WO2022101463A1 - Utilisation des derniers résidus de l'extrémité c-terminale m31/41 de l'ectodomaine zikv m pour déclencher la mort cellulaire apoptotique - Google Patents

Utilisation des derniers résidus de l'extrémité c-terminale m31/41 de l'ectodomaine zikv m pour déclencher la mort cellulaire apoptotique Download PDF

Info

Publication number
WO2022101463A1
WO2022101463A1 PCT/EP2021/081631 EP2021081631W WO2022101463A1 WO 2022101463 A1 WO2022101463 A1 WO 2022101463A1 EP 2021081631 W EP2021081631 W EP 2021081631W WO 2022101463 A1 WO2022101463 A1 WO 2022101463A1
Authority
WO
WIPO (PCT)
Prior art keywords
zikv
peptide
mpf
cells
conjugate
Prior art date
Application number
PCT/EP2021/081631
Other languages
English (en)
Inventor
Philippe Despres
Gilles Gadea
Bénédicte VANWALSCAPPEL
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Centre National De La Recherche Scientifique (Cnrs)
Institut De Recherche Pour Le Développement (Ird)
Universite De La Reunion Saint Denis
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 INSERM (Institut National de la Santé et de la Recherche Médicale), Centre National De La Recherche Scientifique (Cnrs), Institut De Recherche Pour Le Développement (Ird), Universite De La Reunion Saint Denis filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2022101463A1 publication Critical patent/WO2022101463A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4748Tumour specific antigens; Tumour rejection antigen precursors [TRAP], e.g. MAGE
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • 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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/24011Flaviviridae
    • C12N2770/24111Flavivirus, e.g. yellow fever virus, dengue, JEV
    • C12N2770/24122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention is in the field of medicine, in particular virology and oncology.
  • Zika virus is an emergent mosquito-borne enveloped RNA virus belonging to flavivirus genus of Flaviviridae family.
  • ZIKV is a neurotropic pathogen that mainly targets the central nervous system (CNS) [1], leading to several neurological diseases such as congenital neurological disorders and Guillain-Barre syndrome in adults [2, 3], ZIKV strains are clustered into two major lineages, the African and Asian genotypes [4], the latter being responsible for the current epidemics with million cases of infection reported in particular, in South America.
  • CNS central nervous system
  • ZIKV strains are clustered into two major lineages, the African and Asian genotypes [4], the latter being responsible for the current epidemics with million cases of infection reported in particular, in South America.
  • human-to-human sexual or maternal-to-fetal transmission has been confirmed during the recent epidemics.
  • ZIKV contains a single genomic RNA encoding a large polyprotein that is co-and post-translationally processed into three structural proteins C, prM (the intracellular precursor of the small membrane protein M), and E followed by seven non- structural proteins NS1 to NS5.
  • C the intracellular precursor of the small membrane protein M
  • E the extracellular precursor of the small membrane protein M
  • NS1 to NS5 seven non- structural proteins NS1 to NS5.
  • the processing of prM in mature M protein (75 amino acids) by host furin/subtilisin protease family occurs in a post-Golgi compartment leading to release of mature and infectious virus particles.
  • the M protein consists of an ectodomain (hereafter referred as ectoM) composed of amino acids M-l/40 followed by a transmembrane-anchoring region including two transmembrane domains (TMDs) (Fig. 1A). It is of note that dengue M sequences are highly conserved among the four serotypes unlike other structural proteins.
  • ectoM ectodomain
  • TMDs transmembrane domains
  • the present invention is defined by the claims.
  • the present invention relates to the use of the last C-terminal residues M31/41 of ZIKV M ectodomain for triggering apoptotic cell death.
  • ZIKV Mosquito-borne Zika virus
  • Apoptosis is the main mechanism by which ZIKV infection causes cell death.
  • the inventors previously reported that expression of flavivirus M ectodomain (residues M-l/41) resulted in apoptotic cell death. Here, they aimed to determine whether the last C-terminal residues M31/41 of ZIKV M ectodomain can trigger apoptotic cell death.
  • Apoptosis was detected in human hepatoma Huh7 cells expressing of a recombinant GFP which includes ZIKV M oligopeptide at its C-terminus.
  • the triggering of apoptosis requires translocation of ZIKV M oligopeptide in the secretory pathway and involves caspase-3 activation.
  • the inventors assessed whether ZIKV M oligopeptide has ability to confer death-promoting activity to a secreted tumor-associate antigen such as megakaryocytepotentiating factor (MPF) which is a cleaved product of human mesothelin.
  • MPF megakaryocytepotentiating factor
  • ZAMP Zika Apoptosis M Peptide
  • polypeptide As used herein, the terms “polypeptide”, “peptide”, and “protein” are used interchangeably herein to refer to polymers of amino acids of any length. The terms also encompass an amino acid polymer that has been modified; for example, disulfide bond formation, glycosylation, lipidation, phosphorylation, or conjugation with a labeling component. Polypeptides when discussed in the context of gene therapy refer to the respective intact polypeptide, or any fragment or genetically engineered derivative thereof, which retains the desired biochemical function of the intact protein.
  • the expression “derived from” refers to a process whereby a first component (e.g., a first polypeptide), or information from that first component, is used to isolate, derive or make a different second component (e.g., a second polypeptide that is different from the first one).
  • a first component e.g., a first polypeptide
  • a second component e.g., a second polypeptide that is different from the first one
  • the term “having the amino sequence” has to be understood as consisting or comprising the amino sequence.
  • signal peptide refers to a leader sequence ensuring entry into the secretory pathway
  • polynucleotide refers to a polymeric form of nucleotides of any length, including deoxyribonucleotides or ribonucleotides, or analogs thereof.
  • a polynucleotide may comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs, and may be interrupted by non-nucleotide components. If present, modifications to the nucleotide structure may be imparted before or after assembly of the polymer.
  • the term "encoding" refers to the inherent property of specific sequences of nucleotides in a polynucleotide, such as, for example, a gene, a cDNA, or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides e.g., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom.
  • a gene, cDNA, or RNA encodes a protein if transcription and translation of mRNA corresponding to that gene produces the protein in a cell or other biological system.
  • nucleotide sequence encoding an amino acid sequence includes all nucleotide sequences that are degenerate versions of each other and that encode the same amino acid sequence.
  • nucleotide sequence that encodes a protein or a RNA may also include introns to the extent that the nucleotide sequence encoding the protein may in some version contain an intron(s).
  • vector means the vehicle by which a DNA or RNA sequence (e.g., a foreign gene) can be introduced into a host cell, so as to transform the host and promote expression (e.g., transcription and translation) of the introduced sequence.
  • a DNA or RNA sequence e.g., a foreign gene
  • promoter/regulatory sequence refers to a nucleic acid sequence (such as, for example, a DNA sequence) recognized by the synthetic machinery of the cell, or introduced synthetic machinery, required to initiate the specific transcription of a polynucleotide sequence, thereby allowing the expression of a gene product operably linked to the promoter/regulatory sequence.
  • this sequence may be the core promoter sequence and in other instances, this sequence may also include an enhancer sequence and other regulatory elements which are required for expression of the gene product.
  • the promoter/regulatory sequence may, for example, be one which expresses the gene product in a tissue specific manner.
  • operably linked refers to functional linkage between a regulatory sequence and a heterologous nucleic acid sequence resulting in expression of the latter.
  • a first nucleic acid sequence is operably linked with a second nucleic acid sequence when the first nucleic acid sequence is placed in a functional relationship with the second nucleic acid sequence.
  • a promoter is operably linked to a coding sequence if the promoter affects the transcription or expression of the coding sequence.
  • Operably linked DNA sequences can be contiguous with each other and, e.g., where necessary to join two protein coding regions, are in the same reading frame.
  • transformation means the introduction of a "foreign” (/. ⁇ ., extrinsic or extracellular) gene, DNA or RNA sequence to a host cell, so that the host cell will express the introduced gene or sequence to produce a desired substance, typically a protein or enzyme coded by the introduced gene or sequence.
  • a host cell that receives and expresses introduced DNA or RNA has been "transformed”.
  • expression system means a host cell and compatible vector under suitable conditions, e.g., for the expression of a protein coded for by foreign DNA carried by the vector and introduced to the host cell.
  • the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm, as described below.
  • the percent identity between two amino acid sequences can be determined using the Needleman and Wunsch algorithm (NEEDLEMAN, and Wunsch).
  • the percent identity between two nucleotide or amino acid sequences may also be determined using for example algorithms such as EMBOSS Needle (pair wise alignment; available at www.ebi.ac.uk).
  • EMBOSS Needle may be used with a BLOSUM62 matrix, a “gap open penalty” of 10, a “gap extend penalty” of 0.5, a false “end gap penalty”, an “end gap open penalty” of 10 and an “end gap extend penalty” of 0.5.
  • the “percent identity” is a function of the number of matching positions divided by the number of positions compared and multiplied by 100. For instance, if 6 out of 10 sequence positions are identical between the two compared sequences after alignment, then the identity is 60%. The % identity is typically determined over the whole length of the query sequence on which the analysis is performed. Two molecules having the same primary amino acid sequence or nucleic acid sequence are identical irrespective of any chemical and/or biological modification.
  • a first amino acid sequence having at least 80% of identity with a second amino acid sequence means that the first sequence has 80; 81; 82; 83; 84; 85; 86; 87; 88; 89; 90; 91; 92; 93; 94; 95; 96; 97; 98; 99 or 100% of identity with the second amino acid sequence.
  • conjugate or interchangeably “conjugated polypeptide” is intended to indicate a composite or chimeric molecule formed by the covalent attachment of one or more polypeptides.
  • covalent attachment or “conjugation” means that the polypeptide and the non-peptide moiety are either directly covalently joined to one another, or else are indirectly covalently joined to one another through an intervening moiety or moi eties, such as a bridge, spacer, or linkage moiety or moieties.
  • a particular conjugate is a fusion protein.
  • fusion protein indicates a protein created through the attaching of two or more polypeptides which originated from separate proteins.
  • fusion proteins can be created by recombinant DNA technology and are typically used in biological research or therapeutics. Fusion proteins can also be created through chemical covalent conjugation with or without a linker between the polypeptides portion of the fusion proteins. In the fusion protein the two or more polypeptide are fused directly or via a linker.
  • heterologous polypeptide refers to a polypeptide which does not derive from the same protein to which said heterologous polypeptide is fused.
  • the term "directly" means that the first amino acid at the N-terminal end of a first polypeptide is fused to the last amino acid at the C-terminal end of a second polypeptide. This direct fusion can occur naturally as described in (Vigneron et al., Science 2004, PMID 15001714), (Warren et al., Science 2006, PMID 16960008), (Berkers et al., J. Immunol. 2015a, PMID 26401000), (Berkers et al., J. Immunol.
  • the term “linker” or “spacer” has its general meaning in the art and refers to an amino acid sequence of a length sufficient to ensure that the proteins form proper secondary and tertiary structures.
  • the linker is a peptidic linker which comprises at least one, but less than 30 amino acids e.g., a peptidic linker of 2-30 amino acids, preferably of 10-30 amino acids, more preferably of 15-30 amino acids, still more preferably of 19-27 amino acids, most preferably of 20-26 amino acids.
  • the linker has 2; 3; 4; 5; 6; 7; 8; 9; 10; 11; 12; 13; 14; 15; 16; 17; 18; 19; 20; 21; 22; 23; 24; 25; 26; 27; 28; 29; 30 amino acid residues.
  • linkers are those which allow the compound to adopt a proper conformation (i.e., a conformation allowing a proper signal transducing activity through the IL-15Rbeta/gamma signalling pathway).
  • the most suitable linker sequences (1) will adopt a flexible extended conformation, (2) will not exhibit a propensity for developing ordered secondary structure which could interact with the functional domains of fusion proteins, and (3) will have minimal hydrophobic or charged character which could promote interaction with the functional protein domains.
  • tumor antigen includes both tumor specific antigen (TSA) and tumor associated antigen (TAA).
  • TSA tumor specific antigen
  • TAA tumor associated antigen
  • a tumor specific antigen is known as an antigen that is expressed only by tumor cells while tumor associated antigen are expressed on tumor cells but may also be expressed on some normal cells.
  • mesothelin has its general meaning in the art and refers to the polypeptide encoded by the MSLN gene.
  • An exemplary amino acid sequence for mesothelin is as set forth in SEQ ID NO:2
  • the signal peptide of mesothelin consists of the of the amino acid sequence that ranges from the amino acid residue at position 1 to the amino acid at position 36 in SEQ ID NO:2
  • the Megakaryocyte-potentiating factor (MPF) typically consists of the amino acid sequence that ranges from the amino acid residue at position 37 to the amino acid at position 286 in SEQ ID NO:2.
  • SEQ ID NO : 2 >sp
  • the Megakaryocyte-potentiating factor is underlined in the sequence .
  • an oncolytic virus refers to a virus capable of selectively replicating in dividing cells (e.g. a proliferative cell such as a cancer cell) with the aim of slowing the growth and/or lysing said dividing cell, either in vitro or in vivo, while showing no or minimal replication in non-dividing cells.
  • an oncolytic virus contains a viral genome packaged into a viral particle (or virion) and is infectious (i.e. capable of infecting and entering into a host cell or subject).
  • cancer has its general meaning in the art and includes, but is not limited to, solid tumors and blood borne tumors.
  • the term cancer includes diseases of the skin, tissues, organs, bone, cartilage, blood and vessels.
  • the term “cancer” further encompasses both primary and metastatic cancers. Examples of cancers that may treated by methods and compositions of the invention include, but are not limited to, cancer cells from the bladder, blood, bone, bone marrow, brain, breast, colon, esophagus, gastrointestine, gum, head, kidney, liver, lung, nasopharynx, neck, ovary, prostate, skin, stomach, testis, tongue, or uterus.
  • the cancer may specifically be of the following histological type, though it is not limited to these: neoplasm, malignant; carcinoma; carcinoma, undifferentiated; giant and spindle cell carcinoma; small cell carcinoma; papillary carcinoma; squamous cell carcinoma; lymphoepithelial carcinoma; basal cell carcinoma; pilomatrix carcinoma; transitional cell carcinoma; papillary transitional cell carcinoma; adenocarcinoma; gastrinoma, malignant; cholangiocarcinoma; hepatocellular carcinoma; combined hepatocellular carcinoma and cholangiocarcinoma; trabecular adenocarcinoma; adenoid cystic carcinoma; adenocarcinoma in adenomatous polyp; adenocarcinoma, familial polyposis coli; solid carcinoma; carcinoid tumor, malignant; branchiolo-alveolar adenocarcinoma; papillary adenocarcinoma; chromophobe carcinoma; acid
  • treatment refers to both prophylactic or preventive treatment as well as curative or disease modifying treatment, including treatment of patient at risk of contracting the disease or suspected to have contracted the disease as well as patients who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse.
  • the treatment may be administered to a patient having a medical disorder or who ultimately may acquire the disorder, in order to prevent, cure, delay the onset of, reduce the severity of, or ameliorate one or more symptoms of a disorder or recurring disorder, or in order to prolong the survival of a patient beyond that expected in the absence of such treatment.
  • therapeutic regimen is meant the pattern of treatment of an illness, e.g., the pattern of dosing used during therapy.
  • a therapeutic regimen may include an induction regimen and a maintenance regimen.
  • the phrase “induction regimen” or “induction period” refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the initial treatment of a disease.
  • the general goal of an induction regimen is to provide a high level of drug to a patient during the initial period of a treatment regimen.
  • An induction regimen may employ (in part or in whole) a "loading regimen", which may include administering a greater dose of the drug than a physician would employ during a maintenance regimen, administering a drug more frequently than a physician would administer the drug during a maintenance regimen, or both.
  • maintenance regimen refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the maintenance of a patient during treatment of an illness, e.g., to keep the patient in remission for long periods of time (months or years).
  • a maintenance regimen may employ continuous therapy (e.g., administering a drug at a regular interval, e.g., weekly, monthly, yearly, etc.) or intermittent therapy (e.g., interrupted treatment, intermittent treatment, treatment at relapse, or treatment upon achievement of a particular predetermined criteria [e.g., pain, disease manifestation, etc.]).
  • composition refers to a composition described herein, or pharmaceutically acceptable salts thereof, with other agents such as carriers and/or excipients.
  • the pharmaceutical compositions as provided herewith typically include a pharmaceutically acceptable carrier.
  • the term “pharmaceutically acceptable carrier” includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • Remington's Pharmaceutical-Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses various carriers used in formulating pharmaceutical compositions and known techniques for the preparation thereof.
  • the expression "therapeutically effective amount” is meant a sufficient amount of the active ingredient of the present invention to induce an immune response at a reasonable benefit/risk ratio applicable to the medical treatment.
  • the first object of the present invention relates to the peptide that derives from the ZIKV M oligopeptide and having the amino acid sequence as set forth in SEQ ID NO:1 (RVENWIFRNPG).
  • the present invention relates to a peptide that derives from the ZIKV M oligopeptide and comprising the amino acid sequence as set forth in SEQ ID NO:1 (RVENWIFRNPG).
  • the present invention relates to a peptide that derives from the ZIKV M oligopeptide consisting in SEQ ID NO:1 (RVENWIFRNPG).
  • the present invention relates to a conjugate wherein a heterologous polypeptide is conjugated or fused to the peptide of the present invention.
  • the heterologous polypeptide is conjugated to the peptide of the present invention of the present invention by using chemical coupling.
  • chemical coupling Several methods are known in the art for the attachment or conjugation of an antibody to its conjugate moiety. Examples of linker types that have been used to conjugate a moiety to an antibody include, but are not limited to, hydrazones, thioethers, esters, disulfides and peptide-containing linkers, such as valine- citruline linker.
  • a linker can be chosen that is, for example, susceptible to cleavage by low pH within the lysosomal compartment or susceptible to cleavage by proteases, such as proteases preferentially expressed in tumor tissue such as cathepsins (e.g., cathepsins B, C, D).
  • proteases such as proteases preferentially expressed in tumor tissue such as cathepsins (e.g., cathepsins B, C, D).
  • Techniques for conjugating polypeptides and in particular, are well-known in the art (See, e.g., Amon et al., “Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy,” in Monoclonal Antibodies And Cancer Therapy (Reisfeld et al. eds., Alan R.
  • the heterologous polypeptide is fused to the peptide of the present invention to form a fusion protein.
  • the heterologous polypeptide can be fused to the N-terminus or C- terminus of the peptide of the present invention.
  • the heterologous polypeptide is a tumor antigen. Tumor specific antigens and tumor associated antigens have been described in the art.
  • Such tumor antigen can be, but is not limited to human epithelial cell mucin (Muc-1; a 20 amino acid core repeat for Muc-1 glycoprotein, present on breast cancer cells and pancreatic cancer cells), the Ha-ras oncogene product, p53, carcino-embryonic antigen (CEA), the raf oncogene product, GD2, GD3, GM2, TF, sTn, MAGE-1, MAGE-3, tyrosinase, gp75, Melan-A/Mart-1, gplOO, HER2/neu, EBV-LMP 1 & 2, HPV-F4, 6, 7, prostatic serum antigen (PSA), alpha-fetoprotein (AFP), CO17-1A, GA733, gp72, p53, the ras oncogene product, proteinase 3, Wilm's tumor antigen- 1, telomerase, HPV E7 and melanoma gangliosides, as well as
  • CEA Carcinoembryonic antigen
  • AFP alpha-fetoprotein
  • CEA levels rise in patients with hepatocellular carcinoma: 69% of patients with liver cancer express high levels of AFP in their serum.
  • CEA is a serum glycoprotein of 200 kDa found in adenocarcinoma of colon, as well as cancers of the lung and genitourinary tract.
  • MICA/B ligands of NKG2D are examples of NKG2D. These molecules are expressed on many types of tumors, but not normally on healthy cells.
  • tumor antigens include epithelial cell adhesion molecule (Ep- CAM/TACSTD1), mesothelin, tumor-associated glycoprotein 72 (TAG-72), gplOO, Melan-A, MART-1, KDR, RCAS1, MDA7, cancer-associated viral vaccines (e.g., human papillomavirus antigens), prostate specific antigen (PSA, PSMA), RAGE (renal antigen), CAMEL (CTL- recognized antigen on melanoma), CT antigens (such as MAGE-B5, -B6, -C2, -C3, and D; Mage- 12; CT 10; NY-ESO-1, SSX-2, GAGE, BAGE, MAGE, and SAGE), mucin antigens (e.g., MUC1, mucin-CA125, etc.), cancer-associated ganglioside antigens, tyrosinase, gp75, C- myc, Marti, Melan
  • tumor antigen targets include CA 195 tumor-associated antigen-like antigen (see, e.g., U.S. Pat. No. 5,324,822) and female urine squamous cell carcinoma-like antigens (see, e.g., U.S. Pat. No. 5,306,811), and the breast cell tumor antigens described in U.S. Pat. No. 4,960,716.
  • the heterologous polypeptide is mesothelin and more particularly the megakaryocyte-potentiating factor (MPF, 32-kDa) that derives from mesothelin.
  • the heterologous polypeptide consists of the amino acid sequence that has at least 80% of identity with the amino acid sequence that ranges from the amino acid residue at position 37 to the amino acid at position 286 in SEQ ID NO:2.
  • the peptide of the present invention is fused either directly or via a linker to the heterologous polypeptide.
  • the term "directly” means that the (first or last) amino acid at the terminal end (N or C-terminal end) of the peptide of the present invention is fused to the (first or last) amino acid at the terminal end (N or C-terminal end) of heterologous polypeptide. This direct fusion can occur naturally as described in (Vigneron et al., Science 2004, PMID 15001714), (Warren et al., Science 2006, PMID 16960008), (Berkers et al., J. Immunol.
  • the linker is a peptidic linker which comprises at least one, but less than 30 amino acids e.g., a peptidic linker of 2-30 amino acids, preferably of 10-30 amino acids, more preferably of 15-30 amino acids, still more preferably of 19-27 amino acids, most preferably of 20-26 amino acids.
  • Typical surface amino acids in flexible protein regions include Gly, Asn and Ser (i.e., G, N or S). Virtually any permutation of amino acid sequences containing Gly, Asn and Ser would be expected to satisfy the above criteria for a linker sequence.
  • Other near neutral amino acids such as Thr, Ala, Leu, Gin (i.e., T, A, L, Q) may also be used in the linker sequence.
  • the length of the linker sequence may vary without significantly affecting the biological activity of the fusion protein. If used for therapeutical purposes, the linker is preferably non-immunogenic. Exemplary linker sequences are described in U.S. Pat. Nos. 5,073,627 and 5,108,910. Further preferred examples of linker sequences are Gly/Ser linkers of different length including (gly4ser)3, (gly4ser)4, (gly4ser), (gly3ser), gly3, and (gly3ser2)3.
  • the linker is a glycine-serine linker having the amino acid sequence as set forth in SEQ ID NO:3 (GGGSGGG).
  • the fusion protein of the present invention comprises the amino acid sequence as set forth in SEQ ID NO: 4.
  • the fusion protein of the present invention comprises a signal peptide, preferably at its N-terminal end.
  • Signal peptides are well known in the art.
  • the signal peptide consists of the signal peptide of mesothelin.
  • the fusion protein of the present invention consists of the amino acid sequence as set forth in SEQ ID NO: 5
  • the peptide of the present invention as well as fusion proteins comprising thereof may be produced by any technique known per se in the art, such as, without limitation, any chemical, biological, genetic or enzymatic technique, either alone or in combination. Knowing the amino acid sequence of the desired sequence, one skilled in the art can readily produce said polypeptides, by standard techniques for production of polypeptides. For instance, they can be synthesized using well-known solid phase method, preferably using a commercially available peptide synthesis apparatus (such as that made by Applied Biosystems, Foster City, California) and following the manufacturer’s instructions. Alternatively, the polypeptides and fusions proteins of the invention can be synthesized by recombinant DNA techniques as is now well- known in the art.
  • these fragments can be obtained as DNA expression products after incorporation of DNA sequences encoding the desired (poly) peptide into expression vectors and introduction of such vectors into suitable eukaryotic or prokaryotic hosts that will express the desired polypeptide, from which they can be later isolated using well-known techniques.
  • a further object of the present invention relates to a polynucleotide that encodes for the peptide and the conjugates including the fusion proteins of the present invention.
  • said polynucleotide is a DNA or RNA molecule, which may be included in any suitable vector, such as a plasmid, cosmid, episome, artificial chromosome, phage or a viral vector.
  • the polynucleotide of the present invention can be formulated with cationic polymers including cationic liposomes.
  • Other liposomes also represent effective means to formulate and deliver self-acid nucleic molecule.
  • the DNA can be incorporated into a viral vector, viral particle, or bacterium for pharmacologic delivery.
  • Viral vectors can be infection competent, attenuated (with mutations that reduce capacity to induce disease), or replicationdeficient.
  • Methods utilizing DNA to prevent the deposition, accumulation, or activity of pathogenic self-proteins may be enhanced by use of viral vectors or other delivery systems that increase humoral responses against the encoded autoantigen.
  • the DNA can be conjugated to solid supports including gold particles, polysaccharide-based supports, or other particles or beads that can be injected, inhaled, or delivered by particle bombardment (ballistic delivery).
  • solid supports including gold particles, polysaccharide-based supports, or other particles or beads that can be injected, inhaled, or delivered by particle bombardment (ballistic delivery).
  • particle bombardment ballistic delivery.
  • Methods for delivering nucleic acid preparations are known in the art. See, for example, U.S. Patent Nos. 5,399,346, 5,580,859, and 5,589,466.
  • a number of viral based systems have been developed for transfer into mammalian cells. For example, retroviral systems have been described (U.S. Patent No.
  • Adeno-associated virus (AAV) vector systems have also been developed for nucleic acid delivery.
  • AAV vectors can be readily constructed using techniques well known in the art. See, e.g., U.S. Patent Nos. 5,173,414 and 5,139,941; International Publication Nos.
  • the vector is an oncolytic virus.
  • the oncolytic virus of the present invention can be obtained from any member of virus identified at present time provided that it is oncolytic by its propensity to selectivity replicate and kill dividing cells as compared to nondividing cells. It may be a native virus that is naturally oncolytic or may be engineered by modifying one or more viral genes so as to increase tumor selectivity and/or preferential replication in dividing cells, such as those involved in DNA replication, nucleic acid metabolism, host tropism, surface attachment, virulence, lysis and spread (see for example Kirn et al., 2001, Nat. Med. 7: 781; Wong et al., 2010, Viruses 2: 78-106).
  • exemplary oncolytic viruses include without limitation reovirus, Seneca Valley virus (SVV), vesicular stomatitis virus (VSV), Newcastle disease virus (NDV), herpes simplex virus (HSV), morbillivirus virus, retrovirus, influenza virus, Sin bis virus, poxvirus, adenovirus, or the like.
  • the oncolytic virus of the present invention is obtained from a reovirus.
  • a representative example includes Reolysin (under development by Oncolytics Biotech; NCT01166542).
  • the oncolytic virus of the present invention is obtained from a Seneca Valley virus.
  • a representative example includes NTX-010 (Rudin et al., 2011, Clin. Cancer. Res. 17(4): 888-95).
  • the oncolytic virus of the present invention is obtained from a vesicular stomatitis virus (VSV).
  • VSV vesicular stomatitis virus
  • Representative examples are described in the literature (e.g. Stojdl et al., 2000, Nat. Med. 6(7): 821-5; Stojdl et al., 2003, Cancer Cell 4(4): 263-75).
  • the oncolytic virus of the present invention is obtained from a Newcastle disease virus.
  • Representative examples include without limitation the 73-T PV701 and HDV- HUJ strains as well as those described in the literature (e.g. Phuangsab et al., 2001, Cancer Lett. 172(1): 27-36; Lorence et al., 2007, Curr. Cancer Drug Targets 7(2): 157-67; Freeman et al., 2006, Mol. Ther. 13(1): 221-8).
  • the oncolytic virus of the present invention is obtained from a herpes virus.
  • the Herpesviridae are a large family of DNA viruses that all share a common structure and are composed of relatively large double-stranded, linear DNA genomes encoding 100-200 genes encapsided within an icosahedral capsid which is enveloped in a lipid bilayer membrane.
  • the oncolytic herpes virus can be derived from different types of HSV, particularly preferred are HSV1 and HSV2.
  • the herpes virus may be genetically modified so as to restrict viral replication in tumors or reduce its cytotoxicity in non-dividing cells.
  • any viral gene involved in nucleic acid metabolism may be inactivated, such as thymidine kinase (Martuza et al., 1991, Science 252: 854-6), ribonucleotide reductase (RR) (Boviatsis et al., Gene Ther. 1 : 323-31; Mineta et al., 1994, Cancer Res. 54: 3363-66), or uracil-N-glycosylase (Pyles et al., 1994, J. Virol. 68: 4963-72).
  • RR ribonucleotide reductase
  • RR ribonucleotide reductase
  • uracil-N-glycosylase Proles et al., 1994, J. Virol. 68: 4963-72
  • Another aspect involves viral mutants with defects in the function of genes encoding virulence factors such as the ICP34.5 gene (Chambers et al.
  • oncolytic herpes virus examples include NV1020 (e.g. Geevarghese et al., 2010, Hum. Gene Ther. 21(9): 1119-28) and T-VEC (Andtbacka et al., 2013, J. Clin. Oncol. 31, abstract number LBA9008).
  • the oncolytic virus of the present invention is obtained from a morbillivirus which can be obtained from the paramyxoviridae family, with a specific preference for measles virus.
  • oncolytic measles viruses include without limitation MV-Edm (McDonald et al., 2006; Breast Cancer Treat. 99(2): 177-84) and HMWMAA (Kaufmann et al., 2013, J. Invest. Dermatol. 133(4): 1034-42)
  • the oncolytic virus of the present invention is obtained from an adenovirus.
  • Methods are available in the art to engineer oncolytic adenoviruses.
  • An advantageous strategy includes the replacement of viral promoters with tumor-selective promoters or modifications of the El adenoviral gene product(s) to inactivate its/their binding function with p53 or retinoblastoma (Rb) protein that are altered in tumor cells.
  • the adenovirus E1B55 kDa gene cooperates with another adenoviral product to inactivate p53 (p53 is frequently dysregulated in cancer cells), thus preventing apoptosis.
  • oncolytic adenovirus examples include ONYX-015 (e.g. Khuri et al., 2000, Nat. Med 6(8): 879-85) and H101 also named Oncorine (Xia et al., 2004, Ai Zheng 23(12): 1666-70).
  • the oncolytic virus of the present invention is a poxvirus.
  • poxvirus refers to a virus belonging to the Poxviridae family, with a specific preference for a poxvirus belonging to the Chordopoxviridae subfamily and more preferably to the Orthopoxvirus genus. Sequences of the genome of various poxviruses, for example, the vaccinia virus, cowpox virus, Canarypox virus, Ectromelia virus, Myxoma virus genomes are available in the art and specialized databases such as Genbank (accession number NC 006998, NC_003663, NC_005309, NC_004105, NC_001132 respectively).
  • the oncolytic poxvirus is an oncolytic vaccinia virus.
  • Vaccinia viruses are members of the poxvirus family characterized by a 200 kb double-stranded DNA genome that encodes numerous viral enzymes and factors that enable the virus to replicate independently from the host cell machinery.
  • the majority of vaccinia virus particles is intracellular (IMV for intracellular mature virion) with a single lipid envelop and remains in the cytosol of infected cells until lysis.
  • the other infectious form is a double enveloped particle (EEV for extracellular enveloped virion) that buds out from the infected cell without lysing it.
  • compositions of the present invention are provided.
  • a further of the present invention relates to a pharmaceutical composition that comprises as active ingredient an amount of the peptide, conjugate, fusion protein, polynucleotide or vector of the present invention and a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type.
  • the active principle alone or in combination with another active principle, can be administered in a unit administration form, as a mixture with conventional pharmaceutical supports, to the subjects.
  • Suitable unit administration forms comprise oral-route forms such as tablets, gel capsules, powders, granules and oral suspensions or solutions, sublingual and buccal administration forms, aerosols, implants, subcutaneous, transdermal, topical, intraperitoneal, intramuscular, intravenous, subdermal, transdermal, intrathecal and intranasal administration forms and rectal administration forms.
  • the pharmaceutical compositions contain vehicles, which are pharmaceutically acceptable for a formulation capable of being injected.
  • saline solutions monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts
  • dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists.
  • Solutions comprising compounds of the present invention as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the active ingredient of the present invention can be formulated into a composition in a neutral or salt form.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like.
  • Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine,
  • the carrier can also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetables oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminium monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • sterile powders for the preparation of sterile injectable solutions the preferred methods of preparation are vacuum - drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the preparation of more, or highly concentrated solutions for direct injection is also contemplated, where the use of DMSO as solvent is envisioned to result in extremely rapid penetration, delivering high concentrations of the active agents to a small tumor area.
  • solutions Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • the formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like can also be employed.
  • aqueous solutions For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
  • sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject.
  • a further object of the present invention relates to the peptide, conjugate, fusion protein, polynucleotide or vector of the present invention for use as a drug.
  • a further object of the present invention relates to a method of treating cancer in a patient in need thereof comprising administering to the patient a therapeutically effective amount of the peptide, conjugate, fusion protein, polynucleotide or vector of the present invention.
  • the peptide, conjugate, fusion protein, polynucleotide or vector of the present invention is used in combination with a chemotherapeutic agent.
  • chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaorarnide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a carnptothecin (including the synthetic analogue topotecan); bryostatin; cally statin; CC-1065 (including its adozelesin,
  • calicheamicin especially calicheamicin (11 and calicheamicin 211, see, e.g., Agnew Chem Inti. Ed. Engl. 33: 183-186 (1994); dynemicin, including dynemicin A; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, canninomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolin
  • paclitaxel (TAXOL®, Bristol-Myers Squibb Oncology, Princeton, N.].) and doxetaxel (TAXOTERE®, Rhone-Poulenc Rorer, Antony, France); chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP- 16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; CPT-1 1 ; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoic acid; capecitabine; and phannaceutically acceptable salts, acids or derivatives of any of the above.
  • antihormonal agents that act to regulate or inhibit honnone action on tumors
  • anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and phannaceutically acceptable salts, acids or derivatives of any of the above.
  • the peptide, conjugate, fusion protein, polynucleotide or vector of the present invention in combination with a targeted cancer therapy.
  • Targeted cancer therapies are drugs or other substances that block the growth and spread of cancer by interfering with specific molecules ("molecular targets") that are involved in the growth, progression, and spread of cancer.
  • Targeted cancer therapies are sometimes called “molecularly targeted drugs,” “molecularly targeted therapies,” “precision medicines,” or similar names.
  • the targeted therapy consists of administering the subject with a tyrosine kinase inhibitor.
  • tyrosine kinase inhibitor refers to any of a variety of therapeutic agents or drugs that act as selective or non-selective inhibitors of receptor and/or non-receptor tyrosine kinases. Tyrosine kinase inhibitors and related compounds are well known in the art and described in U.S Patent Publication 2007/0254295, which is incorporated by reference herein in its entirety.
  • a compound related to a tyrosine kinase inhibitor will recapitulate the effect of the tyrosine kinase inhibitor, e.g., the related compound will act on a different member of the tyrosine kinase signaling pathway to produce the same effect as would a tyrosine kinase inhibitor of that tyrosine kinase.
  • tyrosine kinase inhibitors and related compounds suitable for use in methods of embodiments of the present invention include, but are not limited to, dasatinib (BMS-354825), PP2, BEZ235, saracatinib, gefitinib (Iressa), sunitinib (Sutent; SU11248), erlotinib (Tarceva; OSI-1774), lapatinib (GW572016; GW2016), canertinib (CI 1033), semaxinib (SU5416), vatalanib (PTK787/ZK222584), sorafenib (BAY 43-9006), imatinib (Gleevec; STI571), leflunomide (SU101), vandetanib (Zactima; ZD6474), MK-2206 (8-[4-aminocyclobutyl)phenyl]-9-phenyl- l,2,4-triazolo[3,4
  • the tyrosine kinase inhibitor is a small molecule kinase inhibitor that has been orally administered and that has been the subject of at least one Phase I clinical trial, more preferably at least one Phase II clinical, even more preferably at least one Phase III clinical trial, and most preferably approved by the FDA for at least one hematological or oncological indication.
  • inhibitors include, but are not limited to, Gefitinib, Erlotinib, Lapatinib, Canertinib, BMS-599626 (AC-480), Neratinib, KRN-633, CEP-11981, Imatinib, Nilotinib, Dasatinib, AZM-475271, CP-724714, TAK-165, Sunitinib, Vatalanib, CP- 547632, Vandetanib, Bosutinib, Lestaurtinib, Tandutinib, Midostaurin, Enzastaurin, AEE-788, Pazopanib, Axitinib, Motasenib, OSI-930, Cediranib, KRN-951, Dovitinib, Seliciclib, SNS- 032, PD-0332991, MKC-I (Ro-317453; R-440), Sorafenib, ABT-869
  • the peptide, conjugate, fusion protein, polynucleotide or vector of the present invention is administered to the patient in combination with an immune checkpoint inhibitor.
  • Preferred immune checkpoint inhibitors are antibodies that specifically recognize immune checkpoint proteins.
  • a number of immune checkpoint inhibitors are known and in analogy of these known immune checkpoint protein inhibitors, alternative immune checkpoint inhibitors may be developed in the (near) future.
  • the immune checkpoint inhibitors include peptides, antibodies, nucleic acid molecules and small molecules.
  • immune checkpoint inhibitor examples include PD-1 antagonist, PD-L1 antagonist, PD-L2 antagonist CTLA-4 antagonist, VISTA antagonist, TIM-3 antagonist, LAG-3 antagonist, IDO antagonist, KIR2D antagonist, A2AR antagonist, B7-H3 antagonist, B7-H4 antagonist, and BTLA antagonist.
  • PD-1 (Programmed Death-1) axis antagonists include PD-1 antagonist (for example anti-PD-1 antibody), PD-L1 (Programmed Death Ligand-1) antagonist (for example anti-PD-Ll antibody) and PD-L2 (Programmed Death Ligand-2) antagonist (for example anti- PD-L2 antibody).
  • the anti-PD-1 antibody is selected from the group consisting of MDX-1106 (also known as Nivolumab, MDX-1106-04, ONO-4538, BMS- 936558, and Opdivo®), Merck 3475 (also known as Pembrolizumab, MK-3475, Lambrolizumab, Keytruda®, and SCH-900475), and CT-011 (also known as Pidilizumab, hBAT, and hBAT-1).
  • the PD-1 binding antagonist is AMP -224 (also known as B7-DCIg).
  • the anti-PD-Ll antibody is selected from the group consisting of YW243.55.S70, MPDL3280A, MDX-1105, and MEDI4736.
  • MDX-1105 also known as BMS-936559, is an anti-PD-Ll antibody described in W02007/005874.
  • Antibody YW243.55. S70 is an anti-PD-Ll described in WO 2010/077634
  • AL MEDI4736 is an anti-PD- Ll antibody described in WO2011/066389 and US2013/034559.
  • MDX-1106 also known as MDX-1 106-04, ONO-4538 or BMS-936558, is an anti-PD-1 antibody described in U.S. Pat. No.
  • Merck 3745 also known as MK-3475 or SCH-900475, is an anti-PD-1 antibody described in U.S. Pat. No. 8,345,509 and W02009/114335.
  • CT-011 Panizilumab
  • AMP-224 also known as B7-DCIg, is a PD-L2-Fc fusion soluble receptor described in W02010/027827 and WO2011/066342.
  • Atezolimumab is an anti-PD-Ll antibody described in U.S. Pat. No. 8,217,149.
  • Avelumab is an anti-PD-Ll antibody described in US 20140341917.
  • CA-170 is a PD-1 antagonist described in W02015033301 & WO2015033299.
  • Other anti-PD-1 antibodies are disclosed in U.S. Pat. No. 8,609,089, US 2010028330, and/or US 20120114649.
  • the PD-1 inhibitor is an anti-PD-1 antibody chosen from Nivolumab, Pembrolizumab or Pidilizumab.
  • PD-L1 antagonist is selected from the group comprising of Avelumab, BMS-936559, CA-170, Durvalumab, MCLA-145, SP142, STI-A1011, STIA1012, STI-A1010, STI-A1014, Al 10, KY1003 and Atezolimumab and the preferred one is Avelumab, Durvalumab or Atezolimumab.
  • FIGURES are a diagrammatic representation of FIGURES.
  • FIG. 1 Schematic representation of the GFP constructs assembled with flavivirus M oligopeptides.
  • (A) schematic representation of mature prM protein that is structured into a “pr” polypeptide followed by a 40 residues-long M ectodomain (ectoM) and the transmembrane anchoring region (TMDs).
  • ectoM M ectodomain
  • TMDs transmembrane anchoring region
  • the sequences of ectoM from epidemic Brazilian ZIKV strain BeH819015 (NC 009227208), epidemic Indian ocean DENV-2 strain RUJul (MN272404) and vaccine strain 17D of YFV (NC_776001.1) are listed. The last eleven residues of ectoM are underlined.
  • the GFP constructs include the signal peptide (SP) of ZIKV prM followed by a FLAG epitope at the N-terminus.
  • the soluble sGFP constructs are ended by a glycine-serine spacer followed by the residues M-31/41 of ZIKV, DENV-2 and YFV or without viral sequence.
  • the GFP ZIKV M31/41 construct is lacking of prM signal peptide.
  • the sGFP construct is limited to a glycine-serine spacer at its C-terminus.
  • the SGFP ZIKV M31/41AAA is an Ala-mutant of sGFP ZIKV M31/41 cons t ruc t (hereafter called mutGFP ZIKV M31/41 ) bearing the three Ala substitutions at positions M-33/35/38 which are underlined.
  • FIG. 1 The ZIKV M oligopeptide associated to sGFP has an effect on cell integrity.
  • Cells were transfected 24 h (left) or 48 h (right) with plasmids expressing sGFP, QFpZIKV.M-31/41, QFpDENV-2.M-31/41 an J QppYFV.M-31/41 Qr moc k-transfeCted (control).
  • LDH activity was measured and cell membrane permeability was expressed as signal intensity (O.D.).
  • the results are the mean ( ⁇ SEM) of three or six independent experiments respectively. Pairwise comparisons with sGFP were performed and all significant comparisons are noted (** p ⁇ 10' 2 ; * p ⁇ 10' 1 ); any non- statistically significant comparisons are omitted.
  • ZIKV M oligopeptide has ability to induce caspase-3 activation in Huh7 cells.
  • Cells were transfected 24 h with plasmids coding for sGFP ZIKV M ' 31/41 , SGFP DENV ' 2,M ' 31/41 and sGFP YFV - M ' 31/41 or mock-transfected (control).
  • Plasmid expressing sGFP served as a control.
  • A cell metabolic activity was measured using MTT assay. Viability was expressed as the signal intensity (O.D.).
  • caspase 3/7 activity was expressed as the fold change of caspase activity in assay relative to that in sGFP. The results are the mean ( ⁇ SEM) of six independent assays. Pairwise comparisons with sGFP were performed and all statistically significant comparisons are noted
  • FIG. 4 The pro-apoptotic activity of ZIKV M oligopeptide involves the residues M-33/35/38 and requires its trafficking into the secretory pathway.
  • Huh7 cells were transfected 24 h with plasmids expressing sGFP, sGFP ZIKV M ' 31/41 , the GFP ZIKV M ' 31/41 mutant that lacks of signal peptide, or the mutGFP ZIKV M ' 31/41 mutant bearing the three Ala mutations.
  • Caspase 3/7 activity is expressed as the fold change of caspase activity in assay relative to that in sGFP.
  • the results are the mean ( ⁇ SEM) of three independent experiments. Pairwise comparisons with sGFP or sGFP ZIKV M ' 31/41 were performed and all statistically significant comparisons are noted
  • FIG. 5 Schematic representation of MPF constructs with ZAMP.
  • MSLN preprotein mesothelin
  • MPF membrane-anchored MSLN by furin protease.
  • the signal peptide of MSLN (residues 1/37) is indicated as blue box.
  • the mutant MPF-mutZAMP includes the three mutations Ala inserted into the ZAMP sequence. The amino-acid changes are underlined in the ZAMP sequence.
  • FIG. 6 ZAMP conjugated to MPF has no effect on HEK-293T cells.
  • Cells were transfected 24 h with plasmids encoding MPF-FLAG, MPF-ZAMP, MPF-mutZAMP, or mock- transfected (control).
  • A FACS analysis was performed on cells expressing MPF-FLAG using anti-FLAG antibody. The percentage and the mean of fluorescence intensity (MFI) of positive cells are shown.
  • MFI fluorescence intensity
  • B cell metabolic activity was measured at 48 h post-transfection using an MTT assay. Viability was expressed as signal intensity (O.D.) and the results are the mean ( ⁇ SEM) of four independent assays. Non-statistically differences were observed between the MPF constructs and control.
  • FIG. 7 Expression of MPF-ZAMP induces apoptosis in A549 cells.
  • Cells were transfected 24 h with plasmids expressing MPF-FLAG, MPF-ZAMP, MPF-mutZAMP or transfectant alone (vehicle) or mock-transfected (control).
  • A549 cells were transfected with plasmids expressing MPF-ZAMP or MPF-mutZAMP. Cells in apoptotic state were quantified by staining with FITC-labeled Annexin V. The percentage of Annexin V-positive cells was determined by FACS analysis. The results are the mean ( ⁇ SEM) of two independent assays.
  • B cell metabolic activity was measured using an MTT assay.
  • Viability was expressed as signal intensity (O.D.) and the results are the mean ( ⁇ SEM) of three independent assays. Pairwise comparisons with MPF-FLAG were performed and all statistically comparisons are noted (* p ⁇ 10' 1 ); any non-statistically significant comparisons are omitted. In (C), caspase 3/7 activity was measured as signal intensity (O.D). The results are the mean ( ⁇ SEM) of four independent assays. Pairwise comparisons with MPF-FLAG were performed and all statistically comparisons are noted (** p ⁇ 10' 2 ; * p ⁇ 10' 1 ); any non-statistically significant comparisons are omitted.
  • FIG. 8 Expression of MPF-ZAMP induces caspase-3 activation in Huh7 cells.
  • Cells were transfected with plasmids encoding MPF-FLAG, MPF-ZAMP, MPF-mutZAMP or transfectant alone (vehicle) or mock-transfected (control).
  • A cell metabolic activity was measured at 18 h post-transfection using an MTT assay. Viability was expressed as signal intensity (O.D.) and the results are the mean ( ⁇ SEM) of four independent assays.
  • Caspase 3/7 activity was measured at 18 h post-transfection and was expressed as signal intensity (O.D). The results are the mean ( ⁇ SEM) of six independent assays. Pairwise comparisons with MPF-FLAG were performed and all statistically comparisons are noted (** p ⁇ 10' 4 ; * p ⁇ 10' 2 ); any non- statistically significant comparisons are omitted.
  • Huh7 cells from Nolwenn Jouvenet, Institut Pasteur
  • A549 cells ATCC, CCL-185
  • HEK- 293T cells ATCC, CRL-1573
  • MEM Minimum Essential Media
  • FBS heat-inactivated fetal bovine serum
  • the coding sequence for M31 ' 41 from Brazilian BeH819015 strain preceded in frame by the soluble MPF region of human mesothelin (Genbank access number NM_001177355.1) which contains the signal peptide for the transport of MPF-ZAMP into secretory pathway (Fig. 5) were synthesized and cloned into Nhe I and Not I restriction sites of the pcDNA 3.1 (+) Neo plasmid (Genecust, Boynes, France).
  • Plasmids were transfected in Huh7, A549 and HEK-293T cells using LipofectaminTM 3000 according to the manufacturer’s instructions (Thermo Fischer Scientific, Illkirch-Grafenstaden, France).
  • Huh7 cells 1.5 x 10 5
  • A549 cells (1 xlO 5 ) were fixed with 3.7% paraformaldehyde (PF A) in phosphate buffered saline (PBS) for 10 min and GFP expression was analyzed by flow cytometry using FlowJo software (BD Bioscience).
  • PF A paraformaldehyde
  • PBS phosphate buffered saline
  • Huh7 cells were seeded in 12-well culture plate at a density of 1.5 x 10 5 cells per well. Cell damages were evaluated on supernatants of transfected-cells measuring lactate dehydogenase (LDH) release using Cytotox 96® non-radioactive cytotoxicity assay (Promega) according to manufacturer instructions. Absorbance of converted dye was measured at 490 nm (Tecan) with a background subtraction at 690 nm.
  • LDH lactate dehydogenase
  • Huh7 cells (2 x 10 4 ), A549 cells (2 x 10 4 ) or HEK-293T cells (2 x 10 4 ) were cultured in 96-well culture plate.
  • Cell monolayers were rinsed with PBS IX and incubated with culture growth medium mixed with 5 mg.mL' 1 MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) solution for 1 h at 37°C.
  • MTT medium was removed and the formazan crystals were solubilized with dimethyl sulfoxide (DMSO).
  • Absorbance was measured at 570 nm with a background subtraction at 690 nm.
  • Huh7 cells (2 x 10 4 ) or A549 (2 x 10 4 ) were seeded in 96-well culture plate.
  • Caspase 3/7 activity in raw cell lysates was measured using a Caspase Gio® 3/7 assay kit (Promega, Charbonnieres- les-bains, France) according to the manufacturer’s protocol.
  • Caspase activity was quantified by luminescence using a FLUOstar Omega Microplate Reader (BMG LABTECH, Champigny- sur-Marne, France).
  • A549 cells were seeded in 12-well culture plate at a density of 1 x 10 5 cells per well. Cells were fixed with 3.7% PFA in PBS for 10 min and stained with the Ca 2+ -dependent phosphatidylserine-binding protein FITC-labelled annexin V (BioLegend). FITC expression was analyzed by flow cytometry using Flowjo software (BD Bioscience).
  • GFP -based constructs A same design of GFP -based constructs was applied for DENV-2 and YFV M oligopeptides to serve as positive controls.
  • a sGFP with only the glycine-serine spacer at the C-terminus was used as a negative control (Fig. IB).
  • Huh7 cells were transfected with vector plasmids expressing either GFP ZIKVM ' 31/41 , GFP DENV ’ 2 M ’ 31/41 , OR QFP YFV - M ' 31/41 (pjg ⁇ )
  • a recombinant sGFP without the C-terminal viral sequence was used as a negative control.
  • Cell membrane integrity was evaluated by measuring LDH release (Fig. 2). At 24 h posttransfection, no significant change in LDH activity was observed in transfected Huh7 cells whatever recombinant sGFP constructs tested.
  • Apoptosis mediated by DENV ectoM involves translocation of the viral sequence into the secretory pathway [6], To evaluate whether a same pre-requisite does exist for ZIKV M oligopeptide, the sGFP ZIKV M ' 31/41 construct was deleted from its signal peptide to generate a mutant GFP ZIKV M - 31/41 (Fig. IB). As shown in Fig. 4, lack of signal peptide resulted in severe reduction of caspase-3 activity as compared to sGFP ZIKV M ' 31/41 . Thus, induction of apoptosis mediated by sGFP ZIKV M ' 31/41 requires trafficking of ZIKV M oligopeptide into the secretory pathway.
  • the ZIKV M oligopeptide confers pro-apoptotic properties to a soluble tumor-associated antigen
  • the oligopeptide RVEWIFRNPG (hereafter referred to as ZAMP for Zika Apoptosis M Peptide) corresponding to ZIKV M residues M- 31/41 was linked to the C-terminus of MPF.
  • ZAMP Zika Apoptosis M Peptide
  • the authentic signal peptide of MPF was used for translocation of MPF -ZAMP into the secretory pathway.
  • MPF is a biomarker of lung cancers [10]
  • pulmonary adenocarcinoma A549 cells were tested with plasmids expressing MPF-ZAMP proteins.
  • FACS analysis using anti-FLAG antibody showed that expression of MPF-FLAG protein was efficient in A549 cells as observed with HEK-293T cells (not shown).
  • the death-promoting activity of MPF-ZAMP in A549 cells was assessed by measuring the rate of early apoptosis using the Annexin V affinity assay, which detects phosphatidylserine (PS) translocation to the outer layer of the cell membrane.
  • PS phosphatidylserine
  • caspase- 3/7 assay indicated tjt expression of MPF-ZAMP resulted in a higher level of caspase-3 activity as compared to MPF-mutZAMP and MPF-FLAG (Fig. 7C). Together, these results demonstrated that MPF-ZAMP can trigger apoptosis in pulmonary adenocarcinoma cells through caspase-3 activation.
  • MPF-ZAMP has also ability to induce apoptosis in hepatoma cells such as Huh7 cells.
  • MTT assay we observed that expression of MPF-ZAMP results in a marked decrease of cell metabolic activity at 24 h post-transfection. Consequently, the loss of cell metabolism mediated by MPF-ZAMP was examined in Huh7 cells at an earlier time in the transfection.
  • expression of MPF-ZAMP again resulted in a severe decrease in cell metabolism as compared to MPF-FLAG and MPF-mutZAMP (Fig. 8A).
  • Flaviviruses expose on their surface the structural proteins E and M, the latter being cleaved from its glycosylated precursor form prM into immature virus particles during their exocytic transit towards extracellular compartment [11],
  • the mature M protein comprises a 40- residues long ectodomain followed by a double-membrane anchor. Little is still known on the role of small integral membrane M protein in the biology and pathogenicity of flaviviruses.
  • ZIKV can trigger apoptotic cell death in vitro as well as in vivo [18-21],
  • the structural proteins prM and E have been associated with apoptosis mediated by ZIKV [22]
  • the last C-terminal residues M-31/41 of the ZIKV M ectodomain from Brazilian viral strain BeH819015 have death-promoting activity through caspase-3 activation.
  • ZAMP sequence shares at least 60% of identity to the counterpart from DENV-2 but less than 30% with YFV despite the fact that the death-promoting activity of the last C-terminal residues of the M ectodomain is conserved among the three flaviviruses.
  • residues M-33/35/38 as playing a key role in the pro-apoptotic activity of ZAMP.
  • the residue Tryp to position M-35 is strictly conserved among flavivirus M proteins.
  • ZIKV cannot tolerate a complete change of the three residues M-33/35/38, it would be of great interest to assess the contribution of each the three residues in relation with the death-promoting activity of ZIKV.
  • ZAMP -mediated apoptosis involves the transport of the Zika M oligopeptide into the secretory pathway.
  • Sphingosine kinase SPHK2 can generate pro-apoptotic ceramide through the production of sphingosine 1 -phosphate which acts as intracellular signaling molecule.
  • SPHK2 has been shown to interact with DENV proteins leading to apoptotic cell death [28], Whether the subcellular distribution of ZAMP leads to activation of apoptosis signaling pathway through a protein-protein interaction requires further investigation.
  • TAA tumor-associated antigen mesothelin.
  • cancer-testis like antigens such as MAGE-1 are expressed in a wide range of different cancers including melanoma, blade cancer and neuroblastoma[33]
  • Differentiation antigens including tyrosinase TRP-1 are expressed in differentiation stage-dependent and tissue-specific manners [34, 35]
  • Oncofetal antigens including carcinoembryonic antigen (CEA) are found in embryonic and fetal tissues as well as certain cancers [36, 37]
  • Overexpressed antigens such as mesothelin that are normal proteins whose expression is up-regulated in cancer cells [9],
  • ZIKV has gained attention in oncolytic virotherapy field as a potential oncolytic virus due to its neurotropism and its efficacy against glioblastoma multiforme and aggressive metastatic forms of human CNS tumors [38-40], As aforementioned, association of ZAMP with MPF results in activation of apoptosis executioner caspase-3 in different tumor cells.
  • the ability of ZIKV M oligopeptide RVENWIFRNPG to confer pro-apoptotic activity to TAA could open important perspective for ZAMP in cancer therapy strategy through tumor cell death and immune recognition of the TAA by antigen-presenting cells.
  • ApoptoM sequence results in disruption of mitochondrial potential and caspase activation. Biochimie, 2003. 85(8): p. 789- 93.
  • RNAi screen reveals a role of SPHK2 in dengue virus-mediated apoptosis in hepatic cell lines.

Abstract

L'apoptose est le mécanisme principal au moyen duquel l'infection par le virus Zika véhiculée par les moustiques provoque la mort cellulaire. L'objectif des inventeurs est de déterminer si les derniers résidus de l'extrémité C-terminale M31/41 de l'ectodomaine ZIKV peuvent déclencher la mort cellulaire apoptotique. L'apoptose a été détectée dans des cellules humaines d'hépatome Huh7 exprimant une GFP recombinante qui comprend un oligopeptide ZIKV M à son extrémité C-terminale. Le déclenchement de l'apoptose nécessite la translocation de l'oligopeptide ZIKV M dans la voie sécrétoire et implique l'activation de la caspase-3. Les inventeurs ont ensuite évalué si l'oligopeptide ZIKV M a la capacité de conférer une activité favorisant la mort à un antigène secrété associé à une tumeur tel qu'un facteur de potentialisation de mégacaryocytes (MPF) qui est un produit clivé de la mésothéline humaine. L'expression de MPF avec un oligopeptide ZIKV M lié à son extrémité C-terminale conduit à l'induction de l'apoptose dans des cellules A549 d'un adénocarcinome pulmonaire humain ainsi que des cellules humaines d'hépatome Huh 7. Dans la présente invention, le ZIKV a été proposé en tant que virus oncolytique pour une thérapie anticancéreuse, la capacité de la séquence ZIKV RVENWIFRNPG (appelée ZAMP pour le peptide M d'apoptose Zika) (SEQ ID NO : l) pour conférer une activité pro-apoptotique à un antigène associé à une tumeur tel que MPF ouvre ainsi de nouvelles perspectives d'attraction pour ZAMP en tant qu'agent anticancéreux innovant.
PCT/EP2021/081631 2020-11-16 2021-11-15 Utilisation des derniers résidus de l'extrémité c-terminale m31/41 de l'ectodomaine zikv m pour déclencher la mort cellulaire apoptotique WO2022101463A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP20306386.2 2020-11-16
EP20306386 2020-11-16

Publications (1)

Publication Number Publication Date
WO2022101463A1 true WO2022101463A1 (fr) 2022-05-19

Family

ID=73554400

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2021/081631 WO2022101463A1 (fr) 2020-11-16 2021-11-15 Utilisation des derniers résidus de l'extrémité c-terminale m31/41 de l'ectodomaine zikv m pour déclencher la mort cellulaire apoptotique

Country Status (1)

Country Link
WO (1) WO2022101463A1 (fr)

Citations (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989012624A2 (fr) 1988-06-14 1989-12-28 Cetus Corporation Agents de couplage et conjugues lies a des disulfures a empechement sterique prepares a partir de tels agents
US4960716A (en) 1984-05-01 1990-10-02 Ciba Corning Diagnostics Corp. Monoclonal antibodies specific for 330 KD breast tumor antigen and assay using said monoclonal antibodies
US5073627A (en) 1989-08-22 1991-12-17 Immunex Corporation Fusion proteins comprising GM-CSF and IL-3
WO1992001070A1 (fr) 1990-07-09 1992-01-23 The United States Of America, As Represented By The Secretary, U.S. Department Of Commerce Conditionnement a haute efficacite de virus adeno-associe mutant utilisant la suppression d'ambre
US5108910A (en) 1989-08-22 1992-04-28 Immunex Corporation DNA sequences encoding fusion proteins comprising GM-CSF and IL-3
US5139941A (en) 1985-10-31 1992-08-18 University Of Florida Research Foundation, Inc. AAV transduction vectors
US5173414A (en) 1990-10-30 1992-12-22 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors
WO1993003769A1 (fr) 1991-08-20 1993-03-04 THE UNITED STATES OF AMERICA, represented by THE SECRETARY, DEPARTEMENT OF HEALTH AND HUMAN SERVICES Transfert induit par adenovirus de genes vers la voie gastro-intestinale
US5219740A (en) 1987-02-13 1993-06-15 Fred Hutchinson Cancer Research Center Retroviral gene transfer into diploid fibroblasts for gene therapy
US5306811A (en) 1990-11-27 1994-04-26 Ciba Corning Diagnostics Corp. Squamous cell carcinoma-like immunoreactive antigen from human female urine
US5324822A (en) 1991-04-12 1994-06-28 Ciba Corning Diagnostics Corp. Method of isolating a CA 195-like immunoreactive antigen from human amniotic fluid
US5399346A (en) 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
US5580859A (en) 1989-03-21 1996-12-03 Vical Incorporated Delivery of exogenous DNA sequences in a mammal
US5618829A (en) 1993-01-28 1997-04-08 Mitsubishi Chemical Corporation Tyrosine kinase inhibitors and benzoylacrylamide derivatives
US5639757A (en) 1995-05-23 1997-06-17 Pfizer Inc. 4-aminopyrrolo[2,3-d]pyrimidines as tyrosine kinase inhibitors
US5728868A (en) 1993-07-15 1998-03-17 Cancer Research Campaign Technology Limited Prodrugs of protein tyrosine kinase inhibitors
US5804396A (en) 1994-10-12 1998-09-08 Sugen, Inc. Assay for agents active in proliferative disorders
US6100254A (en) 1997-10-10 2000-08-08 Board Of Regents, The University Of Texas System Inhibitors of protein tyrosine kinases
US6127374A (en) 1997-07-29 2000-10-03 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6245759B1 (en) 1999-03-11 2001-06-12 Merck & Co., Inc. Tyrosine kinase inhibitors
US6306874B1 (en) 1999-10-19 2001-10-23 Merck & Co., Inc. Tyrosine kinase inhibitors
US6313138B1 (en) 2000-02-25 2001-11-06 Merck & Co., Inc. Tyrosine kinase inhibitors
US6316444B1 (en) 1999-06-30 2001-11-13 Merck & Co., Inc. SRC kinase inhibitor compounds
US6329380B1 (en) 1999-06-30 2001-12-11 Merck & Co., Inc. SRC kinase inhibitor compounds
US6344459B1 (en) 1996-04-12 2002-02-05 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6420382B2 (en) 2000-02-25 2002-07-16 Merck & Co., Inc. Tyrosine kinase inhibitors
US6479512B1 (en) 1999-10-19 2002-11-12 Merck & Co., Inc. Tyrosine kinase inhibitors
US6498165B1 (en) 1999-06-30 2002-12-24 Merck & Co., Inc. Src kinase inhibitor compounds
US6586423B2 (en) 1999-09-10 2003-07-01 Merck & Co., Inc. Tyrosine kinase inhibitors
US6740665B1 (en) 1999-02-10 2004-05-25 Ramachandran Murali Tyrosine kinase inhibitors and methods of using the same
US6794393B1 (en) 1999-10-19 2004-09-21 Merck & Co., Inc. Tyrosine kinase inhibitors
US6875767B2 (en) 2001-06-22 2005-04-05 Merck & Co., Inc. (5-cyano-2-thiazolyl)amino-4-pyridine tyrosine kinase inhibitors
US6927293B2 (en) 2001-08-30 2005-08-09 Merck & Co., Inc. Tyrosine kinase inhibitors
US6958340B2 (en) 2001-08-01 2005-10-25 Merck & Co., Inc. Tyrosine kinase inhibitors
WO2006121168A1 (fr) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Anticorps monoclonaux humains pour mort programmee 1 (mp-1) et procedes pour traiter le cancer en utilisant des anticorps anti-mp-1 seuls ou associes a d’autres immunotherapies
WO2007005874A2 (fr) 2005-07-01 2007-01-11 Medarex, Inc. Anticorps monoclonaux humains diriges contre un ligand de mort programmee de type 1(pd-l1)
US20070254295A1 (en) 2006-03-17 2007-11-01 Prometheus Laboratories Inc. Methods of predicting and monitoring tyrosine kinase inhibitor therapy
WO2009101611A1 (fr) 2008-02-11 2009-08-20 Curetech Ltd. Anticorps monoclonaux pour le traitement de tumeurs
WO2009114335A2 (fr) 2008-03-12 2009-09-17 Merck & Co., Inc. Protéines de liaison avec pd-1
US20100028330A1 (en) 2002-12-23 2010-02-04 Medimmune Limited Methods of upmodulating adaptive immune response using anti-pd1 antibodies
WO2010027827A2 (fr) 2008-08-25 2010-03-11 Amplimmune, Inc. Polypeptides co-stimulateurs ciblés et leurs procédés d'utilisation dans le traitement du cancer
WO2010077634A1 (fr) 2008-12-09 2010-07-08 Genentech, Inc. Anticorps anti-pd-l1 et leur utilisation pour améliorer la fonction des lymphocytes t
WO2011066389A1 (fr) 2009-11-24 2011-06-03 Medimmmune, Limited Agents de liaison ciblés dirigés contre b7-h1
WO2011066342A2 (fr) 2009-11-24 2011-06-03 Amplimmune, Inc. Inhibition simultanée de pd-l1/pd-l2
US20120114649A1 (en) 2008-08-25 2012-05-10 Amplimmune, Inc. Delaware Compositions of pd-1 antagonists and methods of use
US8345509B2 (en) 2009-04-16 2013-01-01 Chevron U.S.A., Inc. System and method to create three-dimensional images of non-linear acoustic properties in a region remote from a borehole
US20140341917A1 (en) 2011-11-28 2014-11-20 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof
WO2015033299A1 (fr) 2013-09-06 2015-03-12 Aurigene Discovery Technologies Limited Dérivés 1,2,4-oxadiazole utilisés comme immunomodulateurs
WO2015033301A1 (fr) 2013-09-06 2015-03-12 Aurigene Discovery Technologies Limited Dérivés 1,3,4-oxadiazole et 1,3,4-thiadiazole servant d'immunomodulateurs
WO2020194063A1 (fr) * 2019-03-28 2020-10-01 Institut Pasteur Flavivurus vivant et atténué comprenant une protéine m mutée

Patent Citations (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4960716A (en) 1984-05-01 1990-10-02 Ciba Corning Diagnostics Corp. Monoclonal antibodies specific for 330 KD breast tumor antigen and assay using said monoclonal antibodies
US5139941A (en) 1985-10-31 1992-08-18 University Of Florida Research Foundation, Inc. AAV transduction vectors
US5219740A (en) 1987-02-13 1993-06-15 Fred Hutchinson Cancer Research Center Retroviral gene transfer into diploid fibroblasts for gene therapy
WO1989012624A2 (fr) 1988-06-14 1989-12-28 Cetus Corporation Agents de couplage et conjugues lies a des disulfures a empechement sterique prepares a partir de tels agents
US5589466A (en) 1989-03-21 1996-12-31 Vical Incorporated Induction of a protective immune response in a mammal by injecting a DNA sequence
US5580859A (en) 1989-03-21 1996-12-03 Vical Incorporated Delivery of exogenous DNA sequences in a mammal
US5399346A (en) 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
US5073627A (en) 1989-08-22 1991-12-17 Immunex Corporation Fusion proteins comprising GM-CSF and IL-3
US5108910A (en) 1989-08-22 1992-04-28 Immunex Corporation DNA sequences encoding fusion proteins comprising GM-CSF and IL-3
WO1992001070A1 (fr) 1990-07-09 1992-01-23 The United States Of America, As Represented By The Secretary, U.S. Department Of Commerce Conditionnement a haute efficacite de virus adeno-associe mutant utilisant la suppression d'ambre
US5173414A (en) 1990-10-30 1992-12-22 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors
US5306811A (en) 1990-11-27 1994-04-26 Ciba Corning Diagnostics Corp. Squamous cell carcinoma-like immunoreactive antigen from human female urine
US5324822A (en) 1991-04-12 1994-06-28 Ciba Corning Diagnostics Corp. Method of isolating a CA 195-like immunoreactive antigen from human amniotic fluid
WO1993003769A1 (fr) 1991-08-20 1993-03-04 THE UNITED STATES OF AMERICA, represented by THE SECRETARY, DEPARTEMENT OF HEALTH AND HUMAN SERVICES Transfert induit par adenovirus de genes vers la voie gastro-intestinale
US5618829A (en) 1993-01-28 1997-04-08 Mitsubishi Chemical Corporation Tyrosine kinase inhibitors and benzoylacrylamide derivatives
US5728868A (en) 1993-07-15 1998-03-17 Cancer Research Campaign Technology Limited Prodrugs of protein tyrosine kinase inhibitors
US5804396A (en) 1994-10-12 1998-09-08 Sugen, Inc. Assay for agents active in proliferative disorders
US5639757A (en) 1995-05-23 1997-06-17 Pfizer Inc. 4-aminopyrrolo[2,3-d]pyrimidines as tyrosine kinase inhibitors
US6344459B1 (en) 1996-04-12 2002-02-05 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6127374A (en) 1997-07-29 2000-10-03 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6562818B1 (en) 1997-07-29 2003-05-13 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6100254A (en) 1997-10-10 2000-08-08 Board Of Regents, The University Of Texas System Inhibitors of protein tyrosine kinases
US6740665B1 (en) 1999-02-10 2004-05-25 Ramachandran Murali Tyrosine kinase inhibitors and methods of using the same
US6245759B1 (en) 1999-03-11 2001-06-12 Merck & Co., Inc. Tyrosine kinase inhibitors
US6544988B1 (en) 1999-03-11 2003-04-08 Merck & Co., Inc. Tyrosine kinase inhibitors
US6498165B1 (en) 1999-06-30 2002-12-24 Merck & Co., Inc. Src kinase inhibitor compounds
US6329380B1 (en) 1999-06-30 2001-12-11 Merck & Co., Inc. SRC kinase inhibitor compounds
US6316444B1 (en) 1999-06-30 2001-11-13 Merck & Co., Inc. SRC kinase inhibitor compounds
US6586423B2 (en) 1999-09-10 2003-07-01 Merck & Co., Inc. Tyrosine kinase inhibitors
US6586424B2 (en) 1999-09-10 2003-07-01 Merck & Co., Inc. Tyrosine kinase inhibitors
US6306874B1 (en) 1999-10-19 2001-10-23 Merck & Co., Inc. Tyrosine kinase inhibitors
US6479512B1 (en) 1999-10-19 2002-11-12 Merck & Co., Inc. Tyrosine kinase inhibitors
US6794393B1 (en) 1999-10-19 2004-09-21 Merck & Co., Inc. Tyrosine kinase inhibitors
US6313138B1 (en) 2000-02-25 2001-11-06 Merck & Co., Inc. Tyrosine kinase inhibitors
US6420382B2 (en) 2000-02-25 2002-07-16 Merck & Co., Inc. Tyrosine kinase inhibitors
US6875767B2 (en) 2001-06-22 2005-04-05 Merck & Co., Inc. (5-cyano-2-thiazolyl)amino-4-pyridine tyrosine kinase inhibitors
US6958340B2 (en) 2001-08-01 2005-10-25 Merck & Co., Inc. Tyrosine kinase inhibitors
US6927293B2 (en) 2001-08-30 2005-08-09 Merck & Co., Inc. Tyrosine kinase inhibitors
US20100028330A1 (en) 2002-12-23 2010-02-04 Medimmune Limited Methods of upmodulating adaptive immune response using anti-pd1 antibodies
US8008449B2 (en) 2005-05-09 2011-08-30 Medarex, Inc. Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
WO2006121168A1 (fr) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Anticorps monoclonaux humains pour mort programmee 1 (mp-1) et procedes pour traiter le cancer en utilisant des anticorps anti-mp-1 seuls ou associes a d’autres immunotherapies
WO2007005874A2 (fr) 2005-07-01 2007-01-11 Medarex, Inc. Anticorps monoclonaux humains diriges contre un ligand de mort programmee de type 1(pd-l1)
US20070254295A1 (en) 2006-03-17 2007-11-01 Prometheus Laboratories Inc. Methods of predicting and monitoring tyrosine kinase inhibitor therapy
WO2009101611A1 (fr) 2008-02-11 2009-08-20 Curetech Ltd. Anticorps monoclonaux pour le traitement de tumeurs
WO2009114335A2 (fr) 2008-03-12 2009-09-17 Merck & Co., Inc. Protéines de liaison avec pd-1
US8609089B2 (en) 2008-08-25 2013-12-17 Amplimmune, Inc. Compositions of PD-1 antagonists and methods of use
WO2010027827A2 (fr) 2008-08-25 2010-03-11 Amplimmune, Inc. Polypeptides co-stimulateurs ciblés et leurs procédés d'utilisation dans le traitement du cancer
US20120114649A1 (en) 2008-08-25 2012-05-10 Amplimmune, Inc. Delaware Compositions of pd-1 antagonists and methods of use
US8217149B2 (en) 2008-12-09 2012-07-10 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
WO2010077634A1 (fr) 2008-12-09 2010-07-08 Genentech, Inc. Anticorps anti-pd-l1 et leur utilisation pour améliorer la fonction des lymphocytes t
US8345509B2 (en) 2009-04-16 2013-01-01 Chevron U.S.A., Inc. System and method to create three-dimensional images of non-linear acoustic properties in a region remote from a borehole
WO2011066342A2 (fr) 2009-11-24 2011-06-03 Amplimmune, Inc. Inhibition simultanée de pd-l1/pd-l2
US20130034559A1 (en) 2009-11-24 2013-02-07 Medlmmune Limited Targeted Binding Agents Against B7-H1
WO2011066389A1 (fr) 2009-11-24 2011-06-03 Medimmmune, Limited Agents de liaison ciblés dirigés contre b7-h1
US20140341917A1 (en) 2011-11-28 2014-11-20 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof
WO2015033299A1 (fr) 2013-09-06 2015-03-12 Aurigene Discovery Technologies Limited Dérivés 1,2,4-oxadiazole utilisés comme immunomodulateurs
WO2015033301A1 (fr) 2013-09-06 2015-03-12 Aurigene Discovery Technologies Limited Dérivés 1,3,4-oxadiazole et 1,3,4-thiadiazole servant d'immunomodulateurs
WO2020194063A1 (fr) * 2019-03-28 2020-10-01 Institut Pasteur Flavivurus vivant et atténué comprenant une protéine m mutée

Non-Patent Citations (98)

* Cited by examiner, † Cited by third party
Title
"Genbank", Database accession no. NC_001132
"Monoclonal Antibodies For Cancer Detection And Therapy", 1985, ACADEMIC PRESS, article "Analysis, Results, and Future Prospective of the Therapeutic Use of Radiolabeled Antibody In Cancer Therapy"
"Remington's Pharmaceutical-Sciences", 1980, MACK PUBLISHING CO.
ACRES ET AL., CURR OPIN MOL THER, vol. 6, February 2004 (2004-02-01), pages 40 - 7
AGNEW CHEM INTL. ED. ENGL., vol. 33, 1994, pages 183 - 186
ANDTBACKA ET AL., J. CLIN. ONCOL., no. 31, 2013
ARNON ET AL.: "Monoclonal Antibodies And Cancer Therapy", 1985, ALAN R. LISS, INC., article "Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy"
BABON ET AL., NAT. MED., 2016
BERKERS ET AL., J. IMMUNOL., 2015
BERKNER, BIOTECHNIQUES, vol. 6, 1988, pages 616 - 629
BETT ET AL., J. VIROL., vol. 67, no. 591, 1993, pages 1 - 5921
BORIS-LAWRIE, TEMIN, CUR. OPIN. GENET. DEVELOP., vol. 3, 1993, pages 102 - 109
BOVIATSIS ET AL., GENE THER., vol. 1, pages 323 - 31
BRABANT, M.BAUX, L.CASIMIR, R.BRIAND, J.P.CHALOIN, O.PORCEDDU, M.BURON, N.CHAUVIER, D.LASALLE, M.LECOEUR, H.: "A flavivirus protein M-derived peptide directly permeabilizes mitochondrial membranes, triggers cell death and reduces human tumor growth in nude mice", APOPTOSIS, vol. 14, no. 10, 2009, pages 1190 - 203, XP019727368, DOI: 10.1007/s10495-009-0394-y
BRESSAC, B.GALVIN, K.M.LIANG, T.J.ISSELBACHER, K.J.WANDS, J.R.OZTURK, M.: "Abnormal structure and expression of p53 gene in human hepatocellular carcinoma", PROC NATL ACAD SCI USA, vol. 87, no. 5, 1990, pages 1973 - 7
BROWN, E.BENTHAM, MBEAUMONT, H.BLOY, A.THOMPSON, R.MCKIMMIE, C.FOSTER, R.RANSON, N.MACDONALD, A.KALI, A.: "Flavivirus membrane (M) proteins as potential ion channel antiviral", MICROBIOLOGY, vol. 1, no. 1A, 2019
BURNS ET AL., PROC. NATL ACAD. SCI. USA, vol. 90, 1993, pages 8033 - 8037
CARTER, CURRENT OPINION IN BIOTECHNOLOGY, vol. 3, 1992, pages 533 - 539
CATTEAU, A.KALININA, O.WAGNER, M.C.DEUBEL, V.COURAGEOT, M.PDESPRES, P.: "Dengue virus M protein contains a proapoptotic sequence referred to as ApoptoM", J GEN VIROL, vol. 84, 2003, pages 2781 - 2793, XP002405613, DOI: 10.1099/vir.0.19163-0
CATTEAU, A.ROUE, G.YUSTE, V.J.SUSIN, S.A.DESPRES, P., EXPRESSION OF DENGUE APOPTOM SEQUENCE RESULTS IN DISRUPTION OF MITOCHONDRIAL POTENTIAL AND CASPASE ACTIVATION, vol. 85, no. 8, 2003, pages 789 - 93
CHAMBERS ET AL., PROC. NATL. ACAD. SCI. USA, vol. 92, 1995, pages 1411 - 5
CHANG, K.I. PASTAN: "Molecular cloning of mesothelin, a differentiation antigen present on mesothelium, mesotheliomas, and ovarian cancers", PROC NATL ACAD SCI USA, vol. 93, no. 1, 1996, pages 136 - 40, XP002153471, DOI: 10.1073/pnas.93.1.136
CHEN, Q.WU, J.YE, Q.MA, F.ZHU, Q.WU, Y.SHAN, C.XIE, X.LI, D.ZHAN, X.: "Treatment of Human Glioblastoma with a Live Attenuated Zika Virus Vaccine Candidate", MBIO, vol. 10, no. 2, 2019
CHEN, Y.T.L.J. OLD: "Cancer-testis antigens: targets for cancer immunotherapy", CANCER J SCI AM, vol. 5, no. 1, 1999, pages 16 - 7
CHONG, H.Y.LEOW, C.Y.ABDUL MAJEED, A.B.LEOW, C.H.: "Flavivirus infection-A review of immunopathogenesis, immunological response, and immunodiagnosis", VIRUS RES, vol. 274, 2019, pages 197770, XP085882742, DOI: 10.1016/j.virusres.2019.197770
DATABASE Geneseq [online] 1 October 2020 (2020-10-01), "Zika virus M protein TM1 ectodomain, SEQ ID 29.", XP055797248, retrieved from EBI accession no. GSP:BIJ53317 Database accession no. BIJ53317 *
DE WISPELAERE, M.KHOU, C.FRENKIEL, M.P.DESPRES, P.PARDIGON, N.: "A Single Amino Acid Substitution in the M Protein Attenuates Japanese Encephalitis Virus in Mammalian Hosts", J VIROL, vol. 90, no. 5, 2015, pages 2676 - 89
DELONG ET AL., SCIENCE, 2016
DERMIME, S.GILHAM, D.E.SHAW, D.MDAVIDSON, E.J.MEZIANE EL, K.ARMSTRONG, A.HAWKINS, R.E.STERN, P.L.: "Vaccine and antibody-directed T cell tumour immunotherapy", BIOCHIM BIOPHYS ACTA, vol. 1704, no. 1, 2004, pages 11 - 35, XP004517446, DOI: 10.1016/j.bbcan.2004.03.002
ELKORD, E.BURT, D.J.DRIJHOUT, J.W.HAWKINS, R.E.STERN, P.L.: "CD4+ T-cell recognition of human 5T4 oncofoetal antigen: implications for initial depletion of CD25+ T cells", CANCER IMMUNOL IMMUNOTHER, vol. 57, no. 6, 2008, pages 833 - 47, XP019586731
EMENS ET AL., CANCER BIOL THER., vol. 2, July 2003 (2003-07-01), pages S161 - 8
FREEMAN ET AL., MOL. THER., vol. 13, no. 1, 2006, pages 221 - 8
FRUMENCE, E.VIRANAICKEN, W.BOS, S.ALVAREZ-MARTINEZ, M.T.ROCHE, M.ARNAUD, J.D.GADEA, G.DESPRES, P.: "A Chimeric Zika Virus between Viral Strains MR766 and BeH819015 Highlights a Role for E-glycan Loop in Antibody-mediated Virus Neutralization", VACCINES (BASEL, vol. 7, no. 2, 2019
GEEVARGHESE ET AL., HUM. GENE THER., vol. 21, no. 9, 2010, pages 1119 - 28
HADDOW, A.D.SCHUH, A.J.YASUDA, C.Y.KASPER, M.R.HEANG, V.HUY, R.GUZMAN, H.TESH, R.B.WEAVER, S.C.: "Genetic characterization of Zika virus strains: geographic expansion of the Asian lineage", PLOS NEGL TROP DIS, vol. 6, no. 2, 2012, pages e1477, XP055425625, DOI: 10.1371/journal.pntd.0001477
HAJ-AHMAD ET AL., J. VIROL., vol. 57, 1986, pages 267 - 274
HELLSTROM ET AL.: "Controlled Drug Delivery", 1987, MARCEL DEIKER, INC., article "Antibodies For Drug Delivery"
HO, M.BERA, T.K.WILLINGHAM, M.C.ONDA, M.HASSAN, R.FITZGERALD, D.PASTAN, I.: "Mesothelin expression in human lung cancer", CLIN CANCER RES, vol. 13, no. 5, 2007, pages 1571 - 5, XP055110225, DOI: 10.1158/1078-0432.CCR-06-2161
HSIEH, S.C.WU, Y.C.ZOU, G.NERURKAR, V.R.SHI, P.Y.WANG, W.K.: "Highly conserved residues in the helical domain of dengue virus type 1 precursor membrane protein are involved in assembly, precursor membrane (prM) protein cleavage, and entry", J BIOL CHEM, vol. 289, no. 48, 2014, pages 33149 - 60
KAID, C.GOULART, E.CAIRES-JUNIOR, L.C.ARAUJO, B.H.S.SOARES-SCHANOSKI, A.BUENO, H.M.S.TELLES-SILVA, K.A.ASTRAY, R.M.ASSONI, A.F.JUN: "Zika Virus Selectively Kills Aggressive Human Embryonal CNS Tumor Cells In Vitro and In Vivo", CANCER RES, vol. 78, no. 12, 2018, pages 3363 - 3374, XP055648362, DOI: 10.1158/0008-5472.CAN-17-3201
KAUFMANN ET AL., J. INVEST. DERMATOL., vol. 133, no. 4, 2013, pages 1034 - 42
KHURI ET AL., NAT. MED, vol. 6, no. 8, 2000, pages 879 - 85
KIRN ET AL., NAT. MED., vol. 7, 2001, pages 781
LANE, D.P.: "Cancer. p53, guardian of the genome", NATURE, vol. 358, no. 6381, 1992, pages 15 - 6
LEBKOWSKI ET AL., MOLEC. CELL BIOL., vol. 8, 1988, pages 3988 - 3996
LEVINE, A.J.M. OREN: "The first 30 years of p53: growing ever more complex", NAT REV CANCER, vol. 9, no. 10, 2009, pages 749 - 58, XP009186382, DOI: 10.1038/nrc2723
LI, C.XU, D.YE, Q.HONG, S.JIANG, Y.LIU, X.ZHANG, N.SHI, L.QIN, C.F.XU, Z.: "Zika Virus Disrupts Neural Progenitor Development and Leads to Microcephaly in Mice", CELL STEM CELL, vol. 19, no. 5, 2016, pages 672
LI, G.POULSEN, M.YASHIRODA, Y.SIMARD, J.M.GALLO, R.C.ZHAO, R.Y.: "Characterization of cytopathic factors through genome-wide analysis of the Zika viral proteins in fission yeast", PROC NATL ACAD SCI USA, vol. 114, no. 3, 2017, pages E376 - E385
LIU JUN ET AL: "Zika Virus Envelope Protein induces G2/M Cell Cycle Arrest and Apoptosis via an Intrinsic Cell Death Signaling Pathway in Neuroendocrine PC12 Cells", INTERNATIONAL JOURNAL OF BIOLOGICAL SCIENCES, vol. 14, no. 9, 1 January 2018 (2018-01-01), Australia, pages 1099 - 1108, XP055797060, ISSN: 1449-2288, DOI: 10.7150/ijbs.26400 *
LIU, J.LI, Q.LI, X.QIU, Z.LI, A.LIANG, W.CHEN, H.CAI, X.CHEN, X.DUAN, X.: "Zika Virus Envelope Protein induces G2/M Cell Cycle Arrest and Apoptosis via an Intrinsic Cell Death Signaling Pathway in Neuroendocrine PC12 Cells", INT J BIOL SCI, vol. 14, no. 9, 2018, pages 1099 - 1108, XP055797060, DOI: 10.7150/ijbs.26400
LORENCE ET AL., CURR. CANCER DRUG TARGETS, vol. 7, no. 2, 2007, pages 157 - 67
LU, W.CHENG, F.YAN, W.LI, X.YOA, X.SONG, W.LIU, M.SHEN, X.JIANG, H.CHEN, J.: "Selective targeting p53(WT) lung cancer cells harboring homozygous p53 Arg72 by an inhibitor of CypA", ONCOGENE, vol. 36, no. 33, 2017, pages 4719 - 4731
M. HO ET AL: "Mesothelin Expression in Human Lung Cancer", CLINICAL CANCER RESEARCH, vol. 13, no. 5, 1 March 2007 (2007-03-01), pages 1571 - 1575, XP055110225, ISSN: 1078-0432, DOI: 10.1158/1078-0432.CCR-06-2161 *
MAGALI BRABANT ET AL: "A flavivirus protein M-derived peptide directly permeabilizes mitochondrial membranes, triggers cell death and reduces human tumor growth in nude mice", APOPTOSIS ; AN INTERNATIONAL JOURNAL ON PROGRAMMED CELL DEATH, KLUWER ACADEMIC PUBLISHERS, BO, vol. 14, no. 10, 20 August 2009 (2009-08-20), pages 1190 - 1203, XP019727368, ISSN: 1573-675X, DOI: 10.1007/S10495-009-0394-Y *
MARTUZA ET AL., SCIENCE, vol. 252, 1991, pages 854 - 6
MCDONALD ET AL., BREAST CANCER TREAT, vol. 99, no. 2, 2006, pages 177 - 84
MILLER ET AL., BIOTECHNIQUES, vol. 7, 1989, pages 980 - 990
MILLER, HUMAN GENE THERAPY, vol. 1, 1990, pages 5 - 14
MINETA ET AL., CANCER RES., vol. 54, 1994, pages 3363 - 66
MITTEREDER ET AL., HUMAN GENE THERAPY, vol. 5, 1994, pages 793 - 801
MORCHANG, A.LEE, R.C.H.YENCHITSOMANUS, P.T.SREEKANTH, G.P.NOISAKRAN, S.CHU, J.J.H.LIMJINDAPORN, T.: "RNAi screen reveals a role of SPHK2 in dengue virus-mediated apoptosis in hepatic cell lines", PLOS ONE, vol. 12, no. 11, 2017, pages e0188121
MORELLO, A.M. SADELAINP.S. ADUSUMILLI: "Mesothelin-Targeted CARs: Driving T Cells to Solid Tumors", CANCER DISCOV, vol. 6, no. 2, 2016, pages 133 - 46, XP055239486, DOI: 10.1158/2159-8290.CD-15-0583
MUZYCZKA, CURRENT TOPICS IN MICROBIOL. AND IMMUNOL., vol. 158, 1992, pages 97 - 129
OHSHIMA ET AL., INT J CANCER., vol. 93, no. 1, 1 July 2001 (2001-07-01), pages 91 - 6
PAN, P.ZHANG, Q.LIU, W.WANG, W.LAO, Z.ZHANG, W.SHEN, M.WAN, P.XIAO, F.LIU, F., DENGUE VIRUS M PROTEIN PROMOTES NLRP3 INFLAMMASOME ACTIVATION TO INDUCE VASCULAR LEAKAGE IN MICE, vol. 93, no. 21, 2019
PARQUET, M.C.KUMATORI, A.HASEBE, F.MATHENGE, E.G.MORITA, K.: "St. Louis encephalitis virus induced pathology in cultured cells", ARCH VIROL, vol. 147, no. 6, 2002, pages 1105 - 19
PARQUET, M.C.KUMATORI, A.HASEBE, F.MORITA, K.IGARASHI, A.: "West Nile virus-induced bax- dependent apoptosis", FEBS LETT, vol. 500, no. 1-2, 2001, pages 17 - 24, XP004251370, DOI: 10.1016/S0014-5793(01)02573-X
PENG, J.G.S.C. WU: "Glutamic acid at residue 125 of the prM helix domain interacts with positively charged amino acids in E protein domain II for Japanese encephalitis virus-like-particle production", J VIROL, vol. 88, no. 15, 2014, pages 8386 - 96
PHUANGSAB ET AL., CANCER LETT., vol. 172, no. 1, 2001, pages 27 - 36
PRYOR, M.J.AZZOLA, L.WRIGHT, P.J.DAVIDSON, A.D.: "Histidine 39 in the dengue virus type 2 M protein has an important role in virus assembly", J GEN VIROL, vol. 85, 2004, pages 3627 - 3636
PYLES ET AL., J. VIROL., vol. 68, 1994, pages 4963 - 940
RIBAS, A.WEBER, J.S.CHMIELOWSKI, B.COMIN-ANDUIX, B.LU, D.DOUEK, M.RAGAVENDRA, N.RAMAN, S.SEJA, E.ROSARIO, D.: "Intra- lymph node prime-boost vaccination against Melan A and tyrosinase for the treatment of metastatic melanoma: results of a phase 1 clinical trial", CLIN CANCER RES, vol. 17, no. 9, 2011, pages 2987 - 96
RICH ET AL., HUMAN GENE THERAPY, vol. 4, 1993, pages 461 - 476
ROSA-FERNANDES, L.CUGOLA, F.R.RUSSO, F.B.KAWAHARA, R.DE MELO FREIRE, C.C.LEITE, P.E.C.BASSI STERN, A.C.ANGELI, C.B.DE OLIVERIA, D., ZIKA VIRUS IMPAIRS NEUROGENESIS AND SYNAPTOGENESIS PATHWAYS IN HUMAN NEURAL STEM CELLS AND
RUBIN, E.J.M.F. GREENEL.R. BADEN: "Zika Virus and Microcephaly", N ENGL J MED, vol. 374, no. 10, 2016, pages 984 - 5
RUDIN ET AL., CLIN. CANCER. RES., vol. 17, no. 4, 2011, pages 888 - 95
SCARPA ET AL., VIROLOGY, vol. 180, 1991, pages 849 - 852
SHELLING ET AL., GENE THERAPY, vol. 1, 1994, pages 165 - 169
SOLOMON, I.H.D.A. MILNERR.D. FOLKERTH, NEUROPATHOLOGY OF ZIKA VIRUS INFECTION. J NEUROINFECT DIS, vol. 7, no. 2, 2016
SOUZA, B.S.SAMPAIO, G.L.PEREIRA, C.S.CAMPOS, G.S.SARDI, S.I.FREITAS, L.A.FIGUEIRA, C.P.PAREDES, B.D.NONAKA, C.K.AZEVEDO, C.M.: "Zika virus infection induces mitosis abnormalities and apoptotic cell death of human neural progenitor cells", SCI REP, vol. 6, 2016, pages 39775
STOJDL ET AL., CANCER CELL, vol. 4, no. 4, 2003, pages 263 - 75
STOJDL ET AL., NAT. MED., vol. 6, no. 7, 2000, pages 821 - 5
SUZUKI, T.OKAMOTO, T.KATOH, H.SUGIYAMA, Y.KUSAKABE, S.TOKUNAGA, M.HIRANO, J.MIYATA, Y.FUKUHARA, T.IKAWA, M.: "Infection with flaviviruses requires BCLXL for cell survival", PLOS PATHOG, vol. 14, no. 9, 2018, pages e1007299
TAYLOR-PAPADIMITRIOU ET AL., BIOCHIM BIOPHYS ACTA, vol. 1455, no. 2-3, 8 October 1999 (1999-10-08), pages 301 - 13
THORPE ET AL., IMMUNOL. REV., vol. 62, 1982, pages 119 - 58
THORPE ET AL.: "Monoclonal Antibodies '84: Biological And Clinical Applications", 1985, article "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review"
TURPIN JONATHAN ET AL: "The ZIKA Virus Delays Cell Death Through the Anti-Apoptotic Bcl-2 Family Proteins", CELLS, vol. 8, no. 11, 29 October 2019 (2019-10-29), pages 1338, XP055797056, DOI: 10.3390/cells8111338 *
VAN MEERTEN, T.A. HAGENBEEK: "CD20-targeted therapy: the next generation of antibodies", SEMIN HEMATOL, vol. 47, no. 2, 2010, pages 199 - 210
VANWALSCAPPEL BÉNÉDICTE ET AL: "Zika M Oligopeptide ZAMP Confers Cell Death-Promoting Capability to a Soluble Tumor-Associated Antigen through Caspase-3/7 Activation", INT. J. MOL. SCI., vol. 21, no. 24, 1 December 2020 (2020-12-01), pages 9578, XP055797040, ISSN: 1661-6596, DOI: 10.3390/ijms21249578 *
VIGNERON ET AL., SCIENCE, 2004
WARREN ET AL., SCIENCE, 2006
WILLISON, H.J.MUSSET, L.MANUGUERRA, J.C.DESPRES, P.FOURNIER, E.MALLET, H.P.MUSSO, D.FONTANET, A.NEIL, J.GHAWCHZ, F.: "Guillain-Barre Syndrome outbreak associated with Zika virus infection in French Polynesia: a case-control study", LANCET, vol. 387, no. 10027, 2016, pages 1531 - 1539, XP029496412, DOI: 10.1016/S0140-6736(16)00562-6
WONG ET AL., VIRUSES, vol. 2, 2010, pages 78 - 106
XIAAI ZHENG ET AL., ONCORINE, vol. 23, no. 12, 2004, pages 1666 - 70
YOSHII, KIGARASHI, M.ICHII, O.YOKOZAWA, K.ITO, K.KARIWA, H.TAKASHIMA, I.: "A conserved region in the prM protein is a critical determinant in the assembly of flavivirus particles", J GEN VIROL, vol. 93, 2012, pages 27 - 38
ZHE ZHU ET AL: "Zika virus has oncolytic activity against glioblastoma stem cells", JOURNAL OF EXPERIMENTAL MEDICINE, vol. 214, no. 10, 5 September 2017 (2017-09-05), US, pages 2843 - 2857, XP055436428, ISSN: 0022-1007, DOI: 10.1084/jem.20171093 *
ZHOU ET AL., J. EXP. MED., vol. 179, 1994, pages 1867 - 1875
ZHU, Z.GORMAN, M.J.MCKENZIE, L.D.CHAI, J.N.HUBERT, C.G.PRAGER, B.C.FERNANDEZ, E.RICHNER, J.M.ZHANG, R.SHAN, C.: "Correction: Zika virus has oncolytic activity against glioblastoma stem cells", J EXP MED, vol. 214, no. 10, 2017, pages 3145, XP055436428, DOI: 10.1084/jem.20171093

Similar Documents

Publication Publication Date Title
EP3718565A1 (fr) Vaccins contre le virus respiratoire
EP2477499B1 (fr) Polythérapie anticancéreuse à virus de la vaccine oncolytique
US20200000862A1 (en) Oncolytic virus therapy
EP4137578A1 (fr) Vecteurs de la vaccine modifiés
AU2013315007A1 (en) Compositions and methods for treating amyotrophic lateral sclerosis
US11939609B2 (en) Rewiring aberrant cancer signaling to a therapeutic effector response with a synthetic two-component system
CA3147640A1 (fr) Therapie oncolytique du virus de la vallee seneca de seconde generation : composition et procedes associes
WO2022101463A1 (fr) Utilisation des derniers résidus de l'extrémité c-terminale m31/41 de l'ectodomaine zikv m pour déclencher la mort cellulaire apoptotique
US20220049228A1 (en) Modified Extracellular Enveloped Virus
WO2015168102A2 (fr) Stabilisation de protéine prolongée par le resvératrol
JP7457037B2 (ja) M1ウイルス変異体及びその使用
US20230338384A1 (en) Methods of treating cancer
Crosley Preclinical evaluation of TRAIL combined with PAC-1 to control granulosa cell tumour
US20230346901A1 (en) Methods and vaccine compositions to treat cancers
EP4268837A1 (fr) Nouveau réovirus modifié et son utilisation
Becker The combination of carboxylesterase-expressing oncolytic vaccinia virus and irinotecan
Puigdelloses-Vallcorba Immuno-virotherapy for glioblastoma: Characterization of Delta 24-ACT in combination with different immunomodulators as therapeutic approach
KR20220116191A (ko) 4-1bbl 아쥬반트화 재조합 변형 백시니아 바이러스 앙카라 (mva)의 의약적 용도
Pizzuto Avian influenza virus as an oncolytic therapy for pancreatic cancer

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: 21810020

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: 21810020

Country of ref document: EP

Kind code of ref document: A1