  • RV human respiratory syncytial virus
  • Newcastle disease virus hendravirus
  • nipahvirus measles virus
  • rindeipest virus canine distemper virus
  • Sendai virus human parainfluenza virus (e.g., 1, 2, 3, and 4)
  • rhinovirus and mumps virus
  • Picornaviridae family which includes the poliovirus, human enterovirus (A, B, C, and D), hepatitis A virus, and the coxsackievirus
  • Hepadnaviridae family which includes the hepatitis B virus; a member of the
  • Dezaguanine Dezaguanine Mesylate; Diaziquone; Docetaxel; Dolasatins; Doxorubicin;
  • Fluorouracil 5-FdUMP; Flurocitabine; Fosquidone; Fostriecin Sodium; Gemcitabine; Gemcitabine Hydrochloride; Gold Au 198; Homocamptothecin; Hydroxyurea; Idarubicin
  • Mitogillin Mitomalcin; Mitomycin; Mitosper; Mitotane; Mitoxantrone Hydrochloride; Mycophenolic Acid; Nocodazole; Nogalamycin;Ormaplatin; Oxisuran; Paclitaxel;
  • Testolactone Thiamiprine; Thioguanine; Thiotepa; Thymitaq; Tiazofurin; Tirapazamine; Tomudex; TOP53; Topotecan Hydrochloride; Toremifene Citrate; Trestolone Acetate; Triciribine Phosphate; Trimetrexate; Trimetrexate Glucuronate; Triptorelin; Tubulozole Hydrochloride; Uracil Mustard; Uredepa; Vapreotide; Verteporfin; Vinblastine;
  • cyclophosphamide nielphaian; chlorambucil; ifosfamide; busulfan; N-methyl- Nnitrosourea (MNU); N, N'-Bis (2-chloroethyl)-N-nitrosourea (BCNU); N- (2- chloroethyl)-N' cyclohexyl-N-nitrosourea (CCNU); N- (2-chloroethyl)-N - (trans-4- methylcyclohexyl-N-nitrosourea (MeCCNU); N- (2-chloroethyl)-N'- (diethyl) ethylphosphonate-N-nitrosourea (fotemustine); streptozotocin; diacarbazine (DTIC); mitozolomide; temozolomide; thiotepa; mitomycin C; AZQ; adozelesin; Cisplatin;
  • Dactinomycin (Actinomycin D); amsacrine; pyrazoloacridine; all-trans retinol; 14- hydroxy-retro-retinol; all-trans retinoic acid; N- (4- Hydroxyphenyl) retinamide; 13-cis retinoic acid; 3-Methyl TTNEB; 9-cis retinoic acid; fludarabine (2-F-ara-AMP); or 2- chlorodeoxyadenosine (2-Cda).
  • aclarubicin acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antagonists; altretamine; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; andrographolide; angiogenesis inhibitors;
  • antagonist D antagonist G
  • antarelix anti-dorsalizing morphogenetic protein- 1 ; antiandrogen, prostatic carcinoma; antiestrogen; antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; apurinic acid; ara-CDP-DL-PTBA; argininedeaminase; asulacrine;
  • camptothecin C calphostin C
  • camptothecin derivatives e.g., 10-hydroxy-camptothecin
  • IL-2 capecitabine
  • carboxamide-amino-triazole carboxyamidotriazole
  • CaRest M3 CARN 700; cartilage derived inhibitor; carzelesin; casein kinase inhibitors (ICOS); castanospermine; cecropin B; cetrorelix; chlorins; chloroquinoxaline sulfonamide; cicaprost; cis-porphyrin; cladribine; clomifene analogues; clotrimazole; collismycin A
  • collismycin B combretastatin A4; combretastatin analogue; conagenin;
  • crambescidin 816 crisnatol; cryptophycin 8; cryptophycin A derivatives; curacin A; cyclopentanihraquinones; cycloplatam; cypemycin; cytarabine ocfosfate; cytolytic factor; cytostatin; dacliximab; decitabine; dehydrodidemnin B; 2'deoxycoformycin
  • didox diethylnorsperrnine; dihydro-5-azacytidine; dihydrotaxol, 9- ; dioxamycin; diphenyl spiromustine; discodermolide; docosanol; dolasetron; doxifluridine;
  • ganirelix gelatinase inhibitors; gemcitabine; glutathione inhibitors; hepsulfam;
  • heregulin hexamethylene bisacetamide; homoharringtonine (HHT); hypericin;
  • ibandronic acid idarubicin; idoxifene; idramantone; ilmofosine; ilomastat;
  • imidazoacridones imiquimod; immunostimulant peptides; insulin-like growth factor- 1 receptor inhibitor; interferon agonists; interferons; interleukins; iobenguane;
  • iododoxorubicin ipomeanol, 4- ; irinotecan; iroplact; irsogladine; isobengazole; isohomohalicondrin B; itasetron; jasplakinolide; kahalalide F; lamellarin-N triacetate; lanreotide; leinamycin; lenograstim; lentinan sulfate; leptolstatin: letrozole; leukemia inhibiting factor; leukocyte alpha interferon; leuprolide + estrogen + progesterone; leuprorelin; levamisole; liarozole; linear polyamine analogue; lipophilic disaccharide peptide; lipophilic platinum compounds; lissoclinamide 7; lobaplatin; lombricine; lometrexol; lonidamine; losoxantrone; lovastatin; loxoribine;
  • masoprocol maspin
  • matrilysin inhibitors maspin
  • matrix metalloproteinase inhibitors masoprocol inhibitors
  • mycobacterial cell wall extract myriaporone; N-acetyldinaline; N-substituted benzamides; nafarelin; nagrestip; naloxone + pentazocine; napavin; naphterpin; nartograstim; nedaplatin; nemorubicin; neridronic acid; neutral endopeptidase;
  • nilutamide nisamycin; nitric oxide modulators; nitroxide antioxidant; nitrullyn; 06- benzyl guanine; octreotide; okicenone; oligonucleotides; onapristone; ondansetron; ondansetron; oracin; oral cytokine inducer; ormaplatin; osaterone; oxaliplatin;
  • palmitoylrhizoxin pamidronic acid; panaxytriol; panomifene; parabactin;
  • pazelliptine pazelliptine; pegaspargase; peldesine; pentosan polysulfate sodium; pentostatin; pentrozole; perflubron; perfosfamide; perillyl alcohol; phenazinomycin;
  • phenylacetate phosphatase inhibitors
  • picibanil pilocarpine hydrochloride
  • podophyllotoxin porfimer sodium; porfiromycin; propyl bis-acridone: prostaglandin J2; proteasome inhibitors; protein A-based immune modulator; protein kinase C inhibitor; protein kinase C inhibitors, microalgal; protein tyrosine phosphatase inhibitors; purine nucleoside phosphorylase inhibitors; purpurins; pyrazoloacridine; pyridoxylated hemoglobin polyoxyethylene conjugate; raf antagonists; raltitrexed; ramosetron; ras farnesyl protein transferase inhibitors; ras inhibitors; ras-GAP inhibitor; retelliptine demethylated; rhenium Re 186 etidronate; rhizoxin; ribozymes; RII retinamide; rogletimide; rohitukine; romurtide; roquinimex;
  • glycosaminoglycans tallimustine; tamoxifen methiodide; tauromustine; tazarotene; tecogalan sodium; tegafur; tellurapyrylium; telomerase inhibitors; temoporfin;
  • thalidomide thiocoraline; thrombopoietin; thrombopoietin mimetic; thymalfasin; thymopoietin receptor agonist; thymotrinan; thyroid stimulating hormone; tin ethyl etiopurpurin; tirapazamine; titanocene dichloride; topotecan; topsentin; toremifene; totipotent stem cell factor; translation inhibitors; tretinoin; triacetyluridine; triciribine; trimetrexate; triptorelin; tropisetron; turosteride; tyrosine kinase inhibitors;
  • tyrphostins UBC inhibitors; ubenimex; urogenital sinus-derived growth inhibitory factor; urokinase receptor antagonists; vapreotide; variolin B; vector system, erythrocyte gene therapy; velaresol; veramine; verdins; verteporfin; vinorelbine; vinxaltine; vitaxin; vorozole; zanoterone; zeniplatin; zilascorb; and zinostatin stimalamer.
  • the X moiety may also be a lytic peptide.
  • Such lytic peptides induce cell death and include, but are not limited to, streptolysin O; stoichactis toxin; phallolysin; staphylococcus alpha toxin; holothurin A; digitonin; melittin; lysolecithin;
  • the X moiety may also be a synthetic peptide that shares some sequence homology or chemical characteristics with any of the naturally occurring peptide lysins; such characteristics include, but are not limited to, linearity, positive charge, amphipathicity, and formation of alpha-helical structures in a hydrophobic environment (Leuschner et al, Biology of Reproduction 73:860-865, 2005).
  • Agents of the invention can also be coupled to an agent that induces complement-mediated cell lysis such as, for example, the immunoglobulin F c subunit.
  • Iodopyracet 125 I Iodopyracet 131 I; lofetamine Hydrochloride 12 I; Iomethin 125 I;
  • daunorubicine chlorhydrate doxorubicine chlorhydrate, epirubicine chlorhydrate, idarubicine chlorhydrate, pirarubicine, or zorubicine chlorhydrate; a camptothecin, or its derivatives or related compounds, such as 10, 11 methylenedioxycamptothecin; or a member of the maytansinoid family of compounds, which includes a variety of structurally-related compounds, e.g., ansamitocin P3, maytansine, 2'-N- demethylmaytanbutine, and maytanbicyclinol.
  • ansamitocin P3 maytansine, 2'-N- demethylmaytanbutine, and maytanbicyclinol.
  • Detectable labels can be used as the X moiety to prepare conjugate compounds of the invention for use as diagnostic agents.
  • a detectable label is used as the "cargo" of the compounds of the invention.
  • Detectable labels can be selected from a radioactive, bioluminescent, fluorescent, or heavy metal label, or an epitope tag.
  • Detectable labels of the conjugate compounds can include radioactive metals for use in radiographic imaging or radiotherapy.
  • Preferred radioisotopes also include 99m Tc, 5 I Cr, 67 Ga, 68 Ga, m In, 168 Yb, 14C La, 90 Y, 88 Y, 153 Sm, 156 Ho, 165 Dy, 64 Cu, 97 Ru, 103 Ru, 186 Re, 188 Re, 203 Pb, 2n Bi, 212 Bi, 2I3 Bi, and 214 Bi.
  • the choice of metal is determined based on the desired therapeutic or diagnostic application.
  • the metal complexes of the invention are useful as diagnostic and/or
  • a detectable label may be a metal ion from heavy elements or rare earth ions, such as Gd , Fe , Mn , or Cr .
  • Conjugates that include paramagnetic or superparamagnetic metals are useful as diagnostic agents in MRI imaging
  • Paramagnetic metals that may be used in the conjugates include, but are not limited to, chromium (III), manganese (II), iron (II), iron (III), cobalt (II), nickel
  • Fluorescent molecules that can also serve as detectable labels include green fluorescent protein (GFP), enhanced GFP (eGFP), yellow fluorescent protein (YFP), cyan fluorescent protein (CFP), red fluorescent protein (RFP), and dsRed.
  • GFP green fluorescent protein
  • eGFP enhanced GFP
  • YFP yellow fluorescent protein
  • CFP cyan fluorescent protein
  • RFP red fluorescent protein
  • dsRed red fluorescent protein
  • the bioluminescent molecule is luciferase.
  • the epitope tag is c- myc, hemagglutinin, or a histidine tag.
  • the lysine rich motif can be selected from the transmembrane helix 1 of DT, the Tl motif, or an amino acid sequence having at least 80% or more (e.g., 85%, 90%, 95%, 97%, 99%, or 100%) sequence identity to a contiguous amino acid sequence corresponding to at least amino acids 201 to 222 of DT (e.g., the lysine rich region has substantial sequence identity (80% or more, as described above) to a contiguous amino acid sequence corresponding to at least amino acids 201-235, amino acids 195 to 222, amino acids 195 to 235, amino acids 201 to 300, amino acids 195 to 300, amino acids 201 to 389, or amino acids 195 to 389, of DT).
  • the lysine rich motif has substantial sequence identity (80% or more, as described above) to a contiguous amino acid sequence corresponding to at least amino acids 201-235, amino acids 195 to 222, amino acids 195 to 235, amino acids 201 to 300, amino
  • amino acid sequences for use as the Y moiety can be selected from the transmembrane domains of other toxins (see, e.g., Table 1). These sequences can be modified at their amino-terminal end to include a cysteine residue in order to establish the formation of a disulfide bond between the Y/Z moieties and the X moiety of the conjugate compound.
  • polypeptides for use as the Y moiety include the consensus peptide sequence of CTF-binding moiety: RDKTKTKIESLKEHGPIKNS, the consensus peptide sequence of CTF-binding moiety including KXKXX sequences in bold:
  • polypeptides for use as the Y moiety are described in, e.g., US 2008- 0306003, incorporated by reference herein in its entirety, and below.
  • the compounds of the invention can be targeted to a specific cell or cells by using a targeting moiety (Z) that directs the compounds to a desired target cell.
  • Z targeting moiety is selected based on its ability to target conjugate compounds of the invention to a desired or selected cell population that expresses the corresponding binding partner (e.g., either the corresponding receptor or ligand) for the selected Z targeting moiety.
  • a conjugate compound of the invention could be targeted to cells expressing epidermal growth factor receptor (EGFR) by selected epidermal growth factor (EGF) as the Z targeting moiety.
  • EGFR epidermal growth factor receptor
  • EGF epidermal growth factor
  • the targeting moiety, Z is erythroblastic leukemia viral oncogene homolog (ErbB) receptor (e.g., ErbBl receptor; ErbB2 receptor; ErbB3 receptor; and ErbB4 receptor).
  • ErbB erythroblastic leukemia viral oncogene homolog
  • the Z targeting moiety can also be selected from bombesin, gastrin, gastrin- releasing peptide, tumor growth factors (TGF), such as TGF-a and TGF- ⁇ , and vaccinia virus growth factor (VVGF).
  • TGF tumor growth factors
  • VVGF vaccinia virus growth factor
  • Non-peptidyl ligands can also be used as the Z targeting moiety and may include, for example, steroids, carbohydrates, vitamins, and lectins.
  • the Z targeting moiety may also be selected from a peptide, such as somatostatin (e.g., a somatostatin having the core sequence cyclo[Cys-Phe-D-Trp-Lys-Thr-Cys], and in which, preferably, the C-terminus of the somatostatin analog is: Thr-NFL), a somatostatin analog (e.g., octreotide and lanreotide), bombesin, a bombesin analog, or an antibody, such as a monoclonal antibody.
  • somatostatin e.g., a somatostatin having the core sequence cyclo[Cys-Phe-D-Trp-Lys-Thr-Cys]
  • a somatostatin analog e.g., octreotide and lanreotide
  • bombesin e.g., octreotide and lan
  • peptides for use as the Z targeting moiety in the conjugate compounds of the invention can be selected from iSS peptides and analogs, urotensin II peptides and analogs, GnRH I and II peptides and analogs, octreotide, depreotide, vapreotide, vasoactive intestinal peptide (VIP), cholecystokinin (CCK), RGD-containing
  • ITIPP(psi) annexin-V
  • endothelin endothelin
  • leukotriene B4 LLB4
  • chemotactic peptides e.g., N-formyl-methionyl-leucyl-phenylalanine-lysine (fMLFK)
  • GP Iib/IIIa e.g., N-formyl-methionyl-leucyl-phenylalanine-lysine (fMLFK)
  • Immunoreactive ligands for use as the targeting moiety Z in the invention include an antigen-recognizing immunoglobulin (also referred to as "antibody"), or antigen-recognizing fragment thereof.
  • immunoglobulin refers to any recognized class or subclass of immunoglobulins such as IgG, IgA, IgM, IgD, or IgE. Preferred are those immunoglobulins which fall within the IgG class of immunoglobulins.
  • the immunoglobulin can be derived from any species. Preferably, however, the immunoglobulin is of human, murine, or rabbit origin. In addition, the immunoglobulin may be polyclonal or monoclonal, but is preferably monoclonal.
  • the immunoglobulin used in conjugates of the invention may be a "chimeric antibody” as that term is recognized in the art.
  • the immunoglobulin may be a "bifunctional” or “hybrid” antibody, that is, an antibody which may have one arm having a specificity for one antigenic site, such as a tumor associated antigen while the other arm recognizes a different target, for example, a hapten which is, or to which is bound, an agent lethal to the antigen-bearing tumor cell.
  • the bifunctional antibody may be one in which each arm has specificity for a different epitope of a tumor associated antigen of the cell to be therapeutically or biologically modified.
  • Hybrid antibodies thus have a dual specificity, preferably with one or more binding sites specific for the hapten of choice or one or more binding sites specific for a target antigen, for example, an antigen associated with a tumor, an infectious organism, or other disease state.
  • Biological bifunctional antibodies can also be used as the Z targeting moiety in the conjugate compounds of the invention (such antibodies are described in, for example, European Patent Publication, EPA 0 105 360, which is hereby incorporated by reference in its entirety).
  • Such hybrid or bifunctional antibodies may be derived either biologically, by cell fusion techniques, or chemically, such as with cross- linking agents or disulfide bridge-forming reagents, and may be comprised of whole antibodies and/or fragments thereof.
  • bifunctional antibodies are those biologically prepared from a "polydoma” or “quadroma” or which are synthetically prepared with cross-linking agents such as bis- (maleimido)-methyl ether (“BMME”), or with other cross-linking agents familiar to those skilled in the art.
  • BMME bis- (maleimido)-methyl ether
  • the immunoglobin may be a single chain antibody ("SCA").
  • SCA may consist of single chain Fv fragments ("scFv”) in which the variable light (“V L ”) and variable heavy (“V H ”) domains are linked by a peptide bridge or by disulfide bonds.
  • the immunoglobulin may consist of single VH domains (dAbs) that possess antigen-binding activity. See G. Winter and C. Milstein, Nature 349:295, 1991; R. Glockshuber et al., Biochemistry 29:1362, 1990; and, E. S. Ward et al., Nature 341 :544, 1989.
  • chimeric monoclonal antibodies preferably those chimeric antibodies having specificity toward a tumor associated antigen.
  • the term "chimeric antibody” refers to a monoclonal antibody comprising a variable region, i.e. a binding region, from one source or species and at least a portion of a constant region derived from a different source or species, usually prepared by recombinant DNA techniques.
  • Chimeric antibodies having a murine variable region and a human constant region are especially preferred in certain applications of the invention, particularly human therapy, because such antibodies are readily prepared and may be less immunogenic than purely murine monoclonal antibodies.
  • Such murine/human chimeric antibodies are the product of expressed immunoglobulin genes comprising DNA segments encoding murine immunoglobulin variable regions and DNA segments encoding human immunoglobulin constant regions.
  • Other forms of chimeric antibodies for use in conjugates of the invention are those in which the class or subclass has been modified or changed from that of the original antibody.
  • Such "chimeric" antibodies are also referred to as "class-switched antibodies.”
  • Methods for producing chimeric antibodies involve conventional recombinant DNA and gene transfection techniques now well known in the art. See Morrison, S. L, et al., Proc. Nat'l Acad. Sci., 81 :6851, 1984.
  • chimeric antibody also includes a "humanized antibody,” namely, those antibodies in which the framework or “complementarity determining regions” (“CDR") have been modified to include the CDR of an immunoglobulin of different specificity, as compared to that of the parent immunoglobulin.
  • CDR framework or complementarity determining regions
  • a murine CDR is grafted into the framework region of a human antibody to prepare the "humanized antibody.” See, e.g., L. Riechmann et al., Nature 332:323, 1988; M. S. Neuberger et al, Nature 314:268, 1985.
  • Particularly preferred CDRs correspond to those representing sequences recognizing the antigens noted above for the chimeric and bifunctional antibodies. See, e.g., EPA 0 239 400 (published Sep. 30, 1987), which is hereby incorporated by reference in its entirety.
  • the noted constructions can be prepared with immunoglobulin fragments used as the starting materials; or, if recombinant techniques are used, the DNA sequences, themselves, can be tailored to encode the desired "fragment” which, when expressed, can be combined in vivo or in vitro, by chemical or biological means, to prepare the final desired intact immunoglobulin "fragment.” It is in this context that the term "fragment" is used herein.
  • the immunoglobulin (antibody), or fragment thereof, used as the Z targeting moiety in conjugate compounds of the present invention may be polyclonal or monoclonal in nature. Monoclonal antibodies are the preferred immunoglobulins.
  • the preparation of polyclonal or monoclonal antibodies is well known to those skilled in the art. See, e.g., G. Kohler and C. Milstein, Nature 256:495, 1975.
  • hybridomas and/or monoclonal antibodies which are produced by such hybridomas and which are useful in the practice of the present invention are publicly available. Linkers for Attaching the Cytotoxic, Therapeutic, or Diagnostic Agents
  • conjugate compounds of the invention can include cytotoxic, therapeutic, or diagnostic agents as "cargo” (e.g., nucleic acids molecules, PNAs, peptides, polypeptides, proteins, small molecules, antibodies, or antibody fragments). These agents can be associated with or bonded to the Y and Z moieties of the conjugate compound, which faciliate the delivery and targeting of the conjugate compound, respectfully, using, e.g., a linker or linking component.
  • cytotoxic, therapeutic, or diagnostic agents as "cargo” (e.g., nucleic acids molecules, PNAs, peptides, polypeptides, proteins, small molecules, antibodies, or antibody fragments).
  • conjugate compounds of the invention are prepared by incorporating a peptidic linking group into the the X moiety (e.g., nucleic acid molecules, such as siRNA and dsRNA), PNA molecules, peptides, polypeptides, proteins, small molecules, antibodies, or antibody fragments).
  • the peptide linking group can include, e.g., a cysteine residue that is used to form a disulfide bond between the X moiety and the Y moiety or it can be another peptide sequence that forms a bond that is cleavable by a known cytosolic enzyme.
  • linker can also couple the X moiety (i.e., the "cargo") to the Y moiety by reacting, e.g., a free amino group of a Thr residue of a peptide portion of X moiety to the conjugate compound (e.g., a portion of the Y moiety) with an appropriate functional group of a chelator, such as a carboxyl group or activated ester.
  • a conjugate may
  • EDTA ethylenediaminetetraacetic acid
  • Conjugate compounds of the invention may be administed to a mammalian subject, such as a human, directly or in combination with any pharmaceutically acceptable carrier or salt known in the art for use in the treatment or detection of disease.
  • Pharmaceutically acceptable salts may include non-toxic acid addition salts or metal complexes that are commonly used in the pharmaceutical industry.
  • acid addition salts include organic acids such as acetic, lactic, pamoic, maleic, citric, malic, ascorbic, succinic, benzoic, palmitic, suberic, salicylic, tartaric, methanesulfonic, toluenesulfonic, or trifluoroacetic acids or the like; polymeric acids such as tannic acid, carboxymethyl cellulose, or the like; and inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid phosphoric acid, or the like.
  • Metal complexes include zinc, iron, and the like.
  • One exemplary pharmaceutically acceptable carrier is physiological saline.
  • physiologically acceptable carriers and their formulations are known to one skilled in the art and described, for example, in Remington's Pharmaceutical Sciences. (18 th edition), ed. A. Gennaro, 1990, Mack Publishing Company, Easton, PA.
  • compositions of a therapeutically effective amount of a conjugate compound of the invention, or pharmaceutically acceptable salt-thereof can be administered orally, parenterally (e.g., by intramuscular, intraperitoneal, intravenous, or subcutaneous injection, by inhalation, intradermally, using optical drops, or by implant), nasally, vaginally, rectally, sublingually, or topically, in admixture with a pharmaceutically acceptable carrier adapted for the route of administration.
  • parenterally e.g., by intramuscular, intraperitoneal, intravenous, or subcutaneous injection, by inhalation, intradermally, using optical drops, or by implant
  • nasally, vaginally, rectally, sublingually, or topically in admixture with a pharmaceutically acceptable carrier adapted for the route of administration.
  • compositions intended for oral use may be prepared in solid or liquid forms according to any method known to the art for the manufacture of pharmaceutical compositions.
  • the compositions may optionally contain sweetening, flavoring, coloring, perfuming, and/or preserving agents in order to provide a more palatable preparation.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is admixed with at least one inert pharmaceutically acceptable carrier or excipient.
  • inert pharmaceutically acceptable carrier or excipient may include, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, sucrose, starch, calcium phosphate, sodium phosphate, or kaolin. Binding agents, buffering agents, and/or lubricating agents (e.g., magnesium stearate) may also be used. Tablets and pills can additionally be prepared with enteric coatings.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and soft gelatin capsules. These forms contain inert diluents commonly used in the art, such as water or an oil medium. Besides such inert diluents, compositions can also include adjuvants, such as wetting agents, emulsifying agents, and suspending agents.
  • Liquid formulations can be sterilized by, for example, filtration through a bacteria-retaining filter, by incorporating sterilizing agents into the compositions, or by irradiating or heating the compositions. Alternatively, they can also be manufactured in the form of sterile, solid compositions which can be dissolved in sterile water or some other sterile injectable medium immediately before use.
  • compositions for rectal or vaginal administration are preferably suppositories which may contain, in addition to active substances, excipients such as coca butter or a suppository wax.
  • Compositions for nasal or sublingual administration are also prepared with standard excipients known in the art.
  • Formulations for inhalation may contain excipients, for example, lactose, or may be aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycocholate and deoxycholate, or may be oily solutions for administration in the form of nasal drops or spray, or as a gel.
  • the amount of active ingredient in the compositions of the invention can be varied.
  • One skilled in the art will appreciate that the exact individual dosages may be adjusted somewhat depending upon a variety of factors, including the type of conjugate compound being administered, the time of administration, the route of administration, the nature of the formulation, the rate of excretion, the nature of the subject's conditions, and the age, weight, health, and gender of the patient.
  • the severity of the condition targeted by the conjugate compounds of the invention may also have an impact on the dosage level.
  • dosage levels of between 0.1 ⁇ g/kg to 100 mg/kg of body weight are administered daily as a single dose or divided into multiple doses.
  • the general dosage range is between 250 ⁇ g/kg to 5.0 mg/kg of body weight per day.
  • compositions of the invention may be as frequent as necessary to obtain the desired therapeutic effect. Some patients may respond rapidly to a higher or lower dose and may find much weaker maintenance doses adequate. Other patients, however, receive long-term treatments at the rate of 1 to 4 doses per day, in accordance with the physiological requirements of each patient.
  • the active product may be administered, e.g., intravenously, 1 to 4 times daily or via continuous infusion.
  • sustained released composition will generally be preferred.
  • Toxins as a Platform to Deliver Cytotoxic, Therapeutic, Diagnostic Agents as Cargo
  • Toxins e.g., the transmembrane and receptor binding domains of the DT
  • cargo e.g., nucleic acid molecules, such as siRNA, polypeptides, or chemical compounds
  • the "cargo" affixed to the toxins can be easily substituted to yield multiple different delivery constructs.
  • the toxins can be engineered to include a cysteine residue at its amino-terminal end or an existing cysteine residue within the toxin (e.g., the cysteine residue located at amino acid position 201 of DT (see, e.g., Figure 1 A) can be used to "load” the cargo onto the toxin construct via the formation of disulfide bonds between the cargo (referred to herein as the "X" moiety) and the toxin platform (referred to herein as the
  • diphtheria toxin structural platform as a nano-machine in the
  • the evolution of bacterial protein toxins can be viewed as the natural assembly of highly efficient nano-machines capable of cell surface receptor specific delivery of their respective catalytic domains, "cargo", to the cytosol.
  • the DT structural platform can be exploited to develop a system for the facilitated delivery of, e.g., nucleic acid molecules (e.g., siRNA), polypeptide therapeutics, or small molecules across the endosomal vesicle membrane and their subsequent release into the cytosol of target cells and tissues.
  • the conjugate compounds of the invention deliver their "cargo” using the same mechanism by which native DT delivers its C- domain cargo to the eukaryotic cell cytosol.
  • the intoxication of sensitive eukaryotic cells by DT follows an ordered series of events.
  • the first step in the intoxication process is the binding of toxin to its cell surface receptor, the heparin-binding (hb)-EGF-like precursor (Naglich el ah, 1992).
  • This binding is further enhanced by the DT receptor associated protein DTRAP27, which is the primate homologue of human CD9 (Iwamoto et al., 1994).
  • Receptor bound toxin is concentrated in clathrin coated pits and internalized into clathrin coated vesicles (CCVs), which are then converted into early endosomal vesicles (EEVs) (Moya et al., 1985).
  • CCVs clathrin coated vesicles
  • EEVs early endosomal vesicles
  • the activity of the vacuolar ATPase lowers the luminal pH within the EEV. Acidification of the vesicle lumen is required to trigger the unfolding of the DT transmembrane domain (Boquet et al, 1976).
  • the transmembrane domain undergoes a dynamic reorganization which results in the spontaneous insertion of two a-helical hairpins into the vesicle membrane resulting in the formation a transmembrane 18-25 A pore (Donovan et ah, 1981; Kagan et al., 1981).
  • Pore formation is a critical step in the intoxication process since it provides the conduit for C-domain translocation from the cis to trans side of the endosomal vesicle membrane with subsequent events effecting C-domain release into the target cell cytosol.
  • CTF Cytosolic Translocation Factor
  • the DT C-domain is an ADP-ribosyltransferase that catalyzes the NAD+ -dependent ADP-ribosylation of eukaryotic elongation factor 2 (EF-2) thereby inhibiting cellular protein synthesis (Honjo et ah, 1968; Collier and Kandel, 1971 ; Gill and Pappenheimer, 1971).
  • EF-2 eukaryotic elongation factor 2
  • Tl motif is highly conserved in anthrax LF, anthrax EF, and the botulinum neurotoxins. I have also demonstrated that the Tl motif and its closely associated XKXX sequences mediate the facilitated delivery of anthrax LF into the eukaryotic cell cytosol (Tamayo et al., 2008).
  • the Tl motif and its closely positioned KXKXX sequences closely resemble the dilysine (KKXX) and interrupted dilysine motifs (KXKXX) in the cytoplasmic tails of both p23/24 adaptor and cargo proteins (Cosson & Letourneue, 1994) that bind COPI coatomer complex proteins and direct vesicle trafficking (Fiedler et al., 1966).
  • endosomal vesicle membrane to form a pore through which the "cargo " is threaded by the chaperone-like properties of the transmembrane domain: and, upon the emergence of transmembrane helix 1 sequences on the cytosolic surface of the endosomal vesicle membrane,
  • transmembrane domain Tl motif and/or KXKXX sequences e.g., dibasic signature (e.g., KKXX, KXKXX) and/or an aromatic amino acid sequences (e.g. FFXXBB(X) horr) that function as p23/24 adaptor mimetics and mediate the binding of COPI complex proteins then facilitate the delivery of the catalytic domain "cargo " into the cytosol; the cargo, which is bound to the transmembrane domain and receptor binding domain via a disulfide bond is then reduced by thioredoxin reductase, thereby releasing the cargo into the cytosol.
  • KXKXX sequences e.g., dibasic signature (e.g., KKXX, KXKXX) and/or an aromatic amino acid sequences (e.g. FFXXBB(X) press) that function as p23/24 adaptor mimetics and mediate the binding of COPI complex proteins then facilitate
  • “nicked" toxin may be separated into a 21.1 kDa N-terminal polypeptide (residues 1-193), or Fragment A, and a 41.2 kDa C-terminal Fragment B (residues 194 to 535), which carries both the transmembrane and receptor binding domains (Uchida et al, 1971; Choe et al, 1992; Bennett et al, 1994).
  • DAB 389 lL-2 was the first, and to date only, targeted toxin to be approved by the FDA for human clinical use (Foss, 2000).
  • the DT structural platform is remarkably amenable to receptor binding domain substitution, thereby allowing this domain of DT to be substituted with a range of desired binding domain (e.g., a receptor, the receptor ligand, and other binding molecules, such as antibodies or antibody fragments, that can be used to target the conjugate compound to a desired cell or tissue).
  • desired binding domain e.g., a receptor, the receptor ligand, and other binding molecules, such as antibodies or antibody fragments, that can be used to target the conjugate compound to a desired cell or tissue.
  • transmembrane domain that are required for COPI complex facilitated delivery of "cargo" from the lumen of acidified endosomal vesicles to the cytosol allows for the use of the DT structural platform for the development of a system to efficiently deliver cytotoxic, therapeutic, or diagnostic agents (e.g., nucleic acid molecules, such as siRNA and PNA molecules, polypeptides, such as growth and transcription factors, cytotoxic agents, such as paclitaxel, and anti-viral agents) to the cytosol of target cells.
  • cytotoxic, therapeutic, or diagnostic agents e.g., nucleic acid molecules, such as siRNA and PNA molecules, polypeptides, such as growth and transcription factors, cytotoxic agents, such as paclitaxel, and anti-viral agents
  • This system envisions the use of the DT B-fragment transmembrane and receptor binding domains (and substitutions thereof) for (i) the targeting of specific cell surface receptors or ligands for cell-specific and/or tissue specific delivery, and (ii) the transmembrane domain for endosomal vesicle membrane pore formation and COPI complex binding for the facilitated delivery of siRNA "cargo" to the target cell cytosol. Delivery of siRNA molecules using Conjugate Compounds of the Invention
  • the antisense RISC complex Upon degradation of the sense strand, the antisense RISC complex then seeks out and degrades the messenger (m)RNA that is complementary to the antisense strand (Ameres et al., 2007). Since the antisense siRNA-RISC complex is stable, mRNA that is targeted continues to be degraded over time. In fact, siRNA mediated knockdown of mRNA is known to last for 3-7 days in rapidly dividing cells, and for weeks in non-dividing cells (Bartlett & Davis, 2006).
  • siRNAs have been developed for hepatitis B virus (Morrissey et al., 2005); human papilloma virus (Niu et al., 2006), liver cirrhosis (Sato et al., 2008), ovarian cancer (Haider et al., 2006), and bone cancer (Takeshita et al., 2005); the siRNA molecules described in the publications and others can be incorporated into the conjugate compounds of the invention to treat these and other diseases.
  • cytoplasmic delivery of siRNAs in vitro have been shown to require one of a variety of transfection methods: linkage to cell penetrating peptides (Turner et al., 2007; Deshayes et al., 2008; Lebleu et al., 2008); conjugation to cholesterol
  • RNAi is a cytoplasmic process
  • efficient delivery to the cytosol following either adsorption to the cell surface or from the lumen of an endosomal vesicle after fluid phase endocytosis is absolutely essential.
  • a diphtheria toxin-based system can be used to facilitate cytosolic delivery of oligonucleotide antisense cargo.
  • peptide-PNA can be conjugated with Fragment B from the non-toxic mutant of diphtheria toxin, CRM 197.
  • the approach is based upon methods that were developed for the reconstitution of "native" diphtheria toxin from two non-toxic mutants, CRM 197 and CRM45 (Uchida et al., 1973).
  • Fragment B197 can be purified from CRM197 according to the method of Uchida et al. (1973). Briefly, purified CRM197 (List Biological Laboratories, Campbell, CA) is treated with immobilized trypsin for 10 min at 37°C in the presence of 10 mM dithiothreitol in 0.02M Tris-HCl buffer at pH 8.0. The reaction is stopped by centrifuging the reaction mix through a spin column. This method allows the separation of the cleaved ppolypeptides from the immobilized trypsin which does not pass through the column frit. Under denaturing conditions the trypsin treated CRM 197 is then be purified by HPLC sizing in order to separate fragment A197 from B197.
  • CHO- Kl cells express the hb-EGF-like precursor receptor for diphtheria toxin, the peptide- PNA-Fragment B197 conjugate should readily bind to the cell surface and be internalized into an early endosomal compartment.
  • peptide-PNA alone should not, by itself, be delivered and therefore should not give rise to luciferase expression
  • CRM 197 can be used as a competitive inhibitor of peptide-PNA-Fragment B197 binding to the hb-EGF-like precursor receptor
  • Bafilomycin Al can be used as an inhibitor of the vesicular ATPase in order to establish that vesicle acidification (required for Fragment B transmembrane domain insertion and pore formation in the endosomal membrane) is required for delivery of the peptide-PNA into the target cell cytosol.
  • Trizol extracted RNA can be used for RT-PCR to confirm the expression of full length luc mRNA.
  • RNA extracted from Luc-IVS2 CHO-Kl cells can be used as the positive control for RT-PCR.
  • primers which hybridize to sequences flanking Luc-IVS2 introns can be used: TTGATATGTGGATTTCGAGTCGTC and
  • Diphtheria fragment B be used to successfully deliver a "chemically unique" PNA cargo.
  • the DT platform described herein can be used to deliver to the cytosol of a target cell a chemically unique PNA cargo. It is known that N-terminal extension fusion proteins of fragment A which include a duplicate A fragment and some of apolipoprotein Al can be delivered into the eukaryotic cell cytosol (Madhus et al., 1992). Thus, other cargo, such as a peptide-PNA (see Fig.l 1) disulfide cross-linked to fragment B of diphtheria toxin, can be delivered into the eukaryotic cell cytosol.
  • a peptide-PNA see Fig.l 1
  • An alternative construct may be to cross link a PNA-Cys to a modified Fragment A (A197).
  • A197 modified Fragment A
  • a Cys residue can be inserted at the N-terminal end of fragment A 197 in order to form a construct that is analogous to those described by Madhus et al. (1992).
  • the recombinant protein can be expressed, purified and used to make the conjugate PNA-Cys-S-S-CRM197.
  • the sense or antisense strand can be modified with a 3'-thiopropyl moiety that can be used for peptide coupling (this same modification can be used to couple any of the nucleic acid molecules described herein as "cargo" to the Y moiety for formation of the conjugate compounds of the invention).
  • this same modification can be used to couple any of the nucleic acid molecules described herein as "cargo" to the Y moiety for formation of the conjugate compounds of the invention.
  • peptide-PNAs and peptide-siRNAs can be synthesized chemically using techniques known in the art.
  • Peptide-siRN A/Fragment B 197 mediated silencing of Luc gene expression in CHO-Kl cells: As described above, protective antigen PA63 mediated the effective delivery of peptide-PNA and correction of aberrantly spliced
  • GCCTGAAGTCTCTGATTAAGT-3 ' reverse 5'- ACACCTGCGTCGAAGT-3 '
  • mRNA isolated from siRNA treated and control samples and correlated to direct measurements of luciferase activity using the Promega luciferase assay system with RLU determined by Wallac Microbeta, Victor2 (Perkin Elmer) counter or Turner Designs luminometer.
  • the A-form of RNA can be thread through the pore formed by the fragment B197 transmembrane domain
  • transmembrane domain helices 5-9 of diphtheria toxin has been measured by Kagan et al. (1981) to be >18A and by Zalman & Wisnieski (1984) to be 24A in diameter. Based upon these reports the pore formed in the endosomal vesicle by the diphtheria toxin transmembrane domain is of sufficient size to allow the passage of peptide- siRNA and peptide PNA from the vesicle lumen to the cytosol.
  • the disulfide bond between the cargo (peptide-siRNA and peptide-PNA) and fragment B197 can be reduced in the endosomal vesicle lumen
  • native diphtheria toxin may be "nicked” by proteases either during its purification from culture filtrates of Corynebacterium diphtherias, in serum, or on the cell surface by the endoproteinase furin.
  • both native diphtheria toxin and the diphtheria toxin-related fusion protein toxins must be nicked in the protease sensitive loop between the A and B fragments into or to deliver the A fragment to the target cell cytosol.
  • the A and B fragment of nicked diphtheria toxin are known to remain disulfide bond cross-linked until the fragments emerge from the transmembrane pore and are presented on the cytosolic surface of the endosomal vesicle and into the reducing environment of the cytosol.
  • Ratts et al., (2003) demonstrated that the disulfide bond linking the A and B fragment of the toxin are reduced in the cytosol by thioredoxin reductase.
  • the peptide-siRNA fragment B197 conjugate and the peptide-PNA fragment B197 conjugate are expected to remain disulfide bond cross-linked until the peptide-siRNA is delivered into the cytosol.
  • a bacterial protein toxin can facilitate the cytosolic delivery of antisense oligon ucleotide.
  • anthrax protective antigen binds to its cell surface receptors capillary morphogenesis gene 2 (CMG2) and tumor endothelial cell marker 8 (TEM8) (Bradley et al., 2001 ; Scobie et al., 2003). Once bound to its receptor, PA83 is cleaved to PA63 by the endoprotease furin (Klimpel et al., 1992; Gordon et al., 1995) and then reorganizes into a homo-heptamer (Milne et al., 1994).
  • CMG2 capillary morphogenesis gene 2
  • TEM8 tumor endothelial cell marker 8
  • Each heptamer is then able to bind up to three molecules of Lethal Factor and/or Edema Factor.
  • Anthrax toxin is then internalized in clathrin coated pits which become early endosomal vesicles. Upon acidification of the endosomal lumen, PA63 undergoes a dynamic change and spontaneously inserts into the vesicle membrane forming a pore.
  • the mechanism of Lethal Factor entry into cytosol parallels that of diphtheria toxin and requires COPI complex binding to one or more of multiple KXKXX sequences and a Tl-like motif in the N-terminal region of the protein (Tamayo et al, 2008).
  • PA63 would facilitate both the delivery of PNA-Lys 8 through the PA63 pore and its release into the eukaryotic cell cytosol.
  • CHO-K1 Luc-VIS-654 cells were exposed to PNA-Lys 8 in the absence or presence of either PA83 or PA63 and measured cytosolic delivery by induction of functional luciferase activity by measuring both chcmiluminescence and luc mRNA.
  • Figure 1 A shows a partial amino acid sequence of diphtheria toxin fragment B from Serine] 95 to Asparagine 235 showing transmembrane helices 1 and 2, the Tl motif and the multiple lysine residues that were anticipated to be most likely to mediate binding to COPI complex proteins and thereby facilitate catalytic domain entry.
  • DAB 389 IL-2 is composed of native DT catalytic and transmembrane domain sequences, amino acids 1 -389, to which human interleukin 2 is genetically fused in the correct translational reading frame (Williams et ah, 1990).
  • transmembrane domain helix 1 a combination of at least three lysine residues is most likely to be required for binding to COPI complex proteins and subsequent delivery of the C-domain. Furthermore, the spacing between these residues appears to affect the efficiency of C-domain entry into the cytosol as reflected by the 2-log range in cytotoxic potency of this group of single K ⁇ A mutants. If this were the case, then either the introduction of any pair of K ⁇ A mutations or the quadruple K ⁇ A mutation in transmembrane helix 1 should lead to a complete loss of cytotoxic activity.
  • double mutants DAB(K213A, K215A) 389 IL-2 and DAB(K215A, K217A) 389 IL-2, and the quadruple mutant DAB(K213A, K215A, K217A, K222A) 389 IL-2 were constructed. Following expression and purification, each mutant fusion protein was then assayed for cytotoxic activity. As anticipated, both of the double K ⁇ A mutants as well as the quadruple K ⁇ A mutant were found to be non-toxic (IC 50 > 5 x 10 "7 M) (Figs.lC & ID).
  • the structural gene encoding DAB 389 IL2 was modified such that amino acids 212 - 223 which encompasses transmembrane helix 1 was deleted and replaced with the 13 amino acid sequence encoding the COPI binding portion of the p23 cytoplasmic tail (REILKKAKFFRRL). Following its genetic construction, the plasmid encoding the mutant toxin DAB(212p23) 3 8 9 IL-2 was cloned, sequenced to verify correct insertion and reading frame of the COPI binding segment, and the recombinant mutant protein was expressed and purified as described in Experimental procedures below. As shown in Figure 2, dose response analysis of
  • DAB(212p23) 389 IL-2 on Hutl02 cells is identical to that of the wild type DAB 389 IL-2 (IC50 3 ⁇ 4 10 pM).
  • the functional equivalence between the lysine-rich transmembrane helix 1 and the COPI binding segment from the cytoplasmic tail of the p23 adaptor protein suggests maintenance of the COPI complex binding function of this region is an essential feature in the catalytic domain entry process.
  • CBM 1,3- cyclohexanebis(methylamine)
  • Lemichez et al. (1997) were the first to describe an in vitro assay to investigate the requirements for translocation of the diphtheria toxin catalytic domain from the lumen of acidified endosomal vesicles to the external medium.
  • endosomal vesicles were pre-loaded with native diphtheria toxin in the presence of Bafilomycin Al, and the early endosomal vesicle enriched fraction was isolated by sucrose density gradient ultracentrifugation. Upon removal of
  • TKIESLKEHG transmembrane helix 1 of diphtheria toxin. Since the cytosolic expression of a peptide which carried the Tl motif in HuT102 cells was found to confer resistance to the toxin and knock down of peptide expression restored toxin sensitivity, it was apparent that either the Tl motif or transmembrane helix 1 of diphtheria toxin is likely to play an essential role in the catalytic domain entry process. In addition, pull down experiments using GST-DT 140-271 demonstrated that at least the ⁇ -COP subunit of the COPI complex specifically bound to at least a portion of the Tl motif, and that this association was also essential for catalytic domain entry process.
  • the Tl motif includes two lysine residues which may play a role in COPI binding and catalytic domain entry; however, in anthrax lethal factor and anthrax edema factor the separation of their respective Tl- like motifs from the multiple upstream KXKXX COPI binding sequences raises addition questions as to the role that the Tl motif per se may play in the entry process.
  • a site-directed mutational analysis of the multiple lysine residues in the N-terminal end of lethal factor was begun in order to determine the minimal sequence necessary for both COPI complex binding and delivery of
  • transmembrane helix 1 is dependent upon the presence and spacing of at least three of the four lysine residues, two of which are positioned immediately upstream of the consensus Tl motif ( TKTKIESLKEHG).
  • a site- directed mutational analysis of transmembrane helix 1 was first conducted in order to determine the minimal number of lysine residues that were necessary to facilitate catalytic domain delivery to the eukaryotic cell cytosol. Following site-directed mutagenesis and DNA sequence analysis to ensure the introduction of each mutation, individual mutant recombinant proteins were expressed, purified, and tested for cytotoxic activity by dose response analysis on HuT102 cells.
  • K215A With exception of a single mutation (K215A), all of the single K ⁇ A mutant forms of DAI1 ⁇ 2 ⁇ )IL-2 displayed only a modest 1 - 2-log reduction in their respective cytotoxic potency. In marked contrast, the introduction of double K ⁇ A mutations ⁇ e.g., K213A, K215A or K215A, K217A) resulted in over a 5-log reduction in cytotoxic potency in their respective mutant fusion protein toxins.
  • double K ⁇ A mutations ⁇ e.g., K213A, K215A or K215A, K217A
  • transmembrane helix 1 of diphtheria toxin and COPI components appears to be unlike the interactions mediated by the canonical di- lysine signature KKXX with a-COP and ⁇ '-COP (Eugster et ah, 2004; Letourneur et ah, 1994).
  • cargo and adaptor protein interactions with ⁇ -COP have been reported to exhibit more diversity in their binding profile including KKXX and KXKXX motifs (Eugster et al., 2004; Zerangue et al. 2001), and results presented here suggest this diversity of binding may be extended to transmenbrane helices 1 and 2 of diphtheria toxin as well.
  • the molecular process by which the catalytic domain from DAB 38 9iL-2 is delivered to the eukaryotic cell cytosol appears to follow the following steps: (i) binding of the toxin to its respective cell surface receptor (Naglich et al, 1992), (ii) the internalization of the toxin: :receptor complex by receptor mediated endocytosis into an early endosomal compartment (Moya et al., 1985), (iii) upon acidification of the vesicle lumen by the (v)ATPase, the transmembrane domain spontaneously inserts into the endosomal vesicle membrane forming an 18 - 22A pore or channel (Donovan et al, 1981 ; Kagan et al., 1981 ).
  • transmembrane helices 1-4 of the toxin with its disulfide bond linked catalytic domain appear to be un-tethered and readily “pulled” through the transmembrane vesicle pore formed by helices 5 - 9.
  • Bovine liver COPI purification COPI enriched fractions were prepared from bovine liver cytosol as described by Waters et al. (1991) with some modifications described by Tamayo et al. (2008).
  • the 13S fraction containing intact COPI was further purified by DE52 (Whatman) column following manufacture's specifications in a Biologic LP (Bio-rad). Briefly, the DE52 cellulose was equilibrated with 25 mM Tris-HCl (pH 7.4)/ 100 mM KCl, 1 mM DTT, 10% Glycerol.
  • the column was eluted with a step gradient of 150, 500, 750 and 1000 mM KCl in 25 mM Tris-HCl (pH 7.4)/ 10% Glycerol/ 1 mM DTT.
  • the elution corresponding to 500 mM KCl containing intact COPI complex and associated material was dialyzed against 25 mM Tris.HCl (pH 7.4), 10% Glycerol and used as input material for the precipitation assays.
  • COPI subunits In Vitro Synthesis of COPI subunits.
  • Vector pCMV6-XL5 carrying the structural human gene encoding ⁇ ' COP (NM_004766.1), ⁇ -COP (NM 016128.3) and ⁇ -COP (NM_007263.3) were purchased from OriGene Technologies (Rockville, MD).
  • Full length COPI subunits were synthesized in vitro by using the TNT Quick Coupled Transcription/Translation System (Promega) in the presence of [ 35 S] -methionine following the manufacturer's instructions (2 ⁇ g of plasmid DNA/ 100 ⁇ of reaction volume).
  • reaction mixture was diluted to 300 ⁇ with binding buffer (50 mM Tris HC1, pH 7.4/150 mM NaCl/1 mM EDTA/1% Nonidet P- 40/ IX protease inhibitor cocktail (Roche)). Pull-down experiments with GST and GST-DT140-271 were performed as described above. Elutions were then analyzed by SDS/PAGE and autoradiographed according to standard methods.
  • binding buffer 50 mM Tris HC1, pH 7.4/150 mM NaCl/1 mM EDTA/1% Nonidet P- 40/ IX protease inhibitor cocktail (Roche)
  • Bacterial strains, plasmids and fusion toxin products Bacterial strains, plasmids and fusion toxin products.
  • the parental plasmid pET- JV127 (vanderSpek et al., 1993) encoding for the fusion toxin DAB 389 IL-2 (AAA72359) was used for cloning and purification of the mutant toxins.
  • the introduction of the alanine exchange mutations and the p23 adaptor COPI binding sequence swap was performed by site-directed mutagenesis between the Nsil and Rsill restriction sites (Table 4).
  • Table 4 Plasmids and IL-2 receptor-targeted fusion toxins used in this study.
  • Plasmid tox gene product amino acid sequence (212-225) pETJV127 DAB 3 89lL-2 DKTKTKIESLKEHG pETCT20 DAB(K213A) 389 IL-2 DATKTKIESLKEHG pETCT30 DAB(K215A)389IL-2 D TATKIESLKEHG pETCT40 DAB(K217A)3 89 IL-2 DKTKTAIESLKEHG pETCT50 DAB(K222A) 389 IL-2 DKTKTKIESLAEHG pETCT80 DAB(K213 A,K215 A) 389 IL-2 DATATKIESLKEHG pETCT90 DAB(K215 A,K217A) 389 lL-2 DKTATAIESLKEHG pETCT60 DAB(K213,K215,K217,K222- >A) 389 IL-2 DATATAIESLAEHG pETCT70 DAB(212p23) 389 IL-2
  • Cytotoxicity Assay Cytotoxicity assays were performed as described by vanderSpek et al. (1994). Figures were created in GraphPad Prism version 5.01 for Windows, GraphPad Software, San Diego California USA. References
  • ADP-ribosylation factor a small GTP-binding protein, is required for binding of coatomer protein beta- COP to Golgi membranes. Proc Natl Acad Sci, USA 89: 6408-6412.
  • Coatomer is essential for retrieval of dilysine- tagged proteins to the endoplasmic reticulum. Cell 79: 1199-1207.
  • Nickel, W., and Wieland, F.T. 2001 Receptor-dependent formation of COPI-coated vesicles from chemically defined donor liposomes. Methods Enzymol 329: 388-404.
  • Boquet P Silverman MS, Pappenheimer AM Jr, & Vernon WB. (1976) Binding of triton X-100 to diphtheria toxin, crossreacting material 45, and their fragments. Proc Natl Acad Sci, USA, 73: 4449- 4453.
  • RNAi is related to intracellular release of SIRNA via a covalently attached signal peptide.
  • RNA [Epub ahead of print].
  • RNA interference-mediated gene silencing of pleiotrophin through poyethylenimine-complexed small interfering RNAs in vivo exerts antitumoral effects in glioblastoma xenografts.
  • RNA interference targeting Fas protects mice from fulminant hepatitis. Nat Med, 9: 347-351.
  • COPI coatomer complex proteins facilitate the translocation of Anthrax Lethal Factor across vesicular membranes in vitro. Proc. Natl. Acad. Sci., USA, 105: 5254-5259.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Biophysics (AREA)
  • Biotechnology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Wood Science & Technology (AREA)
  • Nanotechnology (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Medical Informatics (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Toxicology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Peptides Or Proteins (AREA)

Abstract

L'invention concerne des compositions pour l'administration d'agents cytotoxiques, thérapeutiques, diagnostiques à une cellule cible et des procédés d'utilisation de ces compositions pour le traitement de maladies.
PCT/US2011/033229 2010-04-22 2011-04-20 Compositions et procédés de ciblage et d'administration d'agents thérapeutiques dans des cellules WO2011133658A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US32702410P 2010-04-22 2010-04-22
US61/327,024 2010-04-22

Publications (1)

Publication Number Publication Date
WO2011133658A1 true WO2011133658A1 (fr) 2011-10-27

Family

ID=44210116

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/033229 WO2011133658A1 (fr) 2010-04-22 2011-04-20 Compositions et procédés de ciblage et d'administration d'agents thérapeutiques dans des cellules

Country Status (1)

Country Link
WO (1) WO2011133658A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8513390B2 (en) 2009-01-12 2013-08-20 Cytomx Therapeutics, Inc. Modified antibody compositions, methods of making and using thereof
WO2016191869A1 (fr) * 2015-06-01 2016-12-08 The Hospital For Sick Children Administration de cargo polypeptidique de diverses structures dans des cellules mammaliennes par une toxine bactérienne
WO2020206375A1 (fr) * 2019-04-03 2020-10-08 Technical University Of Denmark Conjugués de protéine de facteur neurotrophique et modes de réalisation associés
US11045546B1 (en) 2020-03-30 2021-06-29 Cytodyn Inc. Methods of treating coronavirus infection

Citations (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1983003679A1 (fr) 1982-04-12 1983-10-27 Hybritech Inc Anticorps ayant des doubles specificites, leur preparation et utilisation
WO1986001533A1 (fr) 1984-09-03 1986-03-13 Celltech Limited Production d'anticorps chimeriques
EP0217577A2 (fr) 1985-09-12 1987-04-08 Hybritech Incorporated Complexes d'anticorps d'agents diagnostiques ou thérapeutiques modifiés par un haptène
EP0239400A2 (fr) 1986-03-27 1987-09-30 Medical Research Council Anticorps recombinants et leurs procédés de production
US5595760A (en) 1994-09-02 1997-01-21 Delab Sustained release of peptides from pharmaceutical compositions
US5672659A (en) 1993-01-06 1997-09-30 Kinerton Limited Ionic molecular conjugates of biodegradable polyesters and bioactive polypeptides
WO1997040055A1 (fr) 1996-04-19 1997-10-30 The Dow Chemical Company Chelates fluorescents utilises en tant qu'agents visuels destines a l'imagerie et specifiques de certains tissus
US5942225A (en) 1995-01-24 1999-08-24 Case Western Reserve University Lineage-directed induction of human mesenchymal stem cell differentiation
US6037329A (en) 1994-03-15 2000-03-14 Selective Genetics, Inc. Compositions containing nucleic acids and ligands for therapeutic treatment
WO2001058936A2 (fr) * 1999-12-02 2001-08-16 Microbiological Research Authority Constructions destinees a la diffusion d'agents therapeutiques dans des cellules neuronales
WO2005014798A2 (fr) 2003-03-31 2005-02-17 Boston Medical Center Corporation Nouvelles compositions et nouveaux procedes pour stimuler, inhiber et detecter l'entree de proteines dans des cellules
US20050118099A1 (en) 2003-03-10 2005-06-02 Braslawsky Gary R. Thiol-mediated drug attachment to targeting peptides
US7056704B2 (en) 2000-12-01 2006-06-06 Max-Planck-Gesellschaft Zur Foerderung Der Wissenschaften E.V. RNA interference mediating small RNA molecules
US20080249046A1 (en) 2006-06-09 2008-10-09 Protiva Biotherapeutics, Inc. MODIFIED siRNA MOLECULES AND USES THEREOF
US20080260854A1 (en) 2005-01-19 2008-10-23 Tobias Walker Sirna Molecules for the Treatment of Blood Vessels
US20080306003A1 (en) 2005-08-30 2008-12-11 Murphy John R Novel Compositions and Methods for Promoting, Inhibiting and Detecting Protein Entry Into Cells
WO2009083738A2 (fr) * 2007-12-31 2009-07-09 Syntaxin Limited Véhicules de délivrance d'arn
US20090318536A1 (en) 2006-11-27 2009-12-24 Iss Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
US20100063134A1 (en) 2002-11-26 2010-03-11 Medtronic, Inc. Treatment of neurodegenerative disease through intracranial delivery of sirna
US20100062967A1 (en) 2004-12-27 2010-03-11 Silence Therapeutics Ag Coated lipid complexes and their use
US20100062051A1 (en) 2006-11-03 2010-03-11 Young Kee Shin Composition for treatment of cervix cancer
US20100063308A1 (en) 2005-02-14 2010-03-11 Tongqian Chen Lipid nanoparticle based compositions and methods for the delivery of biologically active molecules
US20100062436A1 (en) 2006-10-31 2010-03-11 Noxxon Pharma Ag Methods for Detection of a Single- or Double-Stranded Nucleic Acid Molecule
US20100062951A1 (en) 2002-11-14 2010-03-11 Dharmacon, Inc. siRNA targeting TIE-2
US20100063131A1 (en) 2007-03-26 2010-03-11 Hirofumi Takeuchi Prompt nucleic acid delivery carrier composition
US20100063132A1 (en) 2005-03-09 2010-03-11 Mogam Biotechnology Research Institute Small interfering rna and pharmaceutical composition for treatment of hepatitis b comprising the same
US7678897B2 (en) 2002-02-20 2010-03-16 Sirna Therapeutics, Inc. RNA interference mediated inhibition of platelet-derived endothelial cell growth factor (ECGF1) gene expression using short interfering nucleic acid (siNA)
US7691998B2 (en) 2002-11-14 2010-04-06 Dharmacon, Inc. siRNA targeting nucleoporin 62kDa (Nup62)
US20100098664A1 (en) 2007-11-28 2010-04-22 Mathieu Jean-Francois Desclaux Lentiviral vectors allowing RNAi mediated inhibition of GFAP and vimentin expression

Patent Citations (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0105360A1 (fr) 1982-04-12 1984-04-18 Hybritech Incorporated Anticorps ayant des doubles specificites, leur preparation et utilisation
WO1983003679A1 (fr) 1982-04-12 1983-10-27 Hybritech Inc Anticorps ayant des doubles specificites, leur preparation et utilisation
WO1986001533A1 (fr) 1984-09-03 1986-03-13 Celltech Limited Production d'anticorps chimeriques
EP0217577A2 (fr) 1985-09-12 1987-04-08 Hybritech Incorporated Complexes d'anticorps d'agents diagnostiques ou thérapeutiques modifiés par un haptène
EP0239400A2 (fr) 1986-03-27 1987-09-30 Medical Research Council Anticorps recombinants et leurs procédés de production
US5672659A (en) 1993-01-06 1997-09-30 Kinerton Limited Ionic molecular conjugates of biodegradable polyesters and bioactive polypeptides
US6037329A (en) 1994-03-15 2000-03-14 Selective Genetics, Inc. Compositions containing nucleic acids and ligands for therapeutic treatment
US5595760A (en) 1994-09-02 1997-01-21 Delab Sustained release of peptides from pharmaceutical compositions
US5942225A (en) 1995-01-24 1999-08-24 Case Western Reserve University Lineage-directed induction of human mesenchymal stem cell differentiation
WO1997040055A1 (fr) 1996-04-19 1997-10-30 The Dow Chemical Company Chelates fluorescents utilises en tant qu'agents visuels destines a l'imagerie et specifiques de certains tissus
WO2001058936A2 (fr) * 1999-12-02 2001-08-16 Microbiological Research Authority Constructions destinees a la diffusion d'agents therapeutiques dans des cellules neuronales
US7056704B2 (en) 2000-12-01 2006-06-06 Max-Planck-Gesellschaft Zur Foerderung Der Wissenschaften E.V. RNA interference mediating small RNA molecules
US7678897B2 (en) 2002-02-20 2010-03-16 Sirna Therapeutics, Inc. RNA interference mediated inhibition of platelet-derived endothelial cell growth factor (ECGF1) gene expression using short interfering nucleic acid (siNA)
US7691998B2 (en) 2002-11-14 2010-04-06 Dharmacon, Inc. siRNA targeting nucleoporin 62kDa (Nup62)
US20100062951A1 (en) 2002-11-14 2010-03-11 Dharmacon, Inc. siRNA targeting TIE-2
US7678896B2 (en) 2002-11-14 2010-03-16 Dharmacon, Inc. siRNA targeting serine/threonine kinase 12 (STK12 or aurora B kinase)
US20100063134A1 (en) 2002-11-26 2010-03-11 Medtronic, Inc. Treatment of neurodegenerative disease through intracranial delivery of sirna
US20050118099A1 (en) 2003-03-10 2005-06-02 Braslawsky Gary R. Thiol-mediated drug attachment to targeting peptides
WO2005014798A2 (fr) 2003-03-31 2005-02-17 Boston Medical Center Corporation Nouvelles compositions et nouveaux procedes pour stimuler, inhiber et detecter l'entree de proteines dans des cellules
US20100062967A1 (en) 2004-12-27 2010-03-11 Silence Therapeutics Ag Coated lipid complexes and their use
US20080260854A1 (en) 2005-01-19 2008-10-23 Tobias Walker Sirna Molecules for the Treatment of Blood Vessels
US20100063308A1 (en) 2005-02-14 2010-03-11 Tongqian Chen Lipid nanoparticle based compositions and methods for the delivery of biologically active molecules
US20100063132A1 (en) 2005-03-09 2010-03-11 Mogam Biotechnology Research Institute Small interfering rna and pharmaceutical composition for treatment of hepatitis b comprising the same
US20080306003A1 (en) 2005-08-30 2008-12-11 Murphy John R Novel Compositions and Methods for Promoting, Inhibiting and Detecting Protein Entry Into Cells
US20080249046A1 (en) 2006-06-09 2008-10-09 Protiva Biotherapeutics, Inc. MODIFIED siRNA MOLECULES AND USES THEREOF
US20100062436A1 (en) 2006-10-31 2010-03-11 Noxxon Pharma Ag Methods for Detection of a Single- or Double-Stranded Nucleic Acid Molecule
US20100062051A1 (en) 2006-11-03 2010-03-11 Young Kee Shin Composition for treatment of cervix cancer
US20090318536A1 (en) 2006-11-27 2009-12-24 Iss Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
US20100063131A1 (en) 2007-03-26 2010-03-11 Hirofumi Takeuchi Prompt nucleic acid delivery carrier composition
US20100098664A1 (en) 2007-11-28 2010-04-22 Mathieu Jean-Francois Desclaux Lentiviral vectors allowing RNAi mediated inhibition of GFAP and vimentin expression
WO2009083738A2 (fr) * 2007-12-31 2009-07-09 Syntaxin Limited Véhicules de délivrance d'arn

Non-Patent Citations (157)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING COMPANY
A.R. GENNARO: "Remington: The Science and Practice of Pharmacy(20th ed.,", 2000, LIPPINCOTT WILLIAMS & WILKINS
ABRAMI L, LINDSAY M, PARTON RG, LEPPLA SH, VAN DER GOOT FG.: "Membrane insertion of anthrax protective antigen and cytoplasmic delivery of lethal factor occur at different stages of the endocytic pathway", J CELL BIOL, vol. 166, 2004, pages 645 - 651
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
AMERES SL, MARTINEZ J, SCHROEDER R.: "Molecular basis for target RNA recognition and cleavage by human RISC", CELL, vol. 130, 2007, pages 101 - 112
ARIANSEN ET AL., BIOCHEM., vol. 32, 1993, pages 83 - 90
ARYA ET AL., BIOCONJUGATE CHEMISTRY, vol. 2, 1991, pages 323
BACHA ET AL., J. EXP. MED., vol. 167, 1988, pages 612 - 622
BACHA, P., WILLIAMS, D.P., WATERS, C., WILLIAMS, J.M., MURPHY, J.R., STROM, T.B.: "Intcrlcukin 2 receptor-targeted cytotoxicity. Interleukin 2 receptor-mediated action of a diphtheria toxin-related interleukin 2 fusion protein", JEXP MED, vol. 167, 1988, pages 612 - 622
BARONE F, CELLAI L, MATZEU M, MAZZEI F, PEDONE F: "DNA, RNA, and hybrid RNA-DNA oligomers of identical sequence: structural and dynamic differences", BIOPHYS CHCM, vol. 86, 2000, pages 37 - 47
BARTLETT DW, DAVIS ME: "Insights into the kinetics of siRNA-mediated gene silencing from live-cell and live animal bioluminescent imaging", NUCLEIC ACIDS RES, vol. 34, 2006, pages 322 - 334
BENNETT MJ, CHOE S, EISENBERG D: "Refined structure of dimeric diphtheria toxin at 2.OA resolution", PROTEIN SCI, vol. 3, 1994, pages 1444 - 1463
BERNSTEIN E, CAUDY AA, HAMMOND SM, HANNON GJ: "Role for a bidentate ribonuclease in the initiation step of RNA interference", NATURE, vol. 409, 2001, pages 295 - 296
BETHUNE, J., KOL, M., HOFFMANN, J., RECKMANN, I., BRÜGGER, B., WIELAND, F.: "Coatomer, the coat protein of COPl transport vesicles, discriminates endoplasmic reticulum residents from p24 proteins", MOL CELL BIOL, vol. 26, 2006, pages 8011 - 8021
BETHUNE, J., WIELAND, F., MOELLEKEN, J.: "COPI-mediated transport", J MEMBR BIOL, vol. 211, 2006, pages 65 - 79
BLANKE SR, MILNE JC, BENSON EL, COLLIER RJ: "Fused polycationic peptide mediates delivery of diphtheriatoxin A chain to the cytosol in the presence of anthrax protective antigen", PROC NATL ACAD SCI, USA, vol. 93, 1996, pages 8437 - 8442
BOQUET P, SILVERMAN MS, PAPPENHEIMER AM JR, VERNON WB: "Binding of triton X-100 to diphtheria toxin, crossreacting material 45, and their fragments", PROC NATL ACAD SCI, USA, vol. 73, 1976, pages 4449 - 4453
BRADLEY KA, MOGRIDGE J, MOUREZ M, COLLIER RJ, YOUNG JA: "Identification of the cellular receptor for anthrax toxin", NATURE, vol. 414, 2001, pages 225 - 229
BUMS ET AL., CURR. STEM CELL RES. THER., vol. 2, 2006, pages 255 - 266
CAROLINA TRUJILLO ET AL: "Essential lysine residues within transmembrane helix 1 of diphtheria toxin facilitate COPI binding and catalytic domain entry", MOLECULAR MICROBIOLOGY, vol. 76, no. 4, 14 April 2010 (2010-04-14), pages 1010 - 1019, XP055002126, ISSN: 0950-382X, DOI: 10.1111/j.1365-2958.2010.07159.x *
CHEN ET AL., STEM CELL REV., vol. 3, no. 4, 2007, pages 280 - 288
CHENG K, TE Z, GUNTAKA RV, MAHATO RI: "Enhanced hepatic uptake and bioactivity of type alphal (I) collagen gene promoter-specific triplex-forming oligonucleotides after conjugation with cholesterol", J PHARMACOL EXP THER, vol. 317, 2006, pages 797 - 805
CHOE ET AL., NATURE, vol. 357, 1992, pages 216 - 22
CHOE S, BENNETT MJ, FUJII G, CURMI PM, KANTARDJIEFF KA, COLLIER RJ, EISENBERG D: "The crystal structre of diphtheria toxin", NATURE, vol. 357, 1992, pages 216 - 222
COLLIER RJ, KANDEL J: "Structure and activity of diphtheria toxin. 1. Thiol- dependent dissociation of a fraction of toxin into enzymatically activity and inactive fragments", J BIOL CHEM, vol. 246, 1971, pages 1496 - 1503
COSSON P, LETOUMEUR F: "Coatomer interaction with di-lysine endoplasmic retention motifs", SCIENCE, vol. 263, 1994, pages 1629 - 1631
COSSON, P., LETOUMEUR, F.: "Coatomer interaction with di-lysine endoplasmic reticulum retention motifs", SCIENCE, vol. 263, 1994, pages 1629 - 1631
COUTTS, KEIRSTEAD, EXP. NEUROL., vol. 209, no. 2, 2008, pages 368 - 377
CURTER, INVEST. RADIOL., vol. 33, no. 10, 1998, pages 752 - 761
DAVIDSON TJ, HAREL S, ARBOLEDA VA, PRUNELL GF, SHELANSKI ML, GREENE LA, TROY CM: "Highly efficient small interfering RNA delivery to primary mammalian neurons induces MicroRNA-like effects before mRNA degradation", J NEUROSCI, vol. 24, 2004, pages 10040 - 10046
DEROSSI D, JOLIOT AH, CHASSAING G, PROCHIZANTZ A: "The third helix of Antennapedia homeodomain translocates through biological membranes", J BIOL CHEM, vol. 269, 1994, pages 10444 - 10450
DESHAYES S, MORRIS M, HEITZ F, DIVITA G: "Delivery of proteins and nucleic acids using non-covalent peptide-based strategy", ADV DRUG DCLIV REV, vol. 60, 2008, pages 537 - 547
DETZER A, OVERHOFF M, WUNCHE W, ROMPF M, TURNER JJ, IVANOVA GD, GAIT MJ, SCZAKIEL G: "Omcreased RNAi is related to intracellular release of SIRNA via a covalently attached signal peptide", RNA, 2009
DONALDSON,J.G., CASSEL, D., KAHN, R.A., KLAUSNER, R.D.: "ADP-ribosylation factor, a small GTP-binding protein, is required for binding of coatomer protein beta-COP to Golgi membranes", PROC NATL ACAD SCI, USA, vol. 89, 1992, pages 6408 - 6412
DONOVAN ET AL., PROC. NATL. ACAD. SCI., USA, vol. 78, 1981, pages 172
DONOVAN JJ, SIMON MI, MONTAL M: "Diphtheria toxin forms transmembrane channels in planar lipid bilayers", PROC NATL ACAD SCI, USA, vol. 78, 1981, pages 172 - 176
DONOVAN, J.J., SIMON, M.I., MONTAL, M.: "Requirements for the translocation of diphtheria toxin fragment A across lipid membranes", JBIOL CHEM, vol. 260, 1981, pages 8817 - 8823
E. S. WARD ET AL., NATURE, vol. 341, 1989, pages 544
ECKSTEIN F.: "The versatility of oligonucleotides as potential therapeutic", EXPERT OPIN BIOL THER, vol. 7, 2007, pages 1021 - 1034
ELBASHIR SM, HARBOTH J, LENDECKEL W, YALCIN A, WEBER K, TUSCHI T: "Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells", NATURE, vol. 411, 2001, pages 494 - 498
ENDOH T, SISIDO M, OHTSUKI T: "Cellular siRNA delivery mediated by a cell- permeant RNA-binding protein and photoinduced RNA interference", BIOCONJUGATE CHEM, vol. 19, 2008, pages 1017 - 1024
EUGSTER, A., FRIGERIO, G., DALE, M., DUDEN, R.: "The alpha- and beta'-COP WD40 domains mediate cargo-selective interactions with distinct di-lysine motifs", MOL BIOL CELL, vol. 15, 2004, pages 1011 - 1023
FELGNER PL, GADEK TR, HOLM M, ROMAN R, CHAN HW, WENZ M, NORTHROP JP, RINGOLD GM, DANIELSON M: "Lipofection: a highly efficient, lipid-mediated DNA-transfcction procedure", PROC NATL ACAD SCI, USA, vol. 84, 1987, pages 7413 - 7417
FIEDLER K, VEIT M, STAMMES MA, ROTHMAN JE: "Bimodal interaction of coatomer with the p24 family of putative cargo receptors", SCIENCE, vol. 273, 1996, pages 1396 - 1399
FOSS FM.: "DAB(389)IL-2 (ONTAK): a novel fusion toxin therapy for lymphoma", CLIN LYMPHOMA, vol. 1, 2000, pages 110 - 116
FRANCINE M FOSS: "DAB389IL-2 (Denileukin Diftitox, ONTAK): A New Fusion Protein Technology", CLINCIAL LYMPHOMA, MYELOMA & LEUKEMIA, vol. 1, no. Suppl. 1, 1 November 2000 (2000-11-01), pages S27 - S31, XP055002654, ISSN: 2152-2650, DOI: 10.3816/CLM.2000.s.005 *
FRANKEL AD, PABO CO: "Cellular uptake of the tat protein from human immunodeficiency virus", CELL, vol. 55, 1988, pages 1189 - 1193
G. KOHLER, C. MILSTEIN, NATURE, vol. 256, 1975, pages 495
G. WINTER, C. MILSTEIN, NATURE, vol. 349, 1991, pages 295
GE Q, FILIP L, BAI A, NGUYEN T, EISEN HN, CHEN J: "Inhibition of influenza virus production in virus-infected mice by RNA interference", PROC NATL ACAD SCI, USA, vol. 101, 2004, pages 8676 - 8681
GILL DM, PAPPENHEIMER AM, JR: "Structure-activity relationships in diphtheria toxin", J BIOL CHCM, vol. 246, 1971, pages 1492 - 1495
GORDON VM, KLIMPEL KR, ARORA H, HENDERSON MA, LEPPLA SH: "Proteolytic activation of bacterial toxin by eukaryotic cells is performed by furin and by additional cellular proteases", INFECT IMMUN, vol. 63, 1995, pages 82 - 87
GOSWAMI, RAO, DRUGS, vol. 10, no. 10, 2007, pages 713 - 719
GREENFIELD L, BJORN MJ, HORN G, FONG D, BUCK GA, COLLIER RJ, KAPLAN DA: "Nucleotide sequence of the structural gene for diphtheria toxin carried by corynephage beta", PROC NATL ACAD SCI, USA, vol. 80, 1983, pages 6853 - 6857
GRZLINSKI M, URBAN-KLEIN B, MARTENS T, LAMSZUS K, BAKOWSKY U, HOBEL S, CZUBAYKO F, AIGNER A: "RNA interference-mediated gene silencing of pleiotrophin through poyethylenimine-complexed small interfering RNAs in vivo exerts antitumoral effects in glioblastoma xenografts", HUM GENE THER, vol. 17, 2006, pages 751 - 766
HALDER J, KAMAT AA, LANDEN CN JR, HAN LY, LUTGENDORF SK, LIN YG, MERRITT WM, JENNINGS NB, CHAVEZ-REVES A, COLEMAN RL: "Focal adhesion kinase targeting using in vivo short interfering RNA delivery in ncutral liposomes for ovarian caricinoma therapy", CLIN CANCER RES, vol. 12, 2006, pages 4916 - 4924
HARTER, C., WIELAND, F.T.: "A single binding site for dilysine retrieval motifs and p23 with the gamma subunit of coatomer", PROC NATL A CAD SCI, USA, vol. 95, 1998, pages 11649 - 11654
HONJO T, NISHIZUKA Y, HAYAISHI O: "Diphtheria toxin-dependent adenosine diphosphate ribosylation ofaminoacyi transferase II and inhibition of protein synthesis", J BIOL CHEM, vol. 243, 1968, pages 3553 - 3555
HUDSON, R.T., DRAPER, R.K.: "Interaction of coatomer with aminoglycoside antibiotics: evidence that coatomer has at least two dilysine binding sites", MOL BIOL CELL, vol. 8, 1997, pages 1901 - 1910
IWAMOTO R, HIGASHIYAMA S, MITAMURA T, TANIGUCHI N, KLAGSBRUN M, MEKADA E: "Heparin-binding EGF-like growth factor, which acts as the diphtheria toxin rcceptor, forms a complex with membrane protein DRAP27/CD9, which up-regulates functional receptors and diphtheria toxin sensitivity", EMBO J, vol. 13, 1994, pages 2322 - 2330
JANSSENS ET AL., LANCET, vol. 367, 2006, pages 113 - 121
KAGAN BL, FINKELSTEIN A, COLOMBINI M: "Diphtheria toxin fragment forms large pores in phospholipid bilayer membranes", PROC NATL ACAD SCI,USA, vol. 78, 1981, pages 4950 - 4954
KAGAN ET AL., PROC. NATL. ACAD SCI., USA, vol. 78, 1981, pages 4950
KAGAN, B.L., FINKELSTEIN, A., COLOMBINI, M.: "Diphtheria toxin fragment forms large pores in phospholipids bilayer membranes", PROC NATL ACAD SCI, USA, vol. 78, 1981, pages 4950 - 4954
KAHL ET AL., PROC. NATL. ACAD SCI. USA, vol. 87, 1990, pages 7265 - 7269
KANEDA Y, UCHIDA T, MEKADA E, NAKANISHI M, OKADA Y: "Entry of diphtheria toxin into cells: possible existence of cellular factor(s) for entry of diphtheria toxin into cells was studied in somatic cell hybrids and hybrid toxins", J CELL BIOL, vol. 98, 1984, pages 466 - 472
KAZOREK M, DELPEYROUX F, CHENCINER N, STREECK R, MURPHY JR, BOQUET P, TIOLLAIS P: "Nucleotide sequence and expression in Escherichia coli of the tox228 diphtheria toxin", GENE. SCIENCE, vol. 221, 1983, pages 855 - 858
KEM ET AL., J. BIOL. CHEM., vol. 253, no. 16, 1978, pages 5752 - 5757
KLIMPEL KR, MOLLOY SS, THOMAS G, LEPPLA SH: "Anthrax toxin protective antigen is activated by a cell surface protease with the sequence specificity and catalytic properties of furin", PROC NATL ACAD SCI, USA, vol. 89, 1992, pages 10277 - 10281
KOCHI SK, COLLIER RJ: "DNA fragmentation and cytolysis in U937 cells treated with diphtheria toxin or other inhibitors of protein synthesis", EXPERIMENTAL CELL RESEARCH, vol. 208, 1993, pages 296 - 302
KOCHI, COLLIER, EXP. CELL. RES., vol. 208, 1993, pages 296
KOUKIEKOLO R, SAGAN SM, PEZACKI JP: "Effects of pH and salt concentration on the siRNA binding activity of the RNA silencing suppressor protein p19", FEBS LETT, vol. 581, 2007, pages 3051 - 3056
L. RIECHMANN ET AL., NATURE, vol. 332, 1988, pages 323
LAMOYI ET AL., J. IMMUNOLOGICAL METHODS, vol. 56, 1983, pages 235
LEBLEU B, MOULTON HM, ABES R, IVANOVA GD, ABES S, STEIN DA, IVERSEN PL, ARZUMANOV AA, GAIT MJ: "Cell penetrating peptide conjugates of steric clock oligonucleotidcs", ADV DRUG DELIV RES, vol. 60, 2008, pages 517 - 529
LEMICHEZ E, BOMSEL M, DEVILLIERS G, VANDERSPEK J, MURPHY JR, LUKIANOV EV, OLSNER S, BOQUET P: "Membrane translocation of diphtheria toxin fragment A exploits early to late endosome trafficking machinery", MOLE MICROBIOL, vol. 23, 1997, pages 445 - 457
LEMICHEZ, E., BOMSEL, M., VANDERSPEK, J.C., LUKIANOV, E.V., MURPHY, J.R., OLSNES, S., BOQUET, P.: "Membrane translocation of diphtheria toxin fragment A exploits early to late endosomal trafficking machinery", MOL MICROBIOL, vol. 23, 1997, pages 445 - 457
LETOUMEUR, F., GAYNOR, E.C., HENNECKE, S., DÉMOLLIÈRE, C., DUDEN, R., EMR, S.D., RIEZMAN, H., COSSON, P.: "Coatomer is essential for retrieval of dilysine- tagged proteins to the endoplasmic reticulum", CELL, vol. 79, 1994, pages 1199 - 1207
LEUSCHNER ET AL., BIOLOGY OF REPRODUCTION, vol. 73, 2005, pages R60 - 865
LOVE, MURPHY: "Gram-Positive Pathogens", 2000, AMERICAN SOCIETY FOR MICROBIOLOGY, pages: 573 - 582
M. S. NEUBERGER ET AL., NATURE, vol. 314, 1985, pages 268
MADHUS IH, OLSNES S, STENRNARK H: "Membrane translocation of diphtheria toxin carrying passenger protein domains", INFECT IMMUN, vol. 60, 1992, pages 3296 - 3302
MADSHUS I H ET AL: "Membrane translocation of diphtheria toxin carrying passenger protein domains.", INFECTION AND IMMUNITY AUG 1992 LNKD- PUBMED:1639497, vol. 60, no. 8, August 1992 (1992-08-01), pages 3296 - 3302, XP002649736, ISSN: 0019-9567 *
MALONE RW, FELGNER PL, VERMA IM: "Cationic liposome-mediated RNA transfection", PROC NATL ACAD SCI, USA, vol. 86, 1989, pages 6077 - 6081
MATRANGA C, TOMARI Y, SHIN C, BARTEL DP, ZAMORE PD: "Passenger-strand cleavage facilitates assembly of siRNA into Ago2-containing RNAi enzyme complex", CELL, vol. 123, 2005, pages 607 - 620
MAUPETIT J, DERREUMAUX P, TUFFERY P.: "PEP-FOLD: on online resource for de novo peptide structure prediction", NUCLEIC ACIDS RES, 2009
MESCALIN A, DETZER A, WCCKC M, OVERHOFF M, WUNSCHE W, SCZAKIEL G: "Cellular uptake and intracellular release are major obstacles to the therapeutic application of siRNA: novel options by hosphorthionate-stimulated delivery", EXPERT OPIN BIOL THER, vol. 7, 2007, pages 1531 - 1538
MEYER MARTIN ET AL: "Synthesis and Biological Evaluation of a Bioresponsive and Endosomolytic siRNA-Polymer Conjugate", MOLECULAR PHARMACEUTICS, AMERICAN CHEMICAL SOCIETY, US, vol. 6, no. 3, 1 June 2009 (2009-06-01), pages 752 - 762, XP002624572, ISSN: 1543-8384, [retrieved on 20090406], DOI: DOI:10.1021/MP9000124 *
MILNE JC, BLANKE SR, HANNA PC, COLLIER RJ: "Protective antigen-binding domain of anthrax lethal factor mediates translocation of a heterologous protcin fused to its amino- or carboxy-terminus", MOL MICROBIOL, vol. 15, 1995, pages 661 - 666
MINAKUCHI Y, TAKESHITA F, KOSAKA N, SASAKI I I, YAMAMOTO Y, KOUNO M, HONMA K, NAGAHARA S, HANAI K, SATO A: "Atelocollagen- mediated synthetic small interfering RNA delivery for effective gene silencing in vitro and in vivo", NUCLEIC ACIDS RES, vol. 32, 2004, pages 109
MORIZANE ET AL., CELL TISSUE RES., vol. 331, no. 1, 2008, pages 323 - 326
MORRISSEY DV, LOCKRIDGE JA, SHAW L, BLANCHARD K, JANSEN K, BREEN W, HARTSOUGH K, MACHEMER L, RADKA S, JADHAV, V: "Potent and persistent anti-HBV activity of chemically modified siRNAs", NAT BIOTECHNOL, vol. 23, 2005, pages 1002 - 1007
MOSCHOS SA, JONES SW, PERRY MM, WILLIAMS AE, ERJCDALT JS, TURNER JJ, BARNES PJ, SPROAT BS, GAIT MJ, LINDSAY MA, BIOCONJUG CHEM, vol. 18, 2007, pages 1450 - 1459
MOYA ET AL., J CELL. BIOL., vol. 101, 1985, pages 548
MOYA M, DAUTRY-VARSAT A, GOUD B, LOUVARD D, BOQUET P: "Inhibition of coated pit formation in Hep2 cells blocks the cytotoxicity of diphtheria toxin but not that of ricin toxin", J CELL BIOL, vol. 101, 1985, pages 548 - 559
MOYA, M., DAUTRY-VERSAT, A., GOUD, B., LOUVARD, D., BOQUET, P.: "Inhibition of coated pit formation in Hep2 cells blocks the cytotoxicity of diphtheria toxin but not that or ricin toxin", J CELL BIOL, vol. 101, 1985, pages 548 - 549
MURATOVSKA A, ECCLES MR: "Conjugate for efficient delivery of short interfering RNA (siRNA) into mammalian cells", FEBS LETT, vol. 558, 2004, pages 63 - 68
MURPHY JR, BISHAI W, BOROWSKI M, MIYANOHARA A, BOYD J, NAGLE S: "Genetic construction, expression, and melanoma-selective cytotoxicity of a diphtheria toxin-related alpha-melanocyte-stimulating hormone fusion protein", PROC NATL ACAD SCI, USA, vol. 83, 1986, pages 8258 - 8262
NAGLICH JG, METHERALL JE, RUSSELL DW, EIDELS L: "Expression cloning of a diphtheria toxin receptor: identity with a heparin-binding EGF-like growth factor precursor", CELL, vol. 69, 1992, pages 1051 - 1061
NAGLICH, J.G., METHERALL, J.E., RUSSELL, D.W., EIDELS, L.: "Expression cloning of a diphtheria toxin receptor: identity with a heparin-binding EGF-like growth factor precursor", CELL, vol. 69, 1992, pages 1051 - 1061
NICKEL, W., WIELAND, F.T.: "Receptor-dependent formation of COPI-coated vesicles from chemically defined donor liposomes", METHODS ENZYMOL, vol. 329, 2001, pages 388 - 404
NIU XY, PENG ZL, DUAN WQ, WANG H, WANG P: "Inhibition of HPV 16 E6 oncogene expression by RNA interference in vitro and in vivo", INT J GYNECOL CANCER, vol. 16, 2006, pages 743 - 751
OH, K.J., SENZEL, L., COLLIER, R.J., FINKELSTEIN, A.: "Translocation of the catalytic domain of diphtheria toxin across planar phospholipids bilayers by its own T domain", PROC NAIL ACAD SCI, USA, vol. 96, 1999, pages 8467 - 8470
OVERHOFF M, SCZAKIEL G: "Phosphothioate-stimulated uptake of short interfering RNA by human cells", EMBO REP, vol. 6, 2005, pages 1176 - 1181
PALMER,D.J., HELMS, J.B., BECKERS, C.J., ORCI, L., ROTHMAN, J.E.: "Binding of coatomer to Golgi membranes requires ADP-ribosylation factor", J BIOL CHEM, vol. 268, 1993, pages 12083 - 12089
PAPPENHEIMER, ANNU. REV. BIOCHEM., vol. 46, 1977, pages 69
PARHAM, J. IMMUNOLOGY, vol. 131, 1983, pages 2895
PING WANG ET AL: "Diphtheria Toxin Mutant CRM197-Mediated Transcytosis across Blood-Brain Barrier In Vitro", CELLULAR AND MOLECULAR NEUROBIOLOGY, vol. 116, no. 1, 16 January 2010 (2010-01-16), pages 2333 - 725, XP055002647, ISSN: 0272-4340, DOI: 10.1007/s10571-010-9496-x *
R. GLOCKSHUBER ET AL., BIOCHEMISTRY, vol. 29, 1990, pages 1362
R. RATTS: "A conserved motif in transmembrane helix 1 of diphtheria toxin mediates catalytic domain delivery to the cytosol", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 102, no. 43, 25 October 2005 (2005-10-25), pages 15635 - 15640, XP055002656, ISSN: 0027-8424, DOI: 10.1073/pnas.0504937102 *
RAND TA, GINALSKI K, GRISHIN NV, WANG X: "Biochemical identification of Argonaute 2 as the sole protein required for RNA-induced silencing complex activity", PROC NATL ACAD SCI, USA, vol. 101, 2004, pages 14385 - 14389
RATTS R, TRUJILLO C, BHARTI A, VANDERSPEK C, HARRISON R, MURPHY JR.: "A conserved motif in transmembrane helix 1 of diphtheria toxin mediates catalytic domain delivery to the cytosol", PROC NATL ACAD SCI, IJSA, vol. 102, 2005, pages 15635 - 15640
RATTS R, ZENG H, BERG EA, BLUE C, MCCOMB ME, COSTELLO CE, VANDERSPEK JC, MURPHY JR: "The cytosolic entry of diphtheria toxin catalytic domain requires a host cell cytosolic translocation factor complex", J CELL BIOL, vol. 160, 2003, pages 1139 - 1150
RATTS, R., TRUJILLO, C., BHARTI, A., VANDERSPEK, J., HARRISON, R., MURPHY, J.R.: "A motif in transmembrane helix 1 of diphtheria toxin mediates catalytic domain delivery to the cytosol", PROC NATL ACAD SCI, USA, vol. 102, 2005, pages 15635 - 15640
RATTS, R., ZENG, H., BERG, C., BLUE, C., MCCOMB, M.E., COSTELLO, C.E, MURPHY, J.R.: "The cytosolic entry of diphtheria toxin catalytic domain requires a host cell cytosolic translocation factor complex", J CELL BIOL, vol. 160, 2003, pages 1139 - 1150
RATTS, VANDERSPEK: "Diphtheria Toxin: Structure Function and its Clinical Applications. In Chimeric Toxins", 2002, TAYLOR AND FRANCIS, pages: 14 - 36
REINHARD, C., HARTER, C., BREMSER, M., BRUGGERB., SOHN., K., HELMS, J.B., WIELAND, F.: "Receptor-induced polymerization of coatomer", PROC NATL ACAD SCI, USA, vol. 96, 1999, pages 1224 - 1228
REN, J., KACHEL, K., KIM, H., MALENBAUM, S.E., COLLIER, R.J., LONDON, E.: "Interaction of diphtheria toxin T domain with molten globule-like proteins and its implications for translocation", SCIENCE, vol. 284, 1999, pages 955 - 957
ROSOVITZ MJ, SCHUCK P, VARUGHESE M, CHOPRA AP, MEHRA V, SINGH Y, MCGINNIS LM, LEPPLA SH: "Alanine-scanning mutations in domain 4 o anthrax toxin protective antigen reveal residues important for binding to the cell receptor and to a neutraling monoclonal antibody", J BIOL CHEM, vol. 278, 2003, pages 30936 - 30944
SANTEL A, ALEKU M, KEIL 0, ENDRUSCHAT J, ESCHE V, FISCH G, DAMES S, LOFFLER K, FECHTNER M, ARNOLD W: "A novel si-RNA- lipoplex technology for RNA interference in the mouse vascular endothelium", GENE THER, vol. 13, 2006, pages 1222 - 1234
SATO Y, MURASE K, KATO J, KOBUNE M, SATO T, KAWANO T, TAKIMOTO R, TAKADA K, MIYANISHI K, MATSUNAGA: "Resolution of liver cirrhosis using vitamine A-coupled liposomes to deliver siRNA against a collagen- specific chaperone", NAT BIOTECHNOL, vol. 26, 2008, pages 431 - 442
SAZANI P, KANG SH, MAIER MA, WEI C, DILLMAN J, SUMMERTON J, MANOHARAN M, KOLE R.: "Nuclear antisense effects of neutral, anionic, and cationic oligonucleotide analogs", NUCLEIC ACDIS RES, vol. 29, 2001, pages 3965 - 3974
SCOBIE HM, RAINEY GJ, BRADLEY KA, YOUNG JA: "Human capillary morphogenesis protein 2 functions as an anthrax toxin receptor", PROC NATL ACAD SCI, USA, vol. 100, 2003, pages 5170 - 5174
SERAFINI, T., STENBECK, G., BRECHT, A., LOTTSPEICH, F., ORCI, L., ROTHMAN, J.E., WIELAND, F.T.: "A coat subunit of Golgi-derived nonclathrin-coated vesicles with homology to the clathrin-coated vesicle protein beta-adaptin", NATURE, vol. 349, 1991, pages 215 - 220
SMITH WP, TAI PC, MURPHY JR, DAVIS BD: "A precursor in the co- translational secretion of diphtheria toxin", J BACTERIOL, vol. 141, 1980, pages 184 - 189
SONG E, LEE SK, DYKXHOOM DM, NOVINA C, ZHANG D, CRAWFORD K, CERNY J, SHARP PA, LIEBERMAN J, MANJUNATH N: "Sustained small interfering RNA-mediated human immunodeficiency visur type 1 inhibition in primary macrophages", J VIROL, vol. 77, 2003, pages 7174 - 7181
SONG E, LEE SK, WANG J, INCE N, OUYANG N, MIN J, CHEN J, SHANKAR P, LIEBERMAN J: "RNA interference targeting Fas protects mice from fulminant hepatitis", NAT MED, vol. 9, 2003, pages 347 - 351
SOUTSCHEK J, AKINC A, BRAMIAGE B, CHARISSE K, CONSTEIN R, DONOGHUE M, ELBASHIR S, GEIK A, HADWIGER P, HARBOTH J: "Therapeutic silencing of an endogenous gene by systemic administration of modified siRNAs", NATURE, vol. 432, 2004, pages 173 - 178
TAKESHITA F, MINAKUCHI Y, NAGAHARA S, HONMA K, SASAKI H, HIRAI K, TERATANI T, NAMATAME N, TAMAMOTO Y, HANAI K: "Efficient delivery of small interfering RNA to bone-metastatic tumors by using atelocollagen in vivo", PROC NALT ACAD SCI, USA, vol. 102, 2005, pages 12177 - 12182
TAMAYO A, BHARTI A, TRUJILLO C, HARRISON R, MURPHY JR: "COPI coatomer complex proteins facilitate the translocation of Anthrax Lethal Factor across vesicular membranes in vitro", PROC. NATL. ACAD. SCI., USA, vol. 105, 2008, pages 5254 - 5259
TAMAYO A.G., BHARTI, A., TRUJILLO, C., HARRISON, R., MURPHY, J.R.: "COPI coatomer complex proteins facilitate the translocation of anthrax lethal factor across vesicular membrane in vitro", PROC NATL A CAD SCI, USA, vol. 105, 2008, pages 5254 - 5259
TAN PH, YANG LC, SHIH HC, LAN KC, CHENG JT: "Gene knockdown with intrathecal siRNA ofNMDA receptor NR2B subunit reduces formalin-induced nociception in the rat", GENE THER, vol. 12, 2005, pages 59 - 66
TATIANA ET AL., FEMS MICROBIOL. LETT., vol. 174, 1999, pages 247 - 250
TSUNEOKA ET AL., J BIOL. CHEM., vol. 268, 1993, pages 26461 - 5
TSUNEOKA, M., NAKAYAMA, K., HATSUZAWA, K., KOMADA, M., KITAMURA, N., MEKADA, E.: "Evidence for involvement of furin in cleavage and activation of diphtheria toxin", JBIOL CHEM, vol. 268, 1993, pages 26461 - 26465
TURNER JJ, JONES S, FABANI MM, IVANOVA G, ARZUMANOV AA, GAIT MJ: "RNA turnover with peptide conjugates of oligonucleotides, siRNA and PNA", BLOOD CELLS MOL DIS, vol. 38, 2007, pages 1 - 7
UCHIDA T, GILL DM, PAPPENHEIMER AM JR: "Mutation is the structural gene for diphtheria toxin carried by temperate phage Q", NATURE NEW BIOL, vol. 233, 1971, pages 8 - 11
UCHIDA T, PAPPENHEIMER AM JR, HARPER AA: "Diphtheria toxin and related peptides: 3. Reconstitution of hybrid "diphtheria toxin" from non-toxic mutant proteins", J BIOL CHEM, vol. 248, 1973, pages 3851 - 3854
VANDERSPEK JC, MINDELL JA, FINKELSTEIN A, MURPHY JR: "Structure/function analysis of the transmembrane domain of DAB389-interlcukin-2, and interleukin-2 receptor-targeted fusion toxin. The amphipathic helical region of the transmembrane domain is essential for the efficient delivery of the catalytic domain to the cytosol of target cells", J BIOL CHEM, vol. 268, 1993, pages 12077 - 12082
VANDERSPEK JC, MURPHY JR: "Fusion protein toxins based on diphtheria toxin: selective targeting of growth factor receptors of eukaryotic cells", METH ENZYMOL, vol. 327, 2000, pages 239 - 249
VANDERSPEK, J., CASSIDY, D., GENBAUFFE, F., HUYNH, P.D., MURPHY, J.R.: "An intact transmembrane helix 9 is essential for the efficient delivery of the diphtheria toxin catalytic domain to the cytosol of target cells", JBIOL CHEM, vol. 269, 1994, pages 21455 - 21459
VANDERSPEK, J.C., MINDELL, J.A., FINKELSTEIN, A., MURPHY, J.R.: "Structure / function analysis of the transmembrane domain of DAB389-interleukin-2, an interleukin-2 receptor targeted fusion toxin. The anphipathic helical region of the transmembrane domain is essential for the efficient delivery of the catalytic domain to the cytosol of target cells", JBIOL CHEM, vol. 268, 1993, pages 12077 - 12082
VARUGHESE M, CHI A, TEIXEIRA AV, NICHOLLS PJ, KEITH JM, LEPPLA SH: "Internalization of a Bacillus anthracis protective antigen-c-Myc fusion protein mediated by cell surface c-Myc-antibodies", MOL MED, vol. 4, 1998, pages 87 - 95
VARUGHESE M, TEIXEIRA AV, LUI S, LEPPLA SH: "Identification of a receptor- binding region within domain 4 of the protective antigen component of anthrax toxin", INFECT IMMUN, vol. 67, 1999, pages 1860 - 1865
VORNLOCHER HP.: "Antibody-directed cell-type-spccific delivery of siRNA", TRENDS MOL MED, vol. 12, 2006, pages 1 - 3
WANG J, ZUO X, YU P, XU H, STARICH MR, TIEDE DM, SHAPIRO BA, SCHWIETERS CD, WANG Y-W: "A method for helical RNA global structure determination in solution using small-angle X-ray scattering and NMR measurements", J MOL BIOL, 2009
WATERS ET AL., EUR. J IMMUNOL., vol. 20, 1990, pages 785 - 91
WATERS, M.G., DECKERS, C.J., ROTHMAN, J.E.: "Purification of coat proteins", METHODS ENZYMOL, vol. 219, 1992, pages 331 - 337
WATERS, M.G., SERAFINI, T., ROTHMAN, J.E.: "'Coatomer'" a cytosolic protein complex containing subunits of non-clathrin-coated Golgi transport vesicles", NATURE, vol. 349, 1991, pages 248 - 251
WHITNEY, J.A., GOMEZ, M., SHEFF, D., KREIS, T.E., MELLMAN, I.: "Cytoplasmic coat proteins involved in endosome function", CELL, vol. 83, 1995, pages 703 - 713
WILLIAMS, D.P., PARKER, K., BACHA, P., BISHAI, W., BOROWSKI, M., GENBAUFFE, F., STROM, T.B., MURPHY, J.R.: "Diphtheria toxin receptor binding domain substitution with interleukin-2: genetic construction and properties of a diphtheria toxin-related interleukin-2 fusion protein", PROTEIN ENG, vol. 1, 1987, pages 493 - 498
WILLIAMS, D.P., SNIDER, C.E., STROM, T.B., MURPHY, J.R.: "Structure/function analysis of interleukin-2-toxin (DAB486IL-2), fragment B sequences required for the delivery of fragment A to the cytosol of target cells", J BIOL CHEM, vol. 265, 1990, pages 11885 - 11889
WRIGHT DG, ZHANG Y, MURPHY JR: "Effective delivery of antisense peptide nucleic acid oligomers into cells by anthrax protective antigen", BIOCHEM BIOPHYS RES COMMUN, vol. 376, 2008, pages 200 - 205
WRIGHT, ZHANG, MURPHY, BIOCHEM. BIOPHYS. RES. COMMUN., vol. 376, 2008, pages 200 - 205
ZALMAN LS, WISNIESKI BJ: "Mechanism of insertion of diphtheria toxin: peptide entry and pore size determinations", PROC NATL ACAD SCI, USA, vol. 81, 1984, pages 3341 - 3345
ZERANGUE, N., MALAN, M.J., FRIED, S.R., DAZIN, P.F., JAN Y.N., JAN L.Y., SCHWAPPACH, B.: "Analysis of endoplasmic reticulum trafficking signals by combinatorial screening in mammalian cells", PROC NATL ACAD SCI, USA, vol. 98, 2001, pages 2431 - 2436
ZUCKER DR, MURPHY JR: "Monoclonal antibody analysis of diphtheria toxin. I. Localization of epitopes and neutralization of cytotoxicity", MOLEC IMMUNOL, vol. 21, 1984, pages 785 - 793

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8513390B2 (en) 2009-01-12 2013-08-20 Cytomx Therapeutics, Inc. Modified antibody compositions, methods of making and using thereof
US8563269B2 (en) 2009-01-12 2013-10-22 Cytomx Therapeutics, Inc. Modified antibody compositions, methods of making and using thereof
US9453078B2 (en) 2009-01-12 2016-09-27 Cytomx Therapeutics, Inc. Modified antibody compositions, methods of making and using thereof
US10059762B2 (en) 2009-01-12 2018-08-28 Cytomx Therapeutics, Inc. Anti-EGFR activatable antibodies
US10118961B2 (en) 2009-01-12 2018-11-06 Cytomx Therapeutics, Inc. Modified antibody containing the cleavable peptide with the amino acid sequence TGRGPSWV
US10875913B2 (en) 2009-01-12 2020-12-29 Cytomx Therapeutics, Inc. Methods of treatment using activatable anti-EGFR antibodies
WO2016191869A1 (fr) * 2015-06-01 2016-12-08 The Hospital For Sick Children Administration de cargo polypeptidique de diverses structures dans des cellules mammaliennes par une toxine bactérienne
US10597663B2 (en) 2015-06-01 2020-03-24 The Hospital For Sick Children Delivery of structurally diverse polypeptide cargo into mammalian cells by a bacterial toxin
US10954520B2 (en) 2015-06-01 2021-03-23 The Hospital For Sick Children Delivery of structurally diverse polypeptide cargo into mammalian cells by a bacterial toxin
WO2020206375A1 (fr) * 2019-04-03 2020-10-08 Technical University Of Denmark Conjugués de protéine de facteur neurotrophique et modes de réalisation associés
US11045546B1 (en) 2020-03-30 2021-06-29 Cytodyn Inc. Methods of treating coronavirus infection

Similar Documents

Publication Publication Date Title
KR102569133B1 (ko) 삼중특이적 단백질 및 사용 방법
US11859002B2 (en) Antagonistic anti-tumor necrosis factor receptor 2 antibodies
US8927694B2 (en) Human serum albumin linkers and conjugates thereof
AU2009234253C1 (en) Human serum albumin linkers and conjugates thereof
AU2003220011B2 (en) Conjugates of therapeutic or cytotoxic agents and biologically active peptides
US7326685B2 (en) Diagnostic or therapeutic somatostatin or bombesin analog conjugates and uses thereof
AU2010313122A1 (en) Analogs of pitutary adenylate cyclase-activating polypeptide (PACAP) and methods for their use
AU2002341792A1 (en) Diagnostic or therapeutic somatostatin or bombesin analog conjugates and uses thereof
US20160310609A1 (en) Compositions and methods for delivering inhibitory oligonucleotides
WO2011133658A1 (fr) Compositions et procédés de ciblage et d'administration d'agents thérapeutiques dans des cellules
WO2022256499A2 (fr) Bcma ciblant des protéines trispécifiques et méthodes d'utilisation
AU2014221306A1 (en) Compositions and methods for delivering inhibitory oligonucleotides

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11717139

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11717139

Country of ref document: EP

Kind code of ref document: A1