WO2024084034A1 - Méthodes et compositions pharmaceutiques pour le traitement de l'arthrose - Google Patents

Méthodes et compositions pharmaceutiques pour le traitement de l'arthrose Download PDF

Info

Publication number
WO2024084034A1
WO2024084034A1 PCT/EP2023/079262 EP2023079262W WO2024084034A1 WO 2024084034 A1 WO2024084034 A1 WO 2024084034A1 EP 2023079262 W EP2023079262 W EP 2023079262W WO 2024084034 A1 WO2024084034 A1 WO 2024084034A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
inhibitor
antibody
osteoarthritis
subject
Prior art date
Application number
PCT/EP2023/079262
Other languages
English (en)
Inventor
Julien CHERFILS-VICINI
Eric Gilson
Christian Jorgensen
Jean-Marc BRONDELLO
Yves-Marie PERS
Christina FISSOUN
Original Assignee
Institut National de la Santé et de la Recherche Médicale
Centre National De La Recherche Scientifique
Universite Cote D'azur
Centre Hospitalier Universitaire De Nice
Centre Hospitalier Universitaire De Montpellier
Université De Montpellier
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 Institut National de la Santé et de la Recherche Médicale, Centre National De La Recherche Scientifique, Universite Cote D'azur, Centre Hospitalier Universitaire De Nice, Centre Hospitalier Universitaire De Montpellier, Université De Montpellier filed Critical Institut National de la Santé et de la Recherche Médicale
Publication of WO2024084034A1 publication Critical patent/WO2024084034A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3076Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells against structure-related tumour-associated moieties
    • C07K16/3084Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells against structure-related tumour-associated moieties against tumour-associated gangliosides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the present invention relates to methods and pharmaceutical compositions for the treatment of osteoarthritis.
  • Osteoarthritis is the most common degenerative joint disease among the rheumatic disorders affecting the Western world. This condition is one of the main causes of pain and incapacity in elderly people, becoming a major health problem.
  • Osteoarthritis is characterized by degeneration of the articular cartilage, remodelling of the subchondral bone and changes in the synovial membrane. It commonly affects the hands, feet, spine, and large extraspinal, weight-bearing joints, such as the hips and knees.
  • the etiology of osteoarthritis is multifactorial involving both mechanical and biochemical factors.
  • Clinical classification includes several phenotypes such as post-traumatic, metabolic, ageing or genetic.
  • OA joint pain, stiffness in the morning or after rest, pain at night, limited motion, joint deformity, associated with variable degrees of inflammation of the synovial membrane (synovitis).
  • Joint pain in OA may originate not only from synovitis, but also from stretching of the joint capsule or ligaments, periosteal irritation, trabecular microfractures, intraosseous hypertension or muscle spasms
  • osteoarthrosis Some treatments of osteoarthrosis are available, such as analgesics, nonsteroidal antiinflammatory drugs, corticosteroids, hyaluronic acid injection, surgery (realigning bones or joint replacement), but these treatments are of high cost. Thus, there is a need for an improved method to treat osteoarthritis.
  • the present invention relates to methods and pharmaceutical compositions for the treatment of osteoarthritis.
  • the present invention is defined by the claims. DETAILED DESCRIPTION OF THE INVENTION:
  • Osteoarthritis is a type of degenerative joint disease that results from breakdown of joint cartilage and underlying bone.
  • the inventors studied the effects of intra-articular anti-GD3 monoclonal antibody injection.
  • the present invention relates to a method for treating osteoarthritis in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a GD3 inhibitor.
  • the terms “subject” or “patient” denote a mammal, such as a rodent, a feline, a canine, and a primate.
  • the subject according to the invention is a human.
  • the subject according to the invention is a child, a teenager, an adult or an elderly person.
  • the subject is more than 15 years old.
  • the subject is more than 20 years old.
  • the subject is more than 25 years old.
  • the subject is more than 30 years old.
  • the subject is more than 35 years old.
  • osteoarthritis As used herein, the term “osteoarthritis” (OA) has its general meaning in the art and refers to a degenerative joint disease with moderate local inflammation occurring chiefly in older humans and animals, which is characterized by degeneration of the articular cartilage, remodelling of the subchondral bone and changes in the synovial membrane. Osteoarthritis can affect any joint of the organism, such as knee, hip, elbow, hands joints, shoulder, back (for example, spinal or cervical), foot joints, ankle, etc. Osteoarthritis can be a metabolic osteoarthritis, which is related to an accumulation of metabolic abnormalities or metabolic syndrome.
  • osteoarthritis is metabolic osteoarthritis.
  • osteoarthritis is post-traumatic osteoarthritis.
  • post-traumatic osteoarthritis refers to inflammation in joints that forms after experienced a trauma. It develops quickly after an injury instead of over years of wear and tear like other forms of arthritis. Post-traumatic arthritis can last longer and becomes a chronic (long-term) condition.
  • ganglioside refers to a molecule composed of a glycosphingolipid (ceramide and oligosaccharide) with one or more sialic acids (e.g. n-acetylneuraminic acid, NANA) linked on the sugar chain.
  • sialic acids e.g. n-acetylneuraminic acid, NANA
  • NeuNAc an acetylated derivative of the carbohydrate sialic acid, makes the head groups of gangliosides anionic at pH 7, which distinguishes them from globosides.
  • the present invention relates to the inhibition of the enzyme ST8SIA1.
  • ST8SIA1 also known as “Alpha-N-acetylneuraminide alpha-2, 8- sialyltransferase” has its general meaning in this art and Catalyzes the addition of sialic acid in alpha 2,8-linkage to the sialic acid moiety of the ganglioside GM3 to form ganglioside GD3 (Uniprot accession number Q92185).
  • inhibitor includes not only drugs for inhibiting activity of target molecules, but also drugs for inhibiting the expression of target molecules.
  • the term “GD3” has its general meaning in this art and is defined by the chemical structure: aNeu5Ac(2-8)aNeu5Ac(2-3)bDGalp(l-4)bDGlcp(l-l)Cer.
  • the GD3 is human GD3.
  • the GD3 is rat GD3.
  • the GD3 is mouse GD3.
  • the GD3 is primate GD3.
  • GD3 includes variants, isoforms, homologs, orthologs and paralogs of human ganglioside GD3.
  • the present invention relates to the inhibition of GD3.
  • GD3 inhibitor has its general meaning in the art and refers to an inhibitor of GD3.
  • the inhibitor of GD3 include but are not limited to GD3 antibody, CAR-T cell, CAR NK cell.
  • the present invention relates to senotherapy strategy including any type of targeting (e.g. antibody, drug, CAR T, CAR NK...) of GD3 that aimed at blocking with senomorphic agent or eliminating with senolytic agent senescent cells by targeting GD3.
  • the GD3 inhibitor is an antibody.
  • antibody is used in the broadest sense and specifically covers monoclonal antibodies, polyclonal antibodies, multi-specific antibodies (e.g. bispecific antibodies) formed from at least two intact antibodies, and antibody fragments so long as they exhibit the desired biological activity.
  • the term includes antibody fragments that comprise an antigen binding domain such as Fab', Fab, F(ab')2, single domain antibodies (DABs), TandAbs dimer, Fv, scFv (single chain Fv), dsFv, ds-scFv, Fd, linear antibodies, minibodies, diabodies, bispecific antibody fragments, bibody, tribody (scFv-Fab fusions, bispecific or trispecific, respectively); sc-diabody; kappa(lamda) bodies (scFv-CL fusions); BiTE (Bispecific T-cell Engager, scFv-scFv tandems to attract T cells); DVD-Ig (dual variable domain antibody, bispecific format); SIP (small immunoprotein, a kind of minibody); SMTP ("small modular immunopharmaceutical” scFv-Fc dimer; DART (ds-stabilized diabody "Dual Affinity ReTargeting"
  • Antibodies can be fragmented using conventional techniques. For example, F(ab')2 fragments can be generated by treating the antibody with pepsin. The resulting F(ab')2 fragment can be treated to reduce disulfide bridges to produce Fab' fragments. Papain digestion can lead to the formation of Fab fragments.
  • Fab, Fab' and F(ab')2, scFv, Fv, dsFv, Fd, dAbs, TandAbs, ds-scFv, dimers, minibodies, diabodies, bispecific antibody fragments and other fragments can also be synthesized by recombinant techniques or can be chemically synthesized. Techniques for producing antibody fragments are well known and described in the art. For example, each of Beckman et al., 2006; Holliger & Hudson, 2005; Le Gall et al., 2004; Reff & Heard, 2001 ; Reiter et al., 1996; and Young et al., 1995 further describe and enable the production of effective antibody fragments.
  • the antibody is a “chimeric” antibody as described in U.S. Pat. No. 4,816,567.
  • the antibody is a humanized antibody, such as described U.S. Pat. Nos. 6,982,321 and 7,087,409.
  • the antibody is a human antibody.
  • a “human antibody” such as described in US 6,075,181 and 6,150,584.
  • the antibody is a single domain antibody such as described in EP 0 368 684, WO 06/030220 and WO 06/003388.
  • the inhibitor is a monoclonal antibody.
  • Monoclonal antibodies can be prepared and isolated using any technique to produce antibody molecules by continuous cell lines in culture.
  • the antibody is specific of the isoform B of GD3.
  • the antibody is a single domain antibody.
  • the term “single domain antibody” (sdAb) or "VHH” refers to the single heavy chain variable domain of antibodies of the type that can be found in Camelid mammals which are naturally devoid of light chains. Such VHH are also called “nanobody®”. According to the invention, sdAb can particularly be llama sdAb.
  • Sanofi-Genzyme has developed a blocking and depleting antibody SAR440241.
  • R&D Systems has developed a blocking antibody MAB-160. This kind of antibodies affects mostly immune cells, notably the immune adaptive system and not melanocytes.
  • the GD3 antibody includes but are not limited to the clone MB3.6 or the clone R24 or derivative from those clones (afucosylated, humanized).
  • Examples of GD3 antibody includes but are not limited to Ganglioside GD3 Monoclonal Antibody of LifeSpan BioSciences, ST8 alpha-2, 8-Sialyltransferase 8A /ST8SIA1 /Ganglioside GD3 Antibody of Novus Biologicals, Ganglioside GD3 (2Q631) Antibody and Ganglioside GD3 (MB3.6) Antibody of Santa Cruz Biotechnology, Inc., Mab Mo x human Ganglioside GD3 antibody of United States Biological, Mitumomab, Ecromeximab and clone U36 of Creative Biolabs.
  • the GD3 inhibitor is a CAR-T cell.
  • CAR-T cell refers to a T lymphocyte that has been genetically engineered to express a CAR.
  • the definition of CAR T-cells encompasses all classes and subclasses of T-lymphocytes including CD4+ , CD8+ T cells, gamma delta T cells as well as effector T cells, memory T cells, regulatory T cells, and the like.
  • the T lymphocytes that are genetically modified may be "derived” or “obtained” from the subject who will receive the treatment using the genetically modified T cells or they may "derived” or “obtained” from a different subject.
  • a CAR Chimeric Antigen Receptor or alternatively a “CAR” refers to a set of polypeptides, typically two in the simplest embodiments, which when in an immune effector cell, provides the cell with specificity for a target cell, typically a cancer cell, and with intracellular signal generation.
  • a CAR comprises at least an extracellular antigen binding domain, a transmembrane domain and a cytoplasmic signaling domain (also referred to herein as “an intracellular signaling domain”) comprising a functional signaling domain derived from a stimulatory molecule and/or costimulatory molecule as defined below.
  • the set of polypeptides are contiguous with each other.
  • the set of polypeptides include a dimerization switch that, upon the presence of a dimerization molecule, can couple the polypeptides to one another, e.g., can couple an antigen binding domain to an intracellular signaling domain.
  • the stimulatory molecule is the zeta chain associated with the T cell receptor complex.
  • the cytoplasmic signaling domain further comprises one or more functional signaling domains derived from at least one costimulatory molecule as defined below.
  • the costimulatory molecule is chosen from the costimulatory molecules described herein, e.g., 4-1BB (i.e., CD137), CD27 and/or CD28.
  • the CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain and an intracellular signaling domain comprising a functional signaling domain derived from a stimulatory molecule. In some embodiments, the CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain and an intracellular signaling domain comprising afunctional signaling domain derived from a costimulatory molecule and a functional signaling domain derived from a stimulatory molecule.
  • the CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain and an intracellular signaling domain comprising two functional signaling domains derived from one or more costimulatory molecule(s) and a functional signaling domain derived from a stimulatory molecule. In some embodiments, the CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain and an intracellular signaling domain comprising at least two functional signaling domains derived from one or more costimulatory molecule(s) and a functional signaling domain derived from a stimulatory molecule.
  • the CAR comprises an optional leader sequence at the amino-terminus (N-ter) of the CAR fusion protein.
  • the CAR further comprises a leader sequence at the N-terminus of the extracellular antigen binding domain, wherein the leader sequence is optionally cleaved from the antigen binding domain (e g., a scFv) during cellular processing and localization of the CAR to the cellular membrane.
  • CARs comprise fusions of single-chain variable fragments (scFv) derived from monoclonal antibodies, fused to CD3-zeta a transmembrane domain and endodomain.
  • CARs comprise domains for additional co-stimulatory signaling, such as CD3-zeta, FcR, CD27, CD28, CD137, DAPIO, and/or 0X40.
  • molecules can be co-expressed with the CAR, including co-stimulatory molecules, reporter genes for imaging (e.g., for positron emission tomography), gene products that conditionally ablate the T cells upon addition of a pro-drug, homing receptors, chemokines, chemokine receptors, cytokines, and cytokine receptors.
  • the GD3 inhibitor is a CARNK cell.
  • CAR-NK refers to natural killer (NK) cells that has been genetically engineered to express a CAR.
  • NK cells are defined as CD56+ and CD3- cells and are subdivided into cytotoxic and immunoregulatory. They are of great clinical interest because they contribute to the graft-vs-leukemia/graft-vs-tumor effect but are not responsible for graft- vs-host disease.
  • NK cells can be generated from various sources such as umbilical cord blood, bone marrow, human embryonic stem cells, and induced pluripotent stem cells.
  • NK cells can escape the cytotoxicity of NK cells when they are directed against NKG2D ligands MICA and MICB (major histocompatibility complex class I chain-related protein A/B).
  • MICA and MICB major histocompatibility complex class I chain-related protein A/B.
  • CAR-modified primary human NK cells redirected against CD 19, CD20, CD244, and HER2, as well as CAR-expressing NK-92 cells targeted to a wider range of cancer antigens.
  • Primary NK cells engineered to express CARs have potential benefits compared to CAR-T cells. NK cells have spontaneous cytotoxic activity and can generate target cell death independent of tumor antigen, while T lymphocytes only kill their targets by a CAR-specific mechanism.
  • NK cells would still be effective against tumor cells.
  • primary human NK cells produce cytokines, such as interferon gamma, interleukin 3, and granulocyte-macrophage colony-stimulating factor, that differ from the proinflammatory cytokines produced by T cells that are responsible for the onset of cytokine release syndrome.
  • cytokines such as interferon gamma, interleukin 3, and granulocyte-macrophage colony-stimulating factor, that differ from the proinflammatory cytokines produced by T cells that are responsible for the onset of cytokine release syndrome.
  • Individual NK cells can survive after contacting and killing multiple target cells, possibly reducing the number of cells that need to be adoptively transferred (ie, the ex vivo stimulation and expansion of autologous or allogeneic lymphocytes, followed by reinfusion of the expanded lymphocyte population into the patient, in contrast to T cells).
  • the GD3 inhibitor is a peptide, petptidomimetic, small organic molecule, antibody, aptamers, siRNA or antisense oligonucleotide.
  • peptidomimetic refers to a small protein-like chain designed to mimic a peptide.
  • the GD3 inhibitor is an aptamer.
  • Aptamers refer to a class of molecule that represents an alternative to antibodies in term of molecular recognition.
  • Aptamers are oligonucleotide or oligopeptide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity.
  • Such ligands may be isolated through Systematic Evolution of Ligands by Exponential enrichment (SELEX) of a random sequence library, as described in Tuerk C. and Gold L., Science, 1990, 249(4968):505-10.
  • the random sequence library is obtainable by combinatorial chemical synthesis of DNA. In this library, each member is a linear oligomer, eventually chemically modified, of a unique sequence.
  • Peptide aptamers consists of a conformationally constrained antibody variable region displayed by a platform protein, such as E. coli Thioredoxin A that are selected from combinatorial libraries by two hybrid methods (Colas et al., Nature, 1996,380, 548-50).
  • the GD3 inhibitor is a small organic molecule.
  • small organic molecule refers to a molecule of a size comparable to those organic molecules generally used in pharmaceuticals.
  • Preferred small organic molecules range in size up to about 5000 Da, more preferably up to 2000 Da, and most preferably up to about 1000 Da
  • the GD3 inhibitor is a short hairpin RNA (shRNA), a small interfering RNA (siRNA) or an antisense oligonucleotide which inhibits the expression of GD3.
  • shRNA short hairpin RNA
  • siRNA small interfering RNA
  • antisense oligonucleotide which inhibits the expression of GD3.
  • the inhibitor of GD3 expression is siRNA
  • the inhibitor of GD3 expression is shRNA.
  • a short hairpin RNA is a sequence of RNA that makes a tight hairpin turn that can be used to silence gene expression via RNA interference.
  • shRNA is generally expressed using a vector introduced into cells, wherein the vector utilizes the U6 promoter to ensure that the shRNA is always expressed. This vector is usually passed on to daughter cells, allowing the gene silencing to be inherited.
  • the shRNA hairpin structure is cleaved by the cellular machinery into siRNA, which is then bound to the RNA-induced silencing complex (RISC). This complex binds to and cleaves mRNAs that match the siRNA to which it is bound.
  • RISC RNA-induced silencing complex
  • siRNA Small interfering RNA
  • siRNA small interfering RNA
  • RNAi RNA interference pathway
  • Anti-sense oligonucleotides include anti-sense RNA molecules and anti-sense DNA molecules, would act to directly block the translation of the targeted mRNA by binding thereto and thus preventing protein translation or increasing mRNA degradation, thus decreasing the level of the targeted protein, and thus activity, in a cell.
  • antisense oligonucleotides of at least about 15 bases and complementary to unique regions of the mRNA transcript sequence can be synthesized, e.g., by conventional phosphodiester techniques.
  • Methods for using antisense techniques for specifically inhibiting gene expression of genes whose sequence is known are well known in the art (e.g. see U.S. Pat. Nos. 6,566,135; 6,566,131; 6,365,354; 6,410,323; 6,107,091; 6,046,321; and 5,981,732).
  • Ribozymes can also function as an inhibitor of GD3 expression for use in the present invention.
  • Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA.
  • the mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage.
  • Engineered hairpin or hammerhead motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of mineralocorticoid receptor mRNA sequences are thereby useful within the scope of the present invention.
  • ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites, which typically include the following sequences, GUA, GUU, and GUC. Once identified, short RNA sequences of between about 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site can be evaluated for predicted structural features, such as secondary structure, that can render the oligonucleotide sequence unsuitable. The suitability of candidate targets can also be evaluated by testing their accessibility to hybridization with complementary oligonucleotides, using, e.g., ribonuclease protection assays.
  • Antisense oligonucleotides, siRNAs, shRNAs of the invention may be delivered in vivo alone or in association with a vector.
  • a "vector" is any vehicle capable of facilitating the transfer of the antisense oligonucleotide, siRNA, shRNA or ribozyme nucleic acid to the cells and typically mast cells.
  • the vector transports the nucleic acid to cells with reduced degradation relative to the extent of degradation that would result in the absence of the vector.
  • the vectors useful in the invention include, but are not limited to, plasmids, phagemids, viruses, other vehicles derived from viral or bacterial sources that have been manipulated by the insertion or incorporation of the antisense oligonucleotide, siRNA, shRNA or ribozyme nucleic acid sequences.
  • Viral vectors are a preferred type of vector and include, but are not limited to nucleic acid sequences from the following viruses: retrovirus, such as Moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rous sarcoma virus; adenovirus, adeno-associated virus; SV40-type viruses; polyoma viruses; Epstein-Barr viruses; papilloma viruses; herpes virus; vaccinia virus; polio virus; and RNA virus such as a retrovirus.
  • retrovirus such as Moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rous sarcoma virus
  • adenovirus adeno-associated virus
  • SV40-type viruses polyoma viruses
  • Epstein-Barr viruses Epstein-Barr viruses
  • papilloma viruses herpes virus
  • vaccinia virus poli
  • Non-cytopathic viruses include retroviruses (e.g., lentivirus), the life cycle of which involves reverse transcription of genomic viral RNA into DNA with subsequent proviral integration into host cellular DNA. Retroviruses have been approved for human gene therapy trials. Most useful are those retroviruses that are replicationdeficient (i.e., capable of directing synthesis of the desired proteins, but incapable of manufacturing an infectious particle). Such genetically altered retroviral expression vectors have general utility for the high-efficiency transduction of genes in vivo.
  • viruses for certain applications are the adeno-viruses and adeno-associated viruses, which are double-stranded DNA viruses.
  • the adeno-associated virus can be engineered to be replication deficient and is capable of infecting a wide range of cell types and species.
  • the adeno-associated virus can integrate into human cellular DNA in a site-specific manner, thereby minimizing the possibility of insertional mutagenesis and variability of inserted gene expression characteristic of retroviral infection.
  • wild-type adeno-associated virus infections have been followed in tissue culture for greater than 100 passages in the absence of selective pressure, implying that the adeno-associated virus genomic integration is a relatively stable event.
  • the adeno-associated virus can also function in an extrachromosomal fashion.
  • Plasmid vectors have been extensively described in the art and are well known to those of skill in the art. See e.g. Sambrook et al., 1989. In the last few years, plasmid vectors have been used as DNA vaccines for delivering antigen-encoding genes to cells in vivo. They are particularly advantageous for this because they do not have the same safety concerns as with many of the viral vectors. These plasmids, however, having a promoter compatible with the host cell, can express a peptide from a gene operatively encoded within the plasmid. These plasmids are well known to those of ordinary skill in the art.
  • Plasmids may be custom designed using restriction enzymes and ligation reactions to remove and add specific fragments of DNA. Plasmids may be delivered by a variety of parenteral, mucosal and topical routes.
  • the DNA plasmid can be injected by intramuscular, eye, intradermal, subcutaneous, intra-articular or other routes. It may also be administered by intranasal sprays or drops, rectal suppository and orally. It may also be administered into the epidermis or a mucosal surface using a gene-gun.
  • the plasmids may be given in an aqueous solution, dried onto gold particles or in association with another DNA delivery system including but not limited to liposomes, dendrimers, cochleate and microencapsulation.
  • the GD3 inhibitor is an endonuclease.
  • sequencing technologies have provided an unprecedentedly detailed overview of the multiple genetic aberrations in cancer.
  • these new data strongly emphasize the need of fast and reliable strategies to characterize the normal and pathological function of these genes and assess their role, in particular as driving factors during oncogenesis.
  • the new technologies provide the means to recreate the actual mutations observed in cancer through direct manipulation of the genome. Indeed, natural and engineered nuclease enzymes have attracted considerable attention in the recent years.
  • NHEJ error-prone nonhomologous end-joining
  • HDR high-fidelity homology-directed repair
  • the endonuclease is CRISPR-cas.
  • CRISPR-cas has its general meaning in the art and refers to clustered regularly interspaced short palindromic repeats associated which are the segments of prokaryotic DNA containing short repetitions of base sequences.
  • the endonuclease is CRISPR-cas9 which is from Streptococcus pyogenes.
  • the CRISPR/Cas9 system has been described in US 8697359 Bl and US 2014/0068797. Originally an adaptive immune system in prokaryotes (Barrangou and Marraffini, 2014), CRISPR has been recently engineered into a new powerful tool for genome editing. It has already been successfully used to target important genes in many cell lines and organisms, including human (Mali et al., 2013, Science, Vol. 339 : 823-826), bacteria (Fabre et al., 2014, PLoS Negl. Trop. Dis., Vol.
  • the endonuclease is CRISPR-Cpfl which is the more recently characterized CRISPR from Provotella and Francisella 1 (Cpfl) in Zetsche et al. (“Cpfl is a Single RNA-guided Endonuclease of a Class 2 CRISPR-Cas System (2015); Cell; 163, 1-13).
  • the GD3 inhibitor is an aptamer.
  • aptamers are DNA aptamers such as described in Prodeus et al 2015.
  • a major disadvantage of aptamers as therapeutic entities is their poor pharmacokinetic profiles, as these short DNA strands are rapidly removed from circulation due to renal filtration.
  • aptamers according to the invention are conjugated to with high molecular weight polymers such as polyethylene glycol (PEG).
  • PEG polyethylene glycol
  • the aptamer is an anti-PD-1 aptamer.
  • the anti- PD-1 aptamer is MP7 pegylated as described in Prodeus et al 2015.
  • treatment refers to both prophylactic or preventive treatment as well as curative, improving the patient’s condition 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 subject 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 subject 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 regular intervals, e.g., daily, 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., disease manifestation, etc.]).
  • the term “preventing” intends characterizing a prophylactic method or process that is aimed at delaying or preventing the onset of a disorder or condition to which such term applies.
  • administering refers to the act of injecting or otherwise physically delivering a substance as it exists outside the body (e.g. GD3 inhibitor) into the subject, such as by mucosal, intradermal, intravenous, subcutaneous, intramuscular delivery and/or any other method of physical delivery described herein or known in the art.
  • a substance as it exists outside the body (e.g. GD3 inhibitor) into the subject, such as by mucosal, intradermal, intravenous, subcutaneous, intramuscular delivery and/or any other method of physical delivery described herein or known in the art.
  • administration of the substance typically occurs after the onset of the disease or symptoms thereof.
  • administration of the substance typically occurs before the onset of the disease or symptoms thereof.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired therapeutic result.
  • a therapeutically effective amount of drug may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of drug to elicit a desired response in the individual.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody or antibody portion are outweighed by the therapeutically beneficial effects.
  • the efficient dosages and dosage regimens for drug depend on the disease or condition to be treated and may be determined by the persons skilled in the art. A physician having ordinary skill in the art may readily determine and prescribe the effective amount of the pharmaceutical composition required.
  • a suitable dose of a composition of the present invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect according to a particular dosage regimen.
  • Such an effective dose will generally depend upon the factors described above.
  • a therapeutically effective amount for therapeutic use may be measured by its ability to stabilize the progression of disease.
  • One of ordinary skill in the art would be able to determine such amounts based on such factors as the subject's size, the severity of the subject's symptoms, and the particular composition or route of administration selected.
  • An exemplary, non-limiting range for a therapeutically effective amount of drug is about 0.1-100 mg/kg, such as about 0.1- 50 mg/kg, for example about 0.1-20 mg/kg, such as about 0.1-10 mg/kg, for instance about 0.5, about such as 0.3, about 1, about 3 mg/kg, about 5 mg/kg or about 8 mg/kg.
  • Administration may e.g. be intravenous, intramuscular, intraperitoneal, or subcutaneous, and for instance administered proximal to the site of the target. Dosage regimens in the above methods of treatment and uses are adjusted to provide the optimum desired response (e.g., a therapeutic response).
  • a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation.
  • the efficacy of the treatment is monitored during the therapy, e.g. at predefined points in time.
  • treatment according to the present invention may be provided as a daily dosage of the agent of the present invention in an amount of about 0.1-100 mg/kg, such as 0.2, 0.5, 0.9, 1.0, 1.1, 1.5, 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, 40, 45, 50, 60, 70, 80, 90 or 100 mg/kg, per day, on at least one of days 1, 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, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40, or alternatively, at least one of weeks 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 after initiation of treatment, or any combination thereof, using single or divided doses every 24, 12, 8, 6, 4, or 2 hours, or any combination thereof.
  • 0.1-100 mg/kg such as 0.2, 0.5, 0.9, 1.0, 1.1, 1.5, 2, 3, 4, 5,
  • the subject is administered with a pharmaceutical composition
  • a pharmaceutical composition comprising the GD3 inhibitor as active principle and at least one pharmaceutically acceptable excipient.
  • active principle or “active ingredient” are used interchangeably.
  • the active principle is used to alleviate, treat or prevent a medical condition or disease.
  • pharmaceutically acceptable excipient it is understood a carrier medium which does not interfere with the effectiveness of the biological activity of the active ingredient(s) and which is not excessively toxic to the host at the concentration at which it is administered.
  • Said excipients are selected, depending on the pharmaceutical form and the desired method of administration, from the usual excipients known by a person skilled in the art.
  • the pharmaceutical composition of the present invention does not comprise a second active principle.
  • the term “combination” is intended to refer to all forms of administration that provide a first drug together with a further (second, third%) drug.
  • the drugs may be administered simultaneously, separately or sequentially and in any order.
  • the drug is administered to the subject using any suitable method that enables the drug to reach the chondrocytes of the bone growth plate.
  • the drug administered to the subject systemically (i.e. via systemic administration).
  • the drug is administered to the subject such that it enters the circulatory system and is distributed throughout the body.
  • the drug is administered to the subject by local administration, for example by local administration to the growing bone.
  • the terms “combined treatment”, “combined therapy” or “therapy combination” refer to a treatment that uses more than one medication.
  • the combined therapy may be dual therapy or bi-therapy.
  • administration simultaneously refers to administration of 2 active ingredients by the same route and at the same time or at substantially the same time.
  • administration separately refers to an administration of 2 active ingredients at the same time or at substantially the same time by different routes.
  • administration sequentially refers to an administration of 2 active ingredients at different times, the administration route being identical or different.
  • the GD3 inhibitor of the present invention is administered to the subject in combination with a standard treatment.
  • standard treatment of osteoarthritis is analgesics, nonsteroidal anti-inflammatory drugs, corticosteroids, hyaluronic acid injection, surgery, physical activity, weight management, etc.
  • the treatment consists of administering to the subject an immunotherapeutic agent.
  • immunotherapeutic agent refers to a compound, composition or treatment that indirectly or directly enhances, stimulates, or increases the body's immune response against cancer cells and/or that decreases the side effects of other anticancer therapies.
  • Immunotherapy is thus a therapy that directly or indirectly stimulates or enhances the immune system's responses to cancer cells and/or lessens the side effects that may have been caused by other anti-cancer agents.
  • Immunotherapy is also referred to in the art as immunologic therapy, biological therapy biological response modifier therapy and biotherapy.
  • immunotherapeutic agents include, but are not limited to, cytokines, cancer vaccines, monoclonal antibodies, and non-cytokine adjuvants.
  • the immunotherapeutic treatment may consist of administering the subject with an amount of immune cells (T cells, NK, cells, dendritic cells, B cells).
  • Immunotherapeutic agents can be non-specific, i.e. boost the immune system generally so that the human body becomes more effective in fighting the growth and/or spread of cancer cells, or they can be specific, i.e. targeted to the cancer cells themselves immunotherapy regimens may combine the use of non-specific and specific immunotherapeutic agents.
  • Non-specific immunotherapeutic agents are substances that stimulate or indirectly improve the immune system.
  • Non-specific immunotherapeutic agents have been used alone as a main therapy for the treatment of cancer, as well as in addition to a main therapy, in which case the non-specific immunotherapeutic agent functions as an adjuvant to enhance the effectiveness of other therapies (e.g. cancer vaccines).
  • Non-specific immunotherapeutic agents can also function in this latter context to reduce the side effects of other therapies, for example, bone marrow suppression induced by certain chemotherapeutic agents.
  • Non-specific immunotherapeutic agents can act on key immune system cells and cause secondary responses, such as increased production of cytokines and immunoglobulins. Alternatively, the agents can themselves comprise cytokines.
  • Non-specific immunotherapeutic agents are generally classified as cytokines or non-cytokine adjuvants.
  • cytokines have found application in the treatment of cancer either as general nonspecific immunotherapies designed to boost the immune system, or as adjuvants provided with other therapies.
  • Suitable cytokines include, but are not limited to, interferons, interleukins, and colony-stimulating factors.
  • Interferons contemplated by the present invention include the common types of IFNs, IFN-alpha (IFN-a), IFN-beta (IFN-P) and IFN-gamma (IFN-y).
  • IFNs can act directly on cancer cells, for example, by slowing their growth, promoting their development into cells with more normal behaviour and/or increasing their production of antigens thus making the cancer cells easier for the immune system to recognise and destroy.
  • IFNs can also act indirectly on cancer cells, for example, by slowing down angiogenesis, boosting the immune system and/or stimulating natural killer (NK) cells, T cells and macrophages.
  • Recombinant IFN-alpha is available commercially as Roferon (Roche Pharmaceuticals) and Intron A (Schering Corporation).
  • Interleukins contemplated by the present invention include IL-2, IL-4, IL-11 and IL-12.
  • Examples of commercially available recombinant interleukins include Proleukin® (IL-2; Chiron Corporation) and Neumega® (IL-12; Wyeth Pharmaceuticals).
  • Zymogenetics, Inc. (Seattle, Wash.) is currently testing a recombinant form of IL-21, which is also contemplated for use in the combinations of the present invention.
  • Colony-stimulating factors contemplated by the present invention include granulocyte colony stimulating factor (G-CSF or filgrastim), granulocyte-macrophage colony stimulating factor (GM-CSF or sargramostim) and erythropoietin (epoetin alfa, darbepoietin).
  • G-CSF or filgrastim granulocyte colony stimulating factor
  • GM-CSF or sargramostim granulocyte-macrophage colony stimulating factor
  • erythropoietin epoetin alfa, darbepoietin
  • G-CSF Neupogen®
  • Amgen Neulasta
  • GM-CSF Leukine
  • Berlex Procrit
  • Epogen erythropoietin
  • Amgen erythropoietin
  • Amesp erytropoietin
  • immunotherapeutic agents can be active, i.e. stimulate the body's own immune response, or they can be passive, i.e. comprise immune system components that were generated external to the body.
  • Passive specific immunotherapy typically involves the use of one or more monoclonal antibodies that are specific for a particular antigen found on the surface of a cancer cell or that are specific for a particular cell growth factor.
  • Monoclonal antibodies may be used in the treatment of cancer in a number of ways, for example, to enhance a subject's immune response to a specific type of cancer, to interfere with the growth of cancer cells by targeting specific cell growth factors, such as those involved in angiogenesis, or by enhancing the delivery of other anticancer agents to cancer cells when linked or conjugated to agents such as chemotherapeutic agents, radioactive particles or toxins.
  • the subject will be treated with a GD3 inhibitor in combination with an immune checkpoint inhibitor.
  • the therapy consists of administering to the subject an immune checkpoint inhibitor in combination with GD3 inhibitor.
  • an immune checkpoint inhibitor and ii) GD3 inhibitor as a combined preparation according to the invention for simultaneous, separate, or sequential use in the method for treating osteoarthritis in a subject.
  • ICI immune checkpoint inhibitor
  • immune checkpoint protein has its general meaning in the art and refers to a molecule that is expressed by T cells in that either turn up a signal (stimulatory checkpoint molecules) or turn down a signal (inhibitory checkpoint molecules).
  • Immune checkpoint molecules are recognized in the art to constitute immune checkpoint pathways similar to the CTLA-4 and PD-1 dependent pathways (see e.g. Pardoll, 2012. Nature Rev Cancer 12:252-264; Mellman et al. 2011. Nature 480:480- 489).
  • Examples of stimulatory checkpoint include CD27 CD28 CD40, CD122, CD137, 0X40, GITR, and ICOS.
  • inhibitory checkpoint molecules examples include A2AR, B7-H3, B7-H4, BTLA, CTLA-4, CD277, IDO, KIR, PD-1, LAG-3, TIM-3 and VISTA
  • A2AR Adenosine A2A receptor
  • B7-H4 also called VTCN1
  • B and T Lymphocyte Attenuator (BTLA) and also called CD272 has HVEM (Herpesvirus Entry Mediator) as its ligand.
  • HVEM Herpesvirus Entry Mediator
  • Surface expression of BTLA is gradually downregulated during differentiation of human CD8+ T cells from the naive to effector cell phenotype, however tumor-specific human CD8+ T cells express high levels of BTLA.
  • CTLA-4 Cytotoxic T-Lymphocyte-Associated protein 4 and also called CD152. Expression of CTLA-4 on Treg cells serves to control T cell proliferation.
  • IDO Indoleamine 2,3-dioxygenase, is a tryptophan catabolic enzyme.
  • TDO tryptophan 2,3-dioxygenase
  • IDO is known to suppress T and NK cells, generate and activate Tregs and myeloid-derived suppressor cells, and promote tumour angiogenesis.
  • KIR Killer-cell Immunoglobulin-like Receptor
  • LAG3, Lymphocyte Activation Gene-3 works to suppress an immune response by action to Tregs as well as direct effects on CD8+ T cells.
  • PD- 1 Programmed Death 1 (PD-1) receptor, has two ligands, PD-L1 and PD-L2.
  • TIM-3 short for T-cell Immunoglobulin domain and Mucin domain 3, expresses on activated human CD4+ T cells and regulates Thl and Thl7 cytokines. TIM-3 acts as a negative regulator of Thl/Tcl function by triggering cell death upon interaction with its ligand, galectin-9.
  • VISTA Short for V-domain Ig suppressor of T cell activation, VISTA is primarily expressed on hematopoietic cells so that consistent expression of VISTA on leukocytes within tumors may allow VISTA blockade to be effective across a broad range of solid tumors. Tumor cells often take advantage of these checkpoints to escape detection by the immune system. Thus, inhibiting a checkpoint protein on the immune system may enhance the anti -tumor T-cell response.
  • an immune checkpoint inhibitor refers to any compound inhibiting the function of an immune checkpoint protein. Inhibition includes reduction of function and full blockade.
  • the immune checkpoint inhibitor could be an antibody, synthetic or native sequence peptides, small molecules or aptamers which bind to the immune checkpoint proteins and their ligands.
  • the immune checkpoint inhibitor is an antibody.
  • antibodies are directed against A2AR, B7-H3, B7-H4, BTLA, CTLA-4, CD277, IDO, KIR, PD-1, LAG-3, TIM-3 or VISTA
  • the immune checkpoint inhibitor is an anti-PD-1 antibody such as described in WO2011082400, W02006121168, W02015035606, W02004056875, W02010036959, W02009114335, W02010089411, WO2008156712, WO2011110621, WO2014055648 and WO2014194302.
  • anti-PD-1 antibodies which are commercialized: Nivolumab (Opdivo®, BMS), Pembrolizumab (also called Lambrolizumab, KEYTRUDA® or MK-3475, MERCK).
  • the immune checkpoint inhibitor is an anti-PD-Ll antibody such as described in WO2013079174, W02010077634, W02004004771, WO2014195852, W02010036959, WO2011066389, W02007005874, W02015048520, US8617546 and WO2014055897.
  • anti-PD-Ll antibodies which are on clinical trial: Atezolizumab (MPDL3280A, Genentech/Roche), Durvalumab (AZD9291, AstraZeneca), Avelumab (also known as MSB0010718C, Merck) and BMS-936559 (BMS).
  • the immune checkpoint inhibitor is an anti-PD-L2 antibody such as described in US7709214, US7432059 and US8552154.
  • the immune checkpoint inhibitor inhibits Tim-3 or its ligand.
  • the immune checkpoint inhibitor is an anti-Tim-3 antibody such as described in WO03063792, WO2011155607, WO2015117002, WO2010117057 and WO2013006490.
  • the immune checkpoint inhibitor is a small organic molecule.
  • small organic molecule refers to a molecule of a size comparable to those organic molecules generally used in pharmaceuticals.
  • small organic molecules range in size up to about 5000 Da, more preferably up to 2000 Da, and most preferably up to about 1000 Da.
  • the small organic molecules interfere with transduction pathway of A2AR, B7-H3, B7-H4, BTLA, CTLA-4, CD277, IDO, KIR, PD-1, LAG-3, TIM-3 or VISTA.
  • small organic molecules interfere with transduction pathway of PD-1 and Tim-3.
  • they can interfere with molecules, receptors or enzymes involved in PD-1 and Tim-3 pathway.
  • the small organic molecules interfere with Indoleamine-pyrrole 2, 3 -di oxygenase (IDO) inhibitor.
  • IDO is involved in the tryptophan catabolism (Liu et al 2010, Vacchelli et al 2014, Zhai et al 2015). Examples of IDO inhibitors are described in WO 2014150677.
  • IDO inhibitors include without limitation 1-methyl-tryptophan (IMT), p- (3-benzofuranyl)-alanine, P-(3-benzo(b)thienyl)-alanine), 6-nitro-tryptophan, 6- fluoro-tryptophan, 4-methyl-tryptophan, 5 -methyl tryptophan, 6-methyl-tryptophan, 5- methoxy-tryptophan, 5 -hydroxy-tryptophan, indole 3-carbinol, 3,3'- diindolylmethane, epigallocatechin gallate, 5-Br-4-Cl-indoxyl 1,3-diacetate, 9- vinylcarbazole, acemetacin, 5- bromo-tryptophan, 5 -bromoindoxyl diacetate, 3- Amino-naphtoic acid, pyrrolidine dithiocarbamate, 4-phenylimidazole a brassinin derivative, a thiohydanto
  • the IDO inhibitor is selected from 1-methyl-tryptophan, p-(3- benzofuranyl)-alanine, 6-nitro-L-tryptophan, 3- Amino-naphtoic acid and P-[3- benzo(b)thienyl] -alanine or a derivative or prodrug thereof.
  • the inhibitor of IDO is Epacadostat, (INCB24360, INCB024360) has the following chemical formula in the art and refers to -N-(3-bromo-4-fluorophenyl)-N'- hydroxy-4- ⁇ [2-(sulfamoylamino)-ethyl]amino ⁇ -l,2,5-oxadiazole-3 carboximidamide :
  • the inhibitor is BGB324, also called R428, such as described in W02009054864, refers to lH-l,2,4-Triazole-3,5-diamine, l-(6,7-dihydro-5H- benzo[6,7]cyclohepta[l,2-c]pyridazin-3-yl)-N3-[(7S)-6,7,8,9-tetrahydro-7-(l-pyrrolidinyl)- 5H-benzocyclohepten-2-yl]- and has the following formula in the art:
  • the inhibitor is CA-170 (or AUPM-170): an oral, small molecule immune checkpoint antagonist targeting programmed death ligand-1 (PD-L1) and V-domain Ig suppressor of T cell activation (VISTA) (Liu et al 2015).
  • PD-170 or AUPM-170
  • VISTA V-domain Ig suppressor of T cell activation
  • the immune checkpoint inhibitor is an aptamer.
  • the aptamers are directed against A2AR, B7-H3, B7-H4, BTLA, CTLA-4, CD277, IDO, KIR, PD-1, LAG-3, TIM-3 or VISTA.
  • aptamers are DNA aptamers such as described in Prodeus et al 2015.
  • a major disadvantage of aptamers as therapeutic entities is their poor pharmacokinetic profiles, as these short DNA strands are rapidly removed from circulation due to renal filtration.
  • aptamers according to the invention are conjugated to with high molecular weight polymers such as polyethylene glycol (PEG).
  • PEG polyethylene glycol
  • the aptamer is an anti-PD-1 aptamer.
  • the anti-PD-1 aptamer is MP7 pegylated as described in Prodeus et al 2015.
  • the GD3 inhibitor as described above are administered to the subject in the form of a pharmaceutical composition which comprises a pharmaceutically acceptable carrier.
  • pharmaceutically or “pharmaceutically acceptable” refers to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to a mammal, especially a human, as appropriate.
  • compositions include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, di sodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene- block polymers, polyethylene glycol and wool fat.
  • ion exchangers alumina, aluminum stearate, lecithin
  • serum proteins such as human serum albumin
  • buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial gly
  • compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • the used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques.
  • Sterile injectable forms of the compositions of this invention may be aqueous or an oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3 -butanediol.
  • a non-toxic parenterally acceptable diluent or solvent for example as a solution in 1,3 -butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono-or di glycerides.
  • Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • Other commonly used surfactants such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
  • compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions.
  • carriers commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added
  • useful diluents include, e g., lactose.
  • aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring, or coloring agents may also be added.
  • the compositions of this invention may be administered in the form of suppositories for rectal administration.
  • compositions of this invention may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
  • the compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers.
  • Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
  • the compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2- octyldodecanol, benzyl alcohol and water.
  • Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Patches may also be used.
  • the compositions of this invention may also be administered by nasal aerosol or inhalation.
  • Such compositions are prepared according to techniques well- known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
  • an antibody present in a pharmaceutical composition of this invention can be supplied at a concentration of 10 mg/mL in either 100 mg (10 mL) or 500 mg (50 mL) single-use vials.
  • the product is formulated for IV administration in 9.0 mg/mL sodium chloride, 7.35 mg/mL sodium citrate dihydrate, 0.7 mg/mL polysorbate 80, and Sterile Water for Injection.
  • the pH is adjusted to 6.5.
  • An exemplary suitable dosage range for an antibody in a pharmaceutical composition of this invention may between about 1 mg/m 2 and 500 mg/m 2
  • these schedules are exemplary and that an optimal schedule and regimen can be adapted taking into account the affinity and tolerability of the particular antibody in the pharmaceutical composition that must be determined in clinical trials.
  • a pharmaceutical composition of the invention for injection (e.g., intramuscular, i.v.) could be prepared to contain sterile buffered water (e.g. 1 ml for intramuscular), and between about 1 ng to about 100 mg, e g about 50 ng to about 30 mg or more preferably, about 5 mg to about 25 mg, of the inhibitor of the invention.
  • sterile buffered water e.g. 1 ml for intramuscular
  • FIGURES
  • Figure 1 Effect of intra-articular anti-GD3 monoclonal antibody injection in an experimental murine OA model.
  • C-F) Analysis of the bone volume, ratio bone surface/bone volume (BS/BV), bone thickness and ratio bone surface/tissue volume (BS/TV) between isotype knee and a-GD3 knee (N 10)
  • P-values are calculated by the Mann-Whitney test (unpaired t test, two-tailed).
  • Figure 2 Effect of intra-peritoneal injection of anti-GD3 monoclonal antibodies (R24) prevents bone remodeling in spontaneous age-related OA murine model.
  • R24 anti-GD3 monoclonal antibodies
  • Intra-articular injections of anti-GD3 (clone R24) or isotype (anti-IgG3) were performed in mice following the same procedure: 30 microgrammes in PBS of anti-GD3, or 30 microgrammes in PBS of isotype in the left knee at day 7, followed by a second injection 3 days later.
  • intra-peritoneal injections of anti-GD3 (clone R24) or isotype (anti-IgG3) were performed in mice: 150 microgrammes in PBS of anti-GD3, or 150 microgrammes in PBS of isotype, six times every 2 weeks.
  • mice were fixed in 4% paraformaldehyde at 4°C for 48h, washed in PBS, and then processed for routine histology. Knees were decalcified in 14% EDTA/PBS for three weeks and then paraffin-embedded. Tissue sections (5 pm) were rehydrated through a gradient of ethanol and xylene. Sections were then stained with Safranin-O/Fast Green solution to evaluate cartilage degradation.
  • Cartilage damage was analyzed using an arbitrary score of 0-30, based on the OARSI cartilage OA grading system histopathology, and modified by van den Berg for the assessment of murine knee joints (grading scale of 0-6 for the severity of cartilage destruction and of 0-5 for the extent of damaged cartilage surface).
  • Sections were embedded in paraffin and cut into sections (5 pm). The sections were deparaffinized in xylene and rehydrated in ethanol. Then, tissue sections were incubated with pepsin (DAKO, 40 mh/mL) for 30 min for antigen retrieval, permeabilized with 0,3% Triton X-100 PBS/BSA 3% during 3 min. 1% H202 for 10 min followed by incubation in PBS/BSA 3% for 30 min.
  • pepsin DAKO, 40 mh/mL
  • Hind leg knee were dissected to remove smooth tissues and scanned in a micro-CT scanner SkyScan 1176 (Bruker, Belgium, 0.5 mm aluminum filter, 45 kV, 500 pA, 18 pm resolution, 0.5° rotation angle). Scans were reconstructed using CTAn vl.9, Nrecon vl.6 (Bruker, Belgium) and a three-dimensional (3D) model visualization software program (CTVol v2.0). Misalignment compensation, ring artifacts and beam -hardening were adjusted to obtain the correct re-construction of each paw. Bone degradation was quantified in subchondral bone and the epiphysis region of the medial and/or lateral plateau for each tibia (CTAn software, Bruker, Belgium). Reconstructed 3D images of joints were obtained using the Avizo software (Avizo Lite 9.3.0, FEI, France).
  • Articular cartilage of tibia medial plateau was scanned through their depth in XYZ-mode, with a confocal laser scanning microscope (CLSM; TCS SP5-II, Leica Microsystems, Nanterre, France) with a voxel size of 6 pm, a 5* dry objective and a UVlaser light source (I'A 405 nm). Stacks of images were then done and analyzed to quantitatively evaluate several parameters of articular cartilage. Assessment of cartilage morphometric parameters was performed in medial plateau of each tibia using Avizo software (FEI Visualization Sciences Group, Lyon).
  • RNA quality was checked by spectral analysis (A260/ 280 nm), and then samples were stored at -80°C.
  • Reverse transcription was performed using the M-MLV reverse transcriptase (Invitrogen; 28025013; 5U/pL final concentration), 500ng total RNA, a random hexamer primer (Thermo Scientific, GER; SO 142; lOng/pL final concentration), and dNTPs (Roche, CH; 1 277 049; 5mM final concentration) in M-MLV reverse transcriptase buffer (Invitrogen; 18057-018), for a total volume of 20pL.
  • M-MLV reverse transcriptase Invitrogen; 28025013; 5U/pL final concentration
  • 500ng total RNA 500ng total RNA
  • a random hexamer primer Thermo Scientific, GER; SO 142; lOng/pL final concentration
  • dNTPs Roche, CH; 1 277 049
  • SYBR Green-based quantitative PCR was performed using the LightCycle® 480 SYBR Green I master reaction mix (Roche, 04707516001), 10 ng of cDNA, and the LightCycler 480 real-time PCR system (Roche) (40 cycles of amplification).
  • Raw data Ct values
  • Gene expression data were calculated as relative to the expression of housekeeping gene, Rps9 (2 A -deltaCT method).
  • GD3 expression correlates with p!6INK4a positive senescent cells and increases with cartilage degeneration in OA patients
  • GD3 a proposed senescence hallmark, in OA cartilage patients.
  • GD3 expression correlates with p21 CIP1 andpl5 INK4b positive senescent cells and increases with cartilage degeneration in induced OA murine model

Abstract

L'ostéoarthrose (OA) est un type de maladie articulaire dégénérative qui résulte de la décomposition du cartilage articulaire et de l'os sous-jacent. Ici, les inventeurs ont étudié les effets de l'injection d'anticorps monoclonal anti-GD3 intra-articulaire. La présente invention concerne une méthode de traitement de l'arthrose chez un sujet en ayant besoin, comprenant l'administration audit sujet d'une quantité thérapeutiquement efficace d'un inhibiteur de GD3.
PCT/EP2023/079262 2022-10-21 2023-10-20 Méthodes et compositions pharmaceutiques pour le traitement de l'arthrose WO2024084034A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP22306604.4 2022-10-21
EP22306604 2022-10-21

Publications (1)

Publication Number Publication Date
WO2024084034A1 true WO2024084034A1 (fr) 2024-04-25

Family

ID=84358456

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2023/079262 WO2024084034A1 (fr) 2022-10-21 2023-10-20 Méthodes et compositions pharmaceutiques pour le traitement de l'arthrose

Country Status (1)

Country Link
WO (1) WO2024084034A1 (fr)

Citations (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
EP0368684A1 (fr) 1988-11-11 1990-05-16 Medical Research Council Clonage de séquences d'immunoglobulines de domaines variables.
EP0404097A2 (fr) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Récepteurs mono- et oligovalents, bispécifiques et oligospécifiques, ainsi que leur production et application
WO1993011161A1 (fr) 1991-11-25 1993-06-10 Enzon, Inc. Proteines multivalentes de fixation aux antigenes
US5981732A (en) 1998-12-04 1999-11-09 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-13 expression
US6046321A (en) 1999-04-09 2000-04-04 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-i1 expression
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6107091A (en) 1998-12-03 2000-08-22 Isis Pharmaceuticals Inc. Antisense inhibition of G-alpha-16 expression
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6365354B1 (en) 2000-07-31 2002-04-02 Isis Pharmaceuticals, Inc. Antisense modulation of lysophospholipase I expression
US6410323B1 (en) 1999-08-31 2002-06-25 Isis Pharmaceuticals, Inc. Antisense modulation of human Rho family gene expression
US6566131B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of Smad6 expression
US6566135B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of caspase 6 expression
WO2003063792A2 (fr) 2002-01-30 2003-08-07 The Brigham And Women's Hospital, Inc. Compositions et methodes associees a tim-3, molecule de surface cellulaire specifique a th1
WO2004004771A1 (fr) 2002-07-03 2004-01-15 Ono Pharmaceutical Co., Ltd. Compositions immunostimulantes
WO2004056875A1 (fr) 2002-12-23 2004-07-08 Wyeth Anticorps anti pd-1 et utilisations
US6982321B2 (en) 1986-03-27 2006-01-03 Medical Research Council Altered antibodies
WO2006003388A2 (fr) 2004-06-30 2006-01-12 Domantis Limited Compositions et procedes pour le traitement de troubles inflammatoires
WO2006030220A1 (fr) 2004-09-17 2006-03-23 Domantis Limited Compositions monovalentes pour la liaison au cd40l et procedes d'utilisation
US7087409B2 (en) 1997-12-05 2006-08-08 The Scripps Research Institute Humanization of murine antibody
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)
US7432059B2 (en) 2000-06-28 2008-10-07 Dana-Farber Cancer Institute, Inc. Methods of identifying compounds that upmodulate T cell activation in the presence of a PD-1 mediated signal
WO2008156712A1 (fr) 2007-06-18 2008-12-24 N. V. Organon Anticorps dirigés contre le récepteur humain de mort programmée pd-1
WO2009054864A1 (fr) 2007-10-26 2009-04-30 Rigel Pharmaceuticals, Inc. Triazoles substitués par aryle polycyclique et triazoles substitués par hétéroaryle polycyclique utiles comme inhibiteurs d'axl
WO2009114335A2 (fr) 2008-03-12 2009-09-17 Merck & Co., Inc. Protéines de liaison avec pd-1
WO2010036959A2 (fr) 2008-09-26 2010-04-01 Dana-Farber Cancer Institute Anticorps anti-pd-1, pd-l1, et pd-l2 humains et leurs utilisations
WO2010077634A1 (fr) 2008-12-09 2010-07-08 Genentech, Inc. Anticorps anti-pd-l1 et leur utilisation pour améliorer la fonction des lymphocytes t
WO2010089411A2 (fr) 2009-02-09 2010-08-12 Universite De La Mediterranee Anticorps pd-1 et anticorps pd-l1 et leurs utilisations
WO2010117057A1 (fr) 2009-04-10 2010-10-14 協和発酵キリン株式会社 Procédé pour le traitement d'une tumeur sanguine utilisant un anticorps anti-tim-3
WO2011066389A1 (fr) 2009-11-24 2011-06-03 Medimmmune, Limited Agents de liaison ciblés dirigés contre b7-h1
WO2011082400A2 (fr) 2010-01-04 2011-07-07 President And Fellows Of Harvard College Modulateurs du récepteur immunosuppresseur pd-1 et procédés d'utilisation de ceux-ci
WO2011110621A1 (fr) 2010-03-11 2011-09-15 Ucb Pharma, S.A. Produits biologiques : anticorps anti-pd-1 agonistes humanisés
WO2011155607A1 (fr) 2010-06-11 2011-12-15 協和発酵キリン株式会社 Anticorps anti-tim-3
WO2013006490A2 (fr) 2011-07-01 2013-01-10 Cellerant Therapeutics, Inc. Anticorps se liant spécifiquement à tim3
WO2013079174A1 (fr) 2011-11-28 2013-06-06 Merck Patent Gmbh Anticorps anti-pd-l1 et utilisations associées
US8617546B2 (en) 2008-10-02 2013-12-31 Seoul National University Industry Foundation Anticancer agent comprising anti-PD-1 antibody or anti-PD-L1 antibody
US20140068797A1 (en) 2012-05-25 2014-03-06 University Of Vienna Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
WO2014055648A1 (fr) 2012-10-02 2014-04-10 Bristol-Myers Squibb Company Combinaison d'anticorps anti-kir et d'anticorps anti-pd-1 pour le traitement du cancer
WO2014055897A2 (fr) 2012-10-04 2014-04-10 Dana-Farber Cancer Institute, Inc. Anticorps monoclonaux humains anti pd-l1 et procédés d'utilisation
US8697359B1 (en) 2012-12-12 2014-04-15 The Broad Institute, Inc. CRISPR-Cas systems and methods for altering expression of gene products
WO2014150677A1 (fr) 2013-03-15 2014-09-25 Bristol-Myers Squibb Company Inhibiteurs de l'indoléamine 2,3-dioxygénase (ido)
WO2014194302A2 (fr) 2013-05-31 2014-12-04 Sorrento Therapeutics, Inc. Protéines de liaison à l'antigène qui se lient à pd-1
WO2014195852A1 (fr) 2013-06-03 2014-12-11 Glaxosmithkline Intellectual Property (No.2) Limited Combinaisons d'un anticorps anti-pd-l1 et d'un inhibiteur de mek et/ou d'un inhibiteur de braf
WO2015035606A1 (fr) 2013-09-13 2015-03-19 Beigene, Ltd. Anticorps anti-pd1 et leur utilisation comme produits thérapeutiques et produits de diagnostic
WO2015048520A1 (fr) 2013-09-27 2015-04-02 Genentech, Inc. Formulations d'anticorps anti-pdl1
WO2015117002A1 (fr) 2014-01-31 2015-08-06 Novartis Ag Molécules d'anticorps anti-tim-3 et leurs utilisations
WO2022223791A1 (fr) * 2021-04-23 2022-10-27 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés et compositions pour le traitement des maladies liées à l'accumulation de sénescence cellulaire

Patent Citations (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US6982321B2 (en) 1986-03-27 2006-01-03 Medical Research Council Altered antibodies
EP0368684A1 (fr) 1988-11-11 1990-05-16 Medical Research Council Clonage de séquences d'immunoglobulines de domaines variables.
EP0404097A2 (fr) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Récepteurs mono- et oligovalents, bispécifiques et oligospécifiques, ainsi que leur production et application
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1993011161A1 (fr) 1991-11-25 1993-06-10 Enzon, Inc. Proteines multivalentes de fixation aux antigenes
US7087409B2 (en) 1997-12-05 2006-08-08 The Scripps Research Institute Humanization of murine antibody
US6107091A (en) 1998-12-03 2000-08-22 Isis Pharmaceuticals Inc. Antisense inhibition of G-alpha-16 expression
US5981732A (en) 1998-12-04 1999-11-09 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-13 expression
US6046321A (en) 1999-04-09 2000-04-04 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-i1 expression
US6410323B1 (en) 1999-08-31 2002-06-25 Isis Pharmaceuticals, Inc. Antisense modulation of human Rho family gene expression
US7709214B2 (en) 2000-06-28 2010-05-04 Dana-Farber Cancer Institute, Inc. Methods for upregulating an immune response with agents that inhibit the intereaction between PD-L2 and PD-1
US7432059B2 (en) 2000-06-28 2008-10-07 Dana-Farber Cancer Institute, Inc. Methods of identifying compounds that upmodulate T cell activation in the presence of a PD-1 mediated signal
US6365354B1 (en) 2000-07-31 2002-04-02 Isis Pharmaceuticals, Inc. Antisense modulation of lysophospholipase I expression
US6566135B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of caspase 6 expression
US6566131B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of Smad6 expression
WO2003063792A2 (fr) 2002-01-30 2003-08-07 The Brigham And Women's Hospital, Inc. Compositions et methodes associees a tim-3, molecule de surface cellulaire specifique a th1
WO2004004771A1 (fr) 2002-07-03 2004-01-15 Ono Pharmaceutical Co., Ltd. Compositions immunostimulantes
WO2004056875A1 (fr) 2002-12-23 2004-07-08 Wyeth Anticorps anti pd-1 et utilisations
WO2006003388A2 (fr) 2004-06-30 2006-01-12 Domantis Limited Compositions et procedes pour le traitement de troubles inflammatoires
WO2006030220A1 (fr) 2004-09-17 2006-03-23 Domantis Limited Compositions monovalentes pour la liaison au cd40l et procedes d'utilisation
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)
WO2008156712A1 (fr) 2007-06-18 2008-12-24 N. V. Organon Anticorps dirigés contre le récepteur humain de mort programmée pd-1
WO2009054864A1 (fr) 2007-10-26 2009-04-30 Rigel Pharmaceuticals, Inc. Triazoles substitués par aryle polycyclique et triazoles substitués par hétéroaryle polycyclique utiles comme inhibiteurs d'axl
WO2009114335A2 (fr) 2008-03-12 2009-09-17 Merck & Co., Inc. Protéines de liaison avec pd-1
US8552154B2 (en) 2008-09-26 2013-10-08 Emory University Anti-PD-L1 antibodies and uses therefor
WO2010036959A2 (fr) 2008-09-26 2010-04-01 Dana-Farber Cancer Institute Anticorps anti-pd-1, pd-l1, et pd-l2 humains et leurs utilisations
US8617546B2 (en) 2008-10-02 2013-12-31 Seoul National University Industry Foundation Anticancer agent comprising anti-PD-1 antibody or anti-PD-L1 antibody
WO2010077634A1 (fr) 2008-12-09 2010-07-08 Genentech, Inc. Anticorps anti-pd-l1 et leur utilisation pour améliorer la fonction des lymphocytes t
WO2010089411A2 (fr) 2009-02-09 2010-08-12 Universite De La Mediterranee Anticorps pd-1 et anticorps pd-l1 et leurs utilisations
WO2010117057A1 (fr) 2009-04-10 2010-10-14 協和発酵キリン株式会社 Procédé pour le traitement d'une tumeur sanguine utilisant un anticorps anti-tim-3
WO2011066389A1 (fr) 2009-11-24 2011-06-03 Medimmmune, Limited Agents de liaison ciblés dirigés contre b7-h1
WO2011082400A2 (fr) 2010-01-04 2011-07-07 President And Fellows Of Harvard College Modulateurs du récepteur immunosuppresseur pd-1 et procédés d'utilisation de ceux-ci
WO2011110621A1 (fr) 2010-03-11 2011-09-15 Ucb Pharma, S.A. Produits biologiques : anticorps anti-pd-1 agonistes humanisés
WO2011155607A1 (fr) 2010-06-11 2011-12-15 協和発酵キリン株式会社 Anticorps anti-tim-3
WO2013006490A2 (fr) 2011-07-01 2013-01-10 Cellerant Therapeutics, Inc. Anticorps se liant spécifiquement à tim3
WO2013079174A1 (fr) 2011-11-28 2013-06-06 Merck Patent Gmbh Anticorps anti-pd-l1 et utilisations associées
US20140068797A1 (en) 2012-05-25 2014-03-06 University Of Vienna Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
WO2014055648A1 (fr) 2012-10-02 2014-04-10 Bristol-Myers Squibb Company Combinaison d'anticorps anti-kir et d'anticorps anti-pd-1 pour le traitement du cancer
WO2014055897A2 (fr) 2012-10-04 2014-04-10 Dana-Farber Cancer Institute, Inc. Anticorps monoclonaux humains anti pd-l1 et procédés d'utilisation
US8697359B1 (en) 2012-12-12 2014-04-15 The Broad Institute, Inc. CRISPR-Cas systems and methods for altering expression of gene products
WO2014150677A1 (fr) 2013-03-15 2014-09-25 Bristol-Myers Squibb Company Inhibiteurs de l'indoléamine 2,3-dioxygénase (ido)
WO2014194302A2 (fr) 2013-05-31 2014-12-04 Sorrento Therapeutics, Inc. Protéines de liaison à l'antigène qui se lient à pd-1
WO2014195852A1 (fr) 2013-06-03 2014-12-11 Glaxosmithkline Intellectual Property (No.2) Limited Combinaisons d'un anticorps anti-pd-l1 et d'un inhibiteur de mek et/ou d'un inhibiteur de braf
WO2015035606A1 (fr) 2013-09-13 2015-03-19 Beigene, Ltd. Anticorps anti-pd1 et leur utilisation comme produits thérapeutiques et produits de diagnostic
WO2015048520A1 (fr) 2013-09-27 2015-04-02 Genentech, Inc. Formulations d'anticorps anti-pdl1
WO2015117002A1 (fr) 2014-01-31 2015-08-06 Novartis Ag Molécules d'anticorps anti-tim-3 et leurs utilisations
WO2022223791A1 (fr) * 2021-04-23 2022-10-27 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés et compositions pour le traitement des maladies liées à l'accumulation de sénescence cellulaire

Non-Patent Citations (20)

* Cited by examiner, † Cited by third party
Title
"Uniprot", Database accession no. Q92185
COLAS ET AL., NATURE, vol. 380, 1996, pages 548 - 50
DICARLO ET AL., NUCLEIC ACIDS RES., vol. 41, 2013, pages 4336 - 4343
FABRE ET AL., PLOS NEGL. TROP. DIS., vol. 8, 2014, pages e2671
GRATZ ET AL., GENETICS, DOI: 10. 1534/GENETICS. 113.160713, 2014
GUO ET AL., DEVELOPMENT, vol. 141, 2014, pages 707 - 714
HAI ET AL., CELL RES. DOI: 10.1038/CR.2014.11, 2014
HWANG ET AL., PLOS ONE, vol. 8, 2013, pages e68708
ILTIS CHARLÈNE ET AL: "Rôle de l'immunosurveillance des cellules sénescentes dans le cancer et le vieillissement", 25 May 2020 (2020-05-25), pages 1 - 184, XP093037210, Retrieved from the Internet <URL:https://www.theses.fr/2020COAZ6002.pdf> [retrieved on 20230404] *
JAYASENA S.D., CLIN. CHEM., vol. 45, no. 9, 1999, pages 1628 - 50
KAZUYASU NAKAMURA ET AL: "Construction of humanized anti-ganglioside monoclonal antibodies with potent immune effector functions", CANCER IMMUNOLOGY, IMMUNOTHERAPY, vol. 50, no. 5, 1 July 2001 (2001-07-01), pages 275 - 284, XP055190499, ISSN: 0340-7004, DOI: 10.1007/PL00006689 *
MA ET AL., CELL RES., vol. 24, 2014, pages 122 - 125
MALI ET AL., SCIENCE, vol. 339, 2013, pages 823 - 826
MASHIKO ET AL., DEV. GROWTH DIFFER., vol. 56, 2014, pages 122 - 129
MELLMAN ET AL., NATURE, vol. 480, 2011, pages 480 - 489
NIU ET AL., CELL, vol. 156, 2014, pages 836 - 843
PARDOLL, NATURE REV CANCER, vol. 12, 2012, pages 252 - 264
TUERK C.GOLD L., SCIENCE, vol. 249, no. 4968, 1990, pages 505 - 10
YANG ET AL., J. MOL. CELL BIOL., vol. 6, 2014, pages 97 - 99
ZETSCHE ET AL.: "Cpf1 is a Single RNA-guided Endonuclease of a Class 2 CRISPR-Cas System", CELL, vol. 163, 2015, pages 1 - 13

Similar Documents

Publication Publication Date Title
US20230374503A1 (en) Combination
US9186373B2 (en) SiRNA against Cbl-b and optionally IL-2 and IL-12 for use in the treatment of cancer
EP3368689A2 (fr) Compositions et méthodes d&#39;évaluation et de modulation des réponses immunitaires à l&#39;aide de signatures génétiques de cellules immunitaires
JP2022518207A (ja) 酸化コレステロールを含有する薬物送達システム
CN114525279A (zh) C/EBP α SARNA组合物和使用方法
AU2017331275A1 (en) Modulating responses to checkpoint inhibitor therapy
US20210253731A1 (en) Compositions and methods for preventing or reversing t-cell exhaustion through ectonucleotidase inhibition and antibody-mediated target cytosis
JP2022188159A (ja) 二本鎖ポリリボヌクレオチドとポリアルキレンイミンとの複合体を含む粒子を含む新規な医薬組成物
JP2018524588A (ja) 骨髄由来サプレッサー細胞中のテトラスパニン33(Tspan33)を標的化するがん療法
JP2024020338A (ja) 白斑を処置するための方法及び組成物
WO2018084168A1 (fr) Agent de traitement de la fibrose cutanée
EP4326903A1 (fr) Procédés et compositions pour le traitement des maladies liées à l&#39;accumulation de sénescence cellulaire
WO2024084034A1 (fr) Méthodes et compositions pharmaceutiques pour le traitement de l&#39;arthrose
WO2019222500A1 (fr) Procédés de modulation de l&#39;activité d&#39;un dinucléotide cyclique (cdn) à l&#39;aide d&#39;un agent de modulation du transporteur cdn
BR112020013285A2 (pt) direcionamento do complexo de sinalossoma cbm que induz células t reguladoras a atingirem o microambiente tumoral
US20220354811A1 (en) Methods and compositions for modulating macrophages polarization
US20230043806A1 (en) Induction of highly efficacious anti-tumor and immune modulating activity: cell-free off the shelf therapeutic modality
US20240158861A1 (en) Methods and compositions for treating cell senescence accumulation related disease
US20220305100A1 (en) Methods of vaccination and use of cd47 blockade
WO2023183822A1 (fr) Perte hématopoïétique du chromosome y provoquant une fibrose cardiaque et un dysfonctionnement et étant associée à la mort causée par une insuffisance cardiaque
TW202333802A (zh) 用於肺癌之治療性rna(二)
JPWO2020027217A1 (ja) がんの治療及び/又は予防のための医薬