WO2017212021A1 - Méthodes et compositions pharmaceutiques pour le traitement du cancer - Google Patents

Méthodes et compositions pharmaceutiques pour le traitement du cancer Download PDF

Info

Publication number
WO2017212021A1
WO2017212021A1 PCT/EP2017/064093 EP2017064093W WO2017212021A1 WO 2017212021 A1 WO2017212021 A1 WO 2017212021A1 EP 2017064093 W EP2017064093 W EP 2017064093W WO 2017212021 A1 WO2017212021 A1 WO 2017212021A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
tumor
cell
leukemia
carcinoma
Prior art date
Application number
PCT/EP2017/064093
Other languages
English (en)
Inventor
Renato Monteiro
Sanae BEN MKADDEM
Eric DAUGAS
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Université Paris Diderot - Paris 7
Centre National De La Recherche Scientifique (Cnrs)
Assistance Publique-Hôpitaux De Paris (Aphp)
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), Université Paris Diderot - Paris 7, Centre National De La Recherche Scientifique (Cnrs), Assistance Publique-Hôpitaux De Paris (Aphp) filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2017212021A1 publication Critical patent/WO2017212021A1/fr

Links

Classifications

    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/10Protein-tyrosine kinases (2.7.10)
    • C12Y207/10002Non-specific protein-tyrosine kinase (2.7.10.2), i.e. spleen tyrosine kinase

Definitions

  • the present invention relates to methods and pharmaceutical compositions for the treatment of cancer.
  • T cells reactive against tumor-associated antigens have been detected in the blood of patients with many different types of cancers, suggesting a role for the immune system in fighting cancer.
  • Innate and adaptive immunity maintains effector cells such as lymphocytes and natural killer cells that distinguish normal cells from "modified” cells as in the case of tumor cells.
  • effector cells such as lymphocytes and natural killer cells that distinguish normal cells from "modified” cells as in the case of tumor cells.
  • the immune system is controlled by a finely tuned network of regulatory mechanism (1).
  • certains immunoreceptors have been shown to exert inhibitory and activating signal through ITAM motif (Yxx[L/I]x6-sYxx[L/I]) (2) depending on the valency of their respective ligand.
  • the ITAM motif is found in the cytoplasmic domain of several transmembrane adapter molecules, such as the common ⁇ subunit of FcR (FcRy), the Iga and Ig subunits of the BCR, and the ⁇ , ⁇ , ⁇ and ⁇ subunits of the TCR-associated CD3 complex (1, 2), and in the cytoplasmic tail of other receptors, such as the FcyRIIA (11).
  • FcRy FcR
  • FcRy FcR
  • Iga and Ig subunits of the BCR and the ⁇ , ⁇ , ⁇ and ⁇ subunits of the TCR-associated CD3 complex (1, 2)
  • FcyRIIA FcyRIIA
  • the present invention relates to methods and pharmaceutical compositions for the treatment cancer.
  • the present invention is defined by the claims.
  • Immunoreceptors play a crucial role in the regulation of immune homeostasis and inflammation. Depending on ligand valency they can promote either activating or inhibitory signals.
  • the inventors investigated how these receptors translate outside ligand interactions into opposite signals.
  • the Src-family kinase Fyn was the crucial effector for inhibition of SHP-1, while Lyn/Lck were required for its activation downstream of Fc, B-cell and T-cell antigen receptors.
  • a Fyn-PI3K-PKC axis turns off Lyn-mediated SHP-1 activation by shifting tyrosine into serine phosphorylation.
  • the first object of the present invention relates to a method of treating an autoimmune inflammatory disease in a subject in thereof comprising administering to the subject a therapeutically effective amount of a Lyn/Lck inhibitor.
  • treatment or “treat” refer 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 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.
  • 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.
  • loading regimen 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.
  • the phrase "maintenance regimen” or “maintenance period” 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 intervals, 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., disease manifestation, etc.]).
  • continuous therapy e.g., administering a drug at a regular intervals, e.g., weekly, monthly, yearly, etc.
  • 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.]).
  • 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 be 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, gastrointestinal tract, gum, head, kidney, liver, lung, nasopharynx, neck, ovary, prostate, skin, stomach, testis, tongue, or uterus.
  • the subject suffers from a cancer selected from the group consisting of Acanthoma, Acinic cell carcinoma, Acoustic neuroma, Acral lentiginous melanoma, Acrospiroma, Acute eosinophilic leukemia, Acute lymphoblastic leukemia, Acute megakaryoblastic leukemia, Acute monocytic leukemia, Acute myeloblastic leukemia with maturation, Acute myeloid dendritic cell leukemia, Acute myeloid leukemia, Acute promyelocytic leukemia, Adamantinoma, Adenocarcinoma, Adenoid cystic carcinoma, Adenoma, Adenomatoid odontogenic tumor, Adrenocortical carcinoma, Adult T-cell leukemia, Aggressive NK-cell leukemia, AIDS-Related Cancers, AIDS-related lymphoma, Alveolar soft part sarcoma, Ameloblastic fibroma, Anal cancer
  • Lyn has its general meaning in the art and refers to the LYN proto-oncogene, Src family tyrosine kinase encoded by the LYN gene (Gene ID: 4067) and is also known as JTK8; p53Lyn; p56Lyn.
  • An exemplary human nucleic acid sequence for Lyn is accessibled in GenBank under the access number NM 001111097.2 (isoform B or NM 001111097.2 (isoform A).
  • An exemplary human amino acid sequence for Lyn is accessible in GenBank under the accessible number NP 001104567.1 (isoform B), or NP 002341.1 (isoform A).
  • Lyn inhibitor refers to any compound that is capable of inhibiting the activity or expression of Lyn.
  • the term “Lyn activity” includes any biological activity mediated by Lyn such as described in the EXAMPLE.
  • the Lyn inhibitor of the present invention is particular suitable for abrogating kinase activity.
  • Lyn inhibitors include but are not limited to polypeptides such as dominant- negative protein mutants, peptidomimetics, antibodies, ribozymes, antisense oligonucleotides, or other small molecules which specifically inhibit the activity or expression ofLyn.
  • Lck has its general meaning in the art and refers to the LCK proto-oncogene, Src family tyrosine kinase encoded by the LCK gene (Gene ID: 3932) and I s also known as LSK; YT16; IMD22; p561ck; pp581ck.
  • An exemplary nucleic acid sequence for Lck is accessible in GenBank under the access number NM 001042771.2 or NM 005356.4.
  • An exemplary amino acid sequence for Lck is accessible in GenBank under the access number NP 001036236.1 or NP 005347.3.
  • a "Lck inhibitor” refers to any compound that is capable of inhibiting the activity or expression of Lck.
  • Lck activity includes any biological activity mediated by Lck such as described in the EXAMPLE.
  • the Lck inhibitor of the present invention is particular suitable for abrogating kinase activity.
  • Lck inhibitors include but are not limited to polypeptides such as dominant- negative protein mutants, peptidomimetics, antibodies, ribozymes, antisense oligonucleotides, or other small molecules which specifically inhibit the activity or expression of Lck.
  • the Lyn/Lck inhibitor is particularly suitable for promoting activation downstream of Fc, B-cell and T-cell antigen receptors and thus enhancing the immune system.
  • the Lyn/Lck inhibitor is a small organic molecule.
  • the inhibitor is a short hairpin RNA (shRNA), a small interfering
  • RNA or an antisense oligonucleotide which inhibits the expression of Lyn/Lck.
  • 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.
  • 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 R A virus such as a retrovirus.
  • retrovirus such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rous sarcoma virus
  • retrovirus such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rous sarcoma virus
  • adenovirus adeno-associated virus
  • the inhibitor is an intrabody having specificity for Lyn/Lck.
  • the term "intrabody” generally refer to an intracellular antibody or antibody fragment.
  • Antibodies in particular single chain variable antibody fragments (scFv), can be modified for intracellular localization. Such modification may entail for example, the fusion to a stable intracellular protein, such as, e.g., maltose binding protein, or the addition of intracellular trafficking/localization peptide sequences, such as, e.g., the endoplasmic reticulum retention.
  • the intrabody is a single domain antibody.
  • the antibody according to the invention is a single domain antibody.
  • sdAb single domain antibody
  • VHH single domain antibody
  • sdAb single domain antibody
  • VHH single domain antibody
  • sdAb single domain antibody
  • sdAb single domain antibody
  • VHH single domain 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®”.
  • sdAb can particularly be llama sdAb.
  • a “therapeutically effective amount” of the inhibitor as above described is meant a sufficient amount to provide a therapeutic effect. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed, the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific polypeptide employed; and like factors well known in the medical arts.
  • the daily dosage of the products may be varied over a wide range from 0.01 to 1,000 mg per adult per day.
  • the compositions contain 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 250 and 500 mg of the active ingredient for the symptomatic adjustment of the dosage to the subject to be treated.
  • a medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, preferably from 1 mg to about 100 mg of the active ingredient.
  • An effective amount of the drug is ordinarily supplied at a dosage level from 0.0002 mg/kg to about 20 mg/kg of body weight per day, especially from about 0.001 mg/kg to 7 mg/kg of body weight per day.
  • the inhibitor is administered to the subject in the form of a pharmaceutical composition.
  • the inhibitor may be combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form therapeutic compositions.
  • pharmaceutically acceptable excipients such as a pharmaceutically acceptable graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft copolymer, graft
  • the active principle in the pharmaceutical compositions of the present invention for oral, sublingual, subcutaneous, intramuscular, intravenous, transdermal, local or rectal administration, can be administered in a unit administration form, as a mixture with conventional pharmaceutical supports, to animals and human beings.
  • 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.
  • vehicles which are pharmaceutically acceptable for a formulation capable of being injected.
  • These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts), or 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.
  • the form In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • Solutions comprising compounds of the 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 inhibitor 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 typical 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.
  • 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.
  • the Lyn/Lck inhibitor is administered in combination with an immune checkpoint inhibitor.
  • an immune checkpoint inhibitor has its general meaning in the art and refers to any compound inhibiting the function of an immune inhibitory checkpoint protein. Inhibition includes reduction of function and full blockade.
  • 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.
  • the immune checkpoint inhibitor of the present invention is administered for enhancing the proliferation, migration, persistence and/or cytoxic activity of CD8+ T cells in the subject and in particular the tumor- infiltrating of CD8+ T cells of the subject.
  • the immune checkpoint inhibitor is a PD-1 inhibitor.
  • PD-1 inhibitor refers to a compound, substance or composition that can inhibit the function of PD-1.
  • the inhibitor can inhibit the expression or activity of PD-1, modulate or block the PD-1 signaling pathway and/or block the binding of PD-1 to PD-L1 or PD-L2.
  • the immune checkpoint inhibitor is an antibody selected from the group consisting of anti-CTLA4 antibodies, anti-PDl antibodies, anti-PDLl antibodies, anti-PDL2 antibodies anti-TIM-3 antibodies, anti-LAG3 antibodies, anti-B7H3 antibodies, anti-B7H4 antibodies, anti-BTLA antibodies, and anti-B7H6 antibodies.
  • a further object of the present invention relates to a method of screening a drug suitable for the treatment of cancer comprising i) providing a test compound and ii) determining the ability of said test compound to inhibit the expression or activity of Lyn/Lck.
  • Any biological assay well known in the art could be suitable for determining the ability of the test compound to inhibit the activity or expression of Lyn/Lck.
  • the assay fist comprises determining the ability of the test compound to bind to Lyn/Lck.
  • a population of immune cells (mastocytes, monocytes, B cells, T cells...) is then contacted and activated so as to determine the ability of the test compound to inhibit the activation downstream of Fc, B-cell and T-cell antigen receptors.
  • the SHP-1 activation is determined and in particular the identification of serine into tyrosine phosphorylation.
  • the effect triggered by the test compound is determined relative to that of a population of immune cells incubated in parallel in the absence of the test compound or in the presence of a control agent either of which is analogous to a negative control condition.
  • control substance refers a molecule that is inert or has no activity relating to an ability to modulate a biological activity or expression.
  • Assays for determining the test compound to dampen the immune signal response are well known in the art and are typically described in the EXAMPLE.
  • test compounds capable of inhibiting immune response are likely to exhibit similar modulatory capacity in applications in vivo.
  • the test compound is selected from the group consisting of peptides, petptidomimetics, small organic molecules, antibodies (e.g. intraantibodies), aptamers or nucleic acids.
  • the test compound according to the invention may be selected from a library of compounds previously synthesised, or a library of compounds for which the structure is determined in a database, or from a library of compounds that have been synthesised de novo.
  • the test compound may be selected form small organic molecules.
  • small organic molecule refers to a molecule of size comparable to those organic molecules generally sued in pharmaceuticals.
  • LN Long HI and six LN patients were studied.
  • the LN group was composed of 6 patients attending or referred to the Bichat's Hospital specialist nephrology unit between July 2014 and January 2016 meeting at least four ACR systemic lupus erythematosus criteria (32) presenting with active disease with nephritis proven by kidney biopsy (2 at class IV and 4 at class V) and in whom peripheral blood by venepuncture was obtained immediately prior to immunosuppressive therapy administration. All patients were female with age varying between 25 and 42. Ethical approval for this study was obtained from the Bichat Hospital Local Research. Ethics Committee and informed consent was obtained from all subjects enrolled.
  • mice expressing the WT human FcyRIIA on CD 1 lb-positive cells were from Jackson Laboratory (JAX, Bar Harbor, ME, USA).
  • Fyn ' hFcyRIIA 18 , Lyn _/" hFcyRIIA Tg were obtained by the intercross of hFcyRIIA Tg mice with mice knockout for Fyn (JAX) or for Lyn (previously described in (15)). All mice carrying the hFcyRIIA transgene were used as heterozygous animals. Mice were bred and maintained at the mouse facilities of the Bichat Medical School campus. All experiments were performed in accordance with the French Council of Animal Care guidelines and national ethical guidelines of INSERM Animal Care Committee (Animal Use Protocol number 75-1596).
  • NTN Nephrotoxic nephritis
  • NTN was induced by i.p. injection (200 ⁇ 1/20 g body weight) of rabbit anti-mouse glomerular basement membrane (GBM) in C57BL/6 hFcyRIIA Tg , Lyn _/" , Fyn _/" , Fyn _/" hFcyRIIA Tg , Lyn ' hFcyRIIA ⁇ mice (7 to 9 wk old). Briefly, mice were preimmunized i.p. with normal rabbit IgG (0.5 mg/20 g body weight) in CFA 5 days prior to i.p. administration of NTN serum. Blood samples were collected and animals were sacrificed at day 7 following NTN injection. Renal function parameters (urinary proteins and BUN), histological and immunohistological parameters were studied.
  • GBM rabbit anti-mouse glomerular basement membrane
  • CAIA Collagen Antibody-Induced Arthritis
  • Arthritis was induced as described (9, 30) using the Arthrogen-CIA® Arthritogenic Monoclonal Antibody kit (Chondrex, Inc.). Mice were injected i.v. with anti-CII Ab cocktail (Day 0) followed by LPS (i.p.) 3 days later. Animals were injected i.p. with 10 mg/20 g body weight of 500 ⁇ g serum human IgA (purchased from Biomedicals)/20 g body weight or 100 ⁇ g AT-10 F(ab)' 2 or irrelevant mAb F(ab') 2 (clone 320) for 10 days at 2-day intervals. The first dose was administered 2 days prior to anti-CII Ab cocktail injection. Paw thickness was measured with a pocket thickness gauge. On day 10, animals were sacrificed and hind paws and knees were fixed in formalin or snap-frozen.
  • Jurkat cells were incubated with or without anti-CD3 F(ab') 2 fragment or with preformed complexes of anti-CD3 F(ab') 2 plus anti-kappa F(ab') 2 fragments. Cells were then stimulated or not with flagellin (1 ⁇ g) for 6 hours. PMA (40 nM) and ionomycin (InM) were used as positive stimuli for 6 hours. Brefeldin A was added after 2 hours stimulation and maintained for 4 hours. The stimulation was stopped by adding 1 ml cold PBS.
  • Intracellular cytokine staining was performed on fixed/permeabilized cells in residual permeabilization wash buffer (Biolegend, USA) using a conjugated antibody (anti-IL-2 PE or appropriate isotype control) for 20 min in the dark at room temperature as described (36). Data acquisition was performed using a BD Biosciences LSR Fortessa cytometer, and results were analyzed using Flow Jo analysis software (Tree Star).
  • Cells (5 x 10 6 to 10 7 ) were solubilized in RIPA lysis buffer containing 1% Nonidet P- 40/0.1% sodium dodecyl sulfate (SDS) as described 8 .
  • SDS sodium dodecyl sulfate
  • cell lysates were incubated with 2 ⁇ g/ml of AT- 10 anti-FcyRIIA, A77 anti-FcaRI, HIT-3a anti-CD3 or ZL7-4 anti-CD79a mAbs and immunoprecipitated overnight at 4°C with Protein G-Sepharose (GE Healthcare).
  • Enzyme-linked immunosorbent assay ELISA
  • IL-8 and IL-2 were measured in the supernatants of stimulated cells using ELISA kits (R&D Systems) according to the manufacturer's instructions.
  • kidney sections 4 ⁇ in thickness were stained with PAS for morphological analysis.
  • frozen kidney sections were incubated with biotinylated antibodies against rabbit IgG or mAb anti-mouse CD l ib, -mouse F4/80, - mouse CD3, and -mouse Ly6G (Becton Dickinson) for 1 hour at room temperature.
  • the primary antibody incubation was followed by incubation with anti-rabbit IgG or anti-goat IgG (Southern Biotech Associates).
  • Slides were mounted with the Eukitt mounting medium (Electron Microscopy Sciences) and read with an upright microscope (DM2000; Leica) using the IM50 software (Leica).
  • RNA purification from ho mogen i zed_k i d ney s was performed by using RNAble (Eurobio).
  • cDNA was obtained by reverse transcription using using Moloney murine leukaemia virus (Invitrogen). Samples were analyzed by real-time PCR with Taq Man® Gene Expression Master Mix (Applied Biosystem). Primers were purchased from Euro fins (Supplementary Table 1). Gene quantification was performed using a Chrom o4 Real-Time PCR Detection System (Bio-Rad Laboratories). Data were normalized to ⁇ -actin values.
  • F indicates a forward primer
  • R indicates a reverse primer
  • P indicates a FAM-TAMRA probe.
  • the immune system is controlled by a finely tuned network of regulatory mechanism (1).
  • certains immunoreceptors have been shown to exert inhibitory and activating signal through ITAM motif (Yxx[L/I]x6-sYxx[L/I]) (2) depending on the valency of their respective ligand.
  • Low valency interactions were shown to induce anergy and an inhibitory crosstalk with heterologous receptors thereby reducing the susceptibility to autoimmune and inflammatory diseases (3-10).
  • high valency ligand interactions promote an activating signal launching inflammatory and immune cascades to fight the inflammatory insult and restore homeostasis, but in case of ill-regulation or chronic stimulation can also result in autoimmune and inflammatory diseases (1, 10).
  • the ITAM motif is found in the cytoplasmic domain of several transmembrane adapter molecules, such as the common ⁇ subunit of FcR (FcRy), the Iga and Ig subunits of the BCR, and the ⁇ , ⁇ , ⁇ and ⁇ subunits of the TCR-associated CD3 complex (1, 2), and in the cytoplasmic tail of other receptors, such as the FcyRIIA (11).
  • FcRy FcR
  • FcRy FcR
  • Iga and Ig subunits of the BCR and the ⁇ , ⁇ , ⁇ and ⁇ subunits of the TCR-associated CD3 complex (1, 2)
  • FcyRIIA FcyRIIA
  • Src-family kinases phosphorylate the ITAM motifs upon stimulus- induced receptor clustering leading to downstream effector recruitments and cell activation.
  • ITAM-bearing immunoreceptors translate ligand valency into opposite signals remains elusive.
  • TCR and BCR were performed using anti-CD3 or anti-CD79a F(ab') 2 fragments or complexed with anti- ⁇ light chains in representative lymphocytic cell lines.
  • FcRs while divalent targeting of TCR or BCR resulted in ITAMi signatures, multivalent crosslinking led to expected ITAM signature.
  • Lck or Lyn was required for TCR- or BCR-mediated ITAMi signals, whereas Fyn was essential for ITAM configuration.
  • TCR-Fyn dissociation led to SHP-1 recruitment suggesting that Fyn could inhibit SHP-1 recruitment.
  • TCR- and BCR-divalent targeting resulted in inhibition of IL-2 and IL-8 secretion induced by flagellin or Pam3csk4 which were dependent on Lck or Lyn, respectively
  • Fyn was required for these cytokine production induced upon TCR and BCR multivalent engagement.
  • NTN nephrotoxic nephritis
  • Lyn _/ ⁇ FcyRIIA Tg mice did not develop significant disease despite glomerular Ab deposits. Renal disease development in Lyn _/" FcyRIIA Tg mice was characterized by a marked increase in urinary protein and blood urea nitrogen concentration (BUN). Lyn "/- FcyRIIA Tg mice also exhibited severe renal injury involving extensive mesangial and capillary (subendothelial or even intracapillary) deposits associated with mild mesangial and endocapillary plus extracapillary proliferation.
  • Glomerular lesions were characterized by an intense macrophage infiltrate and cytokine production. These effects involved activation of ITAM signaling, as demonstrated by in situ phosphorylation of the Y 525 residue in Syk.
  • the protective role of Lyn was confirmed in another autoimmune model, the collagen antibody-induced arthritis (CAIA) model.
  • CAIA collagen antibody-induced arthritis
  • targeting human FcyRIIA or FcaRI in Tg animals for ITAMi signaling prevented disease development, and this protection required the presence of Lyn.
  • Lyn was strongly associated with FcyRIIA in healthy individuals but weakly associated in patients, whereas Fyn and Syk were exclusively associated with FcyRIIA in LN patients.
  • SHP-1 and pSHP-l Y536 were associated with FcyRIIA exclusively in cells from healthy individuals.
  • pSHP- 1 S591 and pPKCa were exclusively observed in LN patient cell lysate samples, thus emphasizing an IT AM configuration.
  • this inactive pSHP-l S591 and pPKCa were not associated with FcyRIIA.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des méthodes et des compositions pharmaceutiques pour le traitement du cancer. Les immunorécepteurs jouent un rôle essentiel dans la régulation de l'homéostasie immunitaire et de l'inflammation. Les auteurs de l'invention ont observé comment ces récepteurs traduisent les interactions de ligands extérieurs en signaux opposés. La kinase Fyn de la famille Src est l'effecteur essentiel pour l'inhibition de SHP-1, tandis que Lyn/Lck est nécessaire pour son activation en aval de Fc, des récepteurs d'antigènes des lymphocytes T et des lymphocytes B. Les auteurs de l'invention ont démontré les rôles distincts des kinases de la famille Src associées au récepteur des antigènes dans la régulation des états homéostatiques et inflammatoires, et proposent ainsi un moyen permettant de moduler le système immunitaire et de favoriser la lutte contre le cancer. La présente invention concerne en particulier une méthode de traitement du cancer chez un sujet en ayant besoin, consistant à administrer audit sujet une quantité thérapeutiquement efficace d'un inhibiteur de Lyn/Lck.
PCT/EP2017/064093 2016-06-10 2017-06-09 Méthodes et compositions pharmaceutiques pour le traitement du cancer WO2017212021A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP16305685 2016-06-10
EP16305685.6 2016-06-10

Publications (1)

Publication Number Publication Date
WO2017212021A1 true WO2017212021A1 (fr) 2017-12-14

Family

ID=56289440

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2017/064093 WO2017212021A1 (fr) 2016-06-10 2017-06-09 Méthodes et compositions pharmaceutiques pour le traitement du cancer

Country Status (1)

Country Link
WO (1) WO2017212021A1 (fr)

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US6107091A (en) 1998-12-03 2000-08-22 Isis Pharmaceuticals Inc. Antisense inhibition of G-alpha-16 expression
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
WO2015069770A1 (fr) * 2013-11-05 2015-05-14 Cognate Bioservices, Inc. Combinaisons d'inhibiteurs de point de contrôle et d'agents thérapeutiques pour traiter un cancer
WO2016036886A1 (fr) * 2014-09-02 2016-03-10 Cedars-Sinai Medical Center Compositions et méthodes de traitement de troubles fibrosants et du cancer

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US6365354B1 (en) 2000-07-31 2002-04-02 Isis Pharmaceuticals, Inc. Antisense modulation of lysophospholipase I 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
WO2015069770A1 (fr) * 2013-11-05 2015-05-14 Cognate Bioservices, Inc. Combinaisons d'inhibiteurs de point de contrôle et d'agents thérapeutiques pour traiter un cancer
WO2016036886A1 (fr) * 2014-09-02 2016-03-10 Cedars-Sinai Medical Center Compositions et méthodes de traitement de troubles fibrosants et du cancer

Non-Patent Citations (41)

* Cited by examiner, † Cited by third party
Title
A. B. DANIELS ET AL., SCAND J IMMUNOL, vol. 71, 2010, pages 232
A. GETAHUN; J. C. CAMBIER., IMMUNOL REV, vol. 268, 2015, pages 66
A. S. AKHADE; A. QADRI, J IMMUNOL, vol. 45, 2015, pages 2628
B. PASQUIER ET AL., IMMUNITY, vol. 22, 2005, pages 31
C. TAN SARDJONO ET AL., ARTHRITIS RHEUM, vol. 52, 2005, pages 3220
E. H. PALACIOS; A. WEISS, ONCOGENE, vol. 23, 2004, pages 7990
E. M. TAN ET AL., ARTHRITIS RHEUM, vol. 25, 1982, pages 1271
E. ROSSATO ET AL., ARTHRITIS RHEUMATOL, vol. 67, 2015, pages 1766
EVAN INGLEY: "Functions of the Lyn tyrosine kinase in health and disease", CELL COMMUNICATION AND SIGNALING, BIOMED CENTRAL, LONDON, GB, vol. 10, no. 1, 17 July 2012 (2012-07-17), pages 21, XP021133103, ISSN: 1478-811X, DOI: 10.1186/1478-811X-10-21 *
F. PINHEIRO DA SILVA ET AL., NAT MED, vol. 13, 2007, pages 1368
G. A. KORETZKY; P. S. MYUNG, NAT REV IMMUNOL, vol. 1, 2001, pages 95
H. NISHIZUMI ET AL., IMMUNITY, vol. 3, 1995, pages 549
I. MAGALHAES ET AL., J CLIN INVEST, vol. 125, 2015, pages 1752
I. R. HARDY ET AL., J IMMUNOL, vol. 192, 2014, pages 1641
I. STEFANOVA ET AL., NAT IMMUNOL, vol. 4, 2003, pages 248
J. A. HAMERMAN ET AL, IMMUNOL REV, vol. 232, 2009, pages 42
J. A. HAMERMAN; L. L. LANIER., SCI STKE, 2006
J. S. BEZBRADICA; R. MEDZHITOV, CURR OPIN IMMUNOL, vol. 24, 2012, pages 58
JASON JOHN LUKE ET AL: "Kinase inhibitors and immune check-point blockade for the treatment of metastatic melanoma and advanced cancer: synergistic or antagonistic?", EXPERT OPINION ON PHARMACOTHERAPY, vol. 14, no. 18, 21 October 2013 (2013-10-21), LONDON, UK, pages 2457 - 2462, XP055257629, ISSN: 1465-6566, DOI: 10.1517/14656566.2013.849244 *
KIMMO PORKKA ET AL: "An open-label, phase 1b, dose-escalation study (CA180-373) of dasatinib plus nivolumab, an investigational anti-programmed cell death 1 (PD-1) antibody, in patients (pts) with previously treated chronic myeloid leukemia (CM L)", JOURNAL OF CLINICAL ONCOLOGY, vol. 32, 1 January 2014 (2014-01-01), pages TPS7119, XP055318274 *
L. SHANG ET AL., J BIOL CHEM, vol. 289, 2014, pages 15309
M. ALOULOU ET AL., BLOOD, vol. 119, 2012, pages 3084
M. KRAUS ET AL., J EXP MED, vol. 194, 2001, pages 455
M. L. HIBBS ET AL., CELL, vol. 83, 1995, pages 301
M. L. JONES; J. D. CRAIK; J. M. GIBBINS; A. W. POOLE, J BIOL CHEM, vol. 279, pages 40475
M. P. COOKE ET AL, CELL, vol. 65, pages 281
M. RETH., NATURE, vol. 338, 1989, pages 383
O. FEINERMAN ET AL, SCIENCE, vol. 321, 2008, pages 1081
P. M. HOGARTH, CURR OPIN IMMUNOL, vol. 14, 2002, pages 798
P. SCAPINI ET AL, IMMUNOL REV, vol. 228, 2009, pages 23
S. BEN MKADDEM ET AL., J CLIN INVEST, vol. 124, 2014, pages 3945
S. BOLT ET AL., J IMMUNOL, vol. 23, 1993, pages 403
S. ODOM ET AL., J EXP MED, vol. 199, 2004, pages 1491
S. P. MCADOO ET AL., KIDNEY INT, vol. 88, 2015, pages 52
TOSHIKAGE NAGAO ET AL: "Proliferation and Survival Signaling from Both Jak2-V617F and Lyn Involving GSK3 and mTOR/p70S6K/4EBP1 in PVTL-1 Cell Line Newly Established from Acute Myeloid Leukemia Transformed from Polycythemia Vera", PLOS ONE, vol. 9, no. 1, 3 January 2014 (2014-01-03), pages e84746, XP055318603, DOI: 10.1371/journal.pone.0084746 *
U. BLANK ET AL, IMMUNOL REV, vol. 232, pages 59
V. PARRAVICINI ET AL., NAT IMMUNOL, vol. 3, 2002, pages 741
XIAO W ET AL: "Lyn- and PLC-beta3-dependent regulation of SHP-1 phosphorylation controls Stat5 activity and myelomonocytic leukemia-like disease", BLOOD, AMERICAN SOCIETY OF HEMATOLOGY, US, vol. 116, no. 26, 23 December 2010 (2010-12-23), pages 6003 - 6013, XP002726002, ISSN: 0006-4971, [retrieved on 20100921], DOI: 10.1182/BLOOD-2010-05-283937 *
Y. KANAMARU ET AL., J IMMUNOL, vol. 180, 2008, pages 2669
Y. LIU; M. J. KRUHLAK; J. J. HAO; S. SHAW., JLEUKOC BIOL, vol. 82, 2007, pages 742
Z. YE; Y. H. GAN, J BIOL CHEM, vol. 282, 2007, pages 4479

Similar Documents

Publication Publication Date Title
Eisele et al. Targeting apoptosis pathways in glioblastoma
KR20140019303A (ko) 루푸스의 치료 또는 예방을 위한 조성물 및 방법
US10105384B2 (en) Nucleic acids targeting TCTP for use in the treatment of chemo- or hormone-resistant cancers
JP2020536902A (ja) 炎症性疾患を処置するための方法および組成物
WO2007143629A1 (fr) Traitement de la neurofibromatose avec des inhibiteurs d'une voie de transduction du signal
JP2024028924A (ja) Cbmシグナロソーム複合体を標的にすることにより、制御性t細胞に腫瘍微小環境の炎症を引き起こさせる方法
EP2335717A1 (fr) Antagonistes PAR-1 pour une utilisation dans le traitement ou la prévention d'infections par le virus de la grippe de type A
EP3289104B1 (fr) Procédé pour le traitement de gliomes de haut grade
WO2017212021A1 (fr) Méthodes et compositions pharmaceutiques pour le traitement du cancer
US20220220480A1 (en) Methods and compositions for treatment of nlrp3 inflammasome mediated il-1beta dependent disorders
WO2021035048A1 (fr) Utilisation d'inhibiteurs de yap et de sox2 pour le traitement du cancer
WO2020142646A1 (fr) Co-administration d'inhibiteurs pour produire des cellules intestinales produisant de l'insuline
US9970012B2 (en) Replication factor C-40 (RFC40/RFC2) as a prognostic marker and target in estrogen positive and negative and triple negative breast cancer
KR20200028982A (ko) 시냅스 기능을 증진시키기 위한 hdac2-sp3 복합체 표적화
WO2017174626A1 (fr) Procédés et compositions pharmaceutiques pour inhiber la dégranulation des mastocytes
CA3043030A1 (fr) Inhibition de kmt2d pour le traitement du cancer
Wang et al. ADP‐ribose transferase PARP16 mediated‐unfolded protein response contributes to neuronal cell damage in cerebral ischemia/reperfusion
WO2024037910A1 (fr) Inhibiteurs de syk destinés à être utilisés dans le traitement du cancer
WO2024028476A1 (fr) Méthodes de traitement de maladies médiées par th2
US20130236480A1 (en) Transglutaminase 2 inhibitors for use in the prevention or treatment of rapidly progressive glomerulonephritis
JP6846808B2 (ja) Card14を用いた治療、診断およびスクリーニング
Saisorn Neutrophil Extracellular Traps (NETs) formation of Fcgr2b deficient mice in lupus mouse model with ischemic reperfusion injury
WO2014095916A1 (fr) Ninjurine-1 comme cible thérapeutique pour une tumeur du cerveau
WO2022271955A1 (fr) Nouvelles nanoparticules de sharn ciblées pour la thérapie du cancer
WO2023012343A1 (fr) Procédés de traitement du 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: 17732803

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: 17732803

Country of ref document: EP

Kind code of ref document: A1