WO2022171121A1 - 治疗肿瘤的方法和组合 - Google Patents

治疗肿瘤的方法和组合 Download PDF

Info

Publication number
WO2022171121A1
WO2022171121A1 PCT/CN2022/075642 CN2022075642W WO2022171121A1 WO 2022171121 A1 WO2022171121 A1 WO 2022171121A1 CN 2022075642 W CN2022075642 W CN 2022075642W WO 2022171121 A1 WO2022171121 A1 WO 2022171121A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino
chromen
pyrimidin
pyrazolo
substituted
Prior art date
Application number
PCT/CN2022/075642
Other languages
English (en)
French (fr)
Inventor
吕容真
陈志宏
Original Assignee
同润生物医药(上海)有限公司
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 同润生物医药(上海)有限公司 filed Critical 同润生物医药(上海)有限公司
Priority to EP22752281.0A priority Critical patent/EP4292596A1/en
Priority to US18/264,394 priority patent/US20240102634A1/en
Priority to CN202280012531.5A priority patent/CN117222413A/zh
Publication of WO2022171121A1 publication Critical patent/WO2022171121A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • A61K31/522Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/179Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1841Transforming growth factor [TGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/643Albumins, e.g. HSA, BSA, ovalbumin or a Keyhole Limpet Hemocyanin [KHL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • 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
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/32Fusion polypeptide fusions with soluble part of a cell surface receptor, "decoy receptors"

Definitions

  • the present application relates to the field of biomedicine, in particular to a method and combination for treating tumors.
  • PI3K phosphoinositide 3-kinase signaling pathway
  • PI3Ks are members of a unique and conserved family of intracellular lipid kinases that phosphorylate phosphatidylinositol or the 3'-OH group on phosphoinositides.
  • the PI3K family includes 15 kinases with distinct substrate specificities, expression patterns, and regulatory patterns.
  • Class I PI3Ks (p110 ⁇ , p110 ⁇ , p110 ⁇ , and p110 ⁇ ) are typically activated by tyrosine kinases or G-protein coupled receptors to generate (3,4,5)-phosphatidylinositol triphosphate (PIP3), which engages downstream Effectors, such as those in the AKT/PDK1 pathway, mTOR, Tec family kinases and Rho family GTPases.
  • Class II and class III PI3Ks are transported intracellularly by the synthesis of phosphatidylinositol 3-bisphosphate (PI(3)P) and (3,4)-bisphosphate phosphatidylinositol (PI(3,4)P2) play a key role.
  • PI3K is a protein kinase that controls cell growth (mTORC1) or monitors genome integrity (ATM, ATR, DNA-PK and hSmg-1).
  • PI3K-alpha is the most commonly mutated isoform in cancer and plays a role in insulin signaling and glucose homeostasis (Knight et al Cell (2006) 125(4):733–47; Vanhaesebroeck et al Current Topic Microbiol. Immunol. (2010) 347:1–19).
  • PTEN tensin homolog
  • PI3K-delta and -gamma are preferentially expressed in leukocytes and are important in leukocyte function. These subtypes also contribute to the development and maintenance of hematological malignancies (Vanhaesebroeck et al Current Topic Microbiol. Immunol. (2010) 347:1–19; Clayton et al J Exp Med. (2002) 196(6):753– 63; Fung-Leung Cell Signal. (2011) 23(4):603-8; Okkenhaug et al. Science (2002) 297(5583):1031-34).
  • PI3K-delta is activated by cellular receptors (eg, receptor tyrosine kinases) through interaction with the Sarc homology 2 (SH2) domain of the PI3K regulatory subunit (p85) or through direct interaction with RAS.
  • cellular receptors eg, receptor tyrosine kinases
  • SH2 Sarc homology 2 domain of the PI3K regulatory subunit
  • TGF ⁇ Transforming growth factor beta
  • TGF ⁇ is a potent cytokine with significant effects on the immune system.
  • the main function of TGF ⁇ in the immune system is to maintain tolerance and initial immune response to foreign pathogens.
  • Three isoforms of TGF[beta] have been identified in mammals, namely TGF[beta]1, TGF[beta]2 and TGF[beta]3, of which TGF[beta]1 is the predominant isoform.
  • TGF ⁇ is secreted in a latent form, and only a small fraction of the total TGF ⁇ secreted is activated under physiological conditions.
  • Most of the biological effects of TGF ⁇ are achieved through the binding of TGF ⁇ to the receptors ALK5 and TGF ⁇ receptor II (TGF ⁇ R2).
  • active TGF ⁇ dimers can bind tetrameric ALK5 and TGF ⁇ R2 complexes to initiate signal transduction.
  • ALK5 is not required for initial binding of TGF ⁇ , but is required for signal
  • PD-1 Programmed death 1
  • PD-L1 is a member of the CD28 superfamily. PD-1 is expressed in activated T cells, B cells and myeloid cells, and it has two ligands, programmed death ligand 1 (PD-L1) and PD-L2. PD-L1 interacts with the receptor PD-1 on T cells and plays an important role in the negative regulation of immune responses.
  • the expression of PD-L1 protein can be detected in many human tumor tissues.
  • the microenvironment of the tumor site can induce the expression of PD-L1 on tumor cells.
  • the expressed PD-L1 is beneficial to the occurrence and growth of tumors and induces anti-tumor effects.
  • PD-1/PD-L1 pathway inhibitors can block the binding of PD-1 to PD-L1, block negative regulatory signals, and restore the activity of T cells, thereby enhancing the immune response
  • TGF- ⁇ Transforming growth factor- ⁇ belongs to the TGF- ⁇ superfamily that regulates cell growth and differentiation.
  • TGF-beta signals through a heterotetrameric receptor complex consisting of two type I and two type II transmembrane serine/threonine kinase receptors.
  • TGF[beta] can affect many cellular functions, such as cell proliferation, differentiation, cell-cell and cell-matrix adhesion, cell motility, and activation of lymphocytes. (For a review of the role of TGF[beta] in regulating immune responses, see Li et al. (2006) Annu. Rev. Immunol. 24:99-146.) Furthermore, TGF[beta] is believed to induce or mediate the progression of many diseases, such as osteoporosis disease, hypertension, atherosclerosis, cirrhosis, fibrotic diseases of the kidneys, liver, and lungs, and tumor progression.
  • diseases such as osteoporosis disease, hypertension, atherosclerosis, cirrhosis, fibrotic diseases of the kidneys, liver, and lungs, and tumor progression.
  • TGF ⁇ can enhance chronic inflammation-induced end-organ damage, while TGF ⁇ Antagonists are effective in reducing this damage (Border et al. (1990) Nature 346:371-374; Border et al. (1992) Nature 360:361-364; Isaka et al. (1999) Kidney Int. 55:465-475; Sharma et al. (1996) Diabetes 45: 522; Xin et al (2004) Transplantation 15: 1433; Benigni et al (2003) J. Am. Soc. Nephrol. 14: 1816).
  • TGF ⁇ may have a direct inhibitory effect on malignant cells and may increase the production or activity of a range of tumor growth factors and angiogenic factors.
  • Tumor immunotherapy mainly uses immunological principles and methods to improve the immunogenicity of tumor cells and the sensitivity to effector cell killing, stimulate and enhance the body's anti-tumor immune response, and use immune cells and effector molecules to infuse the host into the body.
  • the immune system kills tumors and inhibits tumor growth. Reducing the toxicity and resistance of tumor drugs and reducing the toxic and side effects are still urgent problems to be solved in tumor therapy.
  • a PI3K inhibitor and a second therapeutic agent of choice comprising a PI3K inhibitor and a second therapeutic agent of choice.
  • the combination of a PI3K inhibitor and a second therapeutic agent selected from one or more of the following has been found to be synergistic in treating tumors (eg, in reducing cancer cell growth or viability, or both) : Immune checkpoint inhibitor, TGF ⁇ inhibitor, or a combination thereof.
  • the combination of a PI3K inhibitor and a second therapeutic agent of choice may allow for lower doses of the PI3K inhibitor, the second therapeutic agent, or both, than the monotherapy doses required to achieve the same therapeutic effect apply.
  • the combination may allow the PI3K inhibitor, the second therapeutic agent, or both, to be administered less frequently than when the PI3K inhibitor or the second therapeutic agent is administered as a monotherapy.
  • Such combinations may provide beneficial effects, eg, in reducing, preventing, delaying and/or reducing the occurrence of one or more of the following consequences: side effects, toxicity or resistance otherwise associated with administration of higher doses of the agent.
  • the application provides a pharmaceutical combination comprising a phosphoinositide 3-kinase (PI3K) inhibitor and a second therapeutic agent, wherein the second therapeutic agent is an immune checkpoint inhibitor, a TGF ⁇ inhibitor, a dual function Immune checkpoint/TGF ⁇ inhibitors or combinations thereof.
  • PI3K phosphoinositide 3-kinase
  • the inhibitor of the immune checkpoint comprises an agent capable of blocking the interaction of programmed death 1 (PD-1) with programmed death ligand 1 (PD-L1).
  • the inhibitor of the immune checkpoint comprises an inhibitor of programmed death ligand 1 (PD-L1) and/or programmed death 1 (PD-1).
  • the dual function immune checkpoint/TGF ⁇ inhibitor comprises a dual PD-L1/TGF ⁇ inhibitor, or a dual PD-1/TGF ⁇ inhibitor.
  • the pharmaceutical combination comprises:
  • the inhibitor of the immune checkpoint comprises nucleic acid (eg, dsRNA, siRNA or shRNA), polypeptide (eg, soluble ligand, antibody or antigen-binding fragment thereof, immunoadhesin, fusion protein) , oligopeptides and other molecules), or compounds.
  • nucleic acid eg, dsRNA, siRNA or shRNA
  • polypeptide eg, soluble ligand, antibody or antigen-binding fragment thereof, immunoadhesin, fusion protein
  • oligopeptides and other molecules oligopeptides and other molecules
  • the antibody is selected from the group consisting of: human antibodies, humanized antibodies, chimeric antibodies, multispecific antibodies, monoclonal antibodies, and polyclonal antibodies.
  • the antigen-binding fragment is selected from the group consisting of: Fab, Fab', F(ab') 2 , Fv, scFv, diabodies, Fd, dAbs, VHHs, large antibodies and complementarity determining regions (CDRs) Fragment.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain variable region (VH) comprising HCDR1, HCDR2 and HCDR3.
  • VH heavy chain variable region
  • the antibody or antigen-binding fragment thereof further comprises or does not comprise a CH1 domain.
  • the antibody or antigen-binding fragment thereof further comprises or does not comprise CH2 and CH3 domains.
  • the antibody or antigen-binding fragment thereof further comprises an antibody heavy chain constant region.
  • the antibody heavy chain constant region is a human antibody heavy chain constant region.
  • the human antibody heavy chain constant region is selected from constant regions derived from the group consisting of IgGl, IgG2, IgG3, IgG4, and variants thereof.
  • the antibody or antigen-binding fragment thereof further comprises a light chain variable region (VL) comprising LCDR1, LCDR2, LCDR3.
  • VL light chain variable region
  • the antibody or antigen-binding fragment thereof further comprises a light chain constant region (CL).
  • CL light chain constant region
  • the inhibitor of the immune checkpoint comprises an anti-PD-L1 antibody or antigen-binding fragment thereof, or an anti-PD-1 antibody or antigen-binding fragment thereof.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain variable region of an antibody
  • the heavy chain variable region comprises HCDR1, HCDR2, HCDR3, each of which is associated with the following molecules, respectively HCDR1, HCDR2, HCDR3 have at least about 80% (eg, about 85%, 90% or 95%) sequence identity to: Atezolizumab, Avelumab, BMS-936559, MPDL3280A (RG7446) or durvalumab (MEDI4736).
  • the heavy chain variable regions have at least about 80% (eg, about 85%, 90% or 95%) sequence identity to the heavy chain variable regions of the following molecules, respectively: Atezolizumab, Avelumab, BMS-936559, MPDL3280A, or durvalumab.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain of an antibody, wherein the heavy chain is at least about 80% (e.g., about 85%, e.g., about 85%, respectively,) 90% or 95%) sequence identity: Atezolizumab, Avelumab, BMS-936559, MPDL3280A, or durvalumab.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof further comprises a light chain variable region of an antibody, wherein the light chain variable region comprises LCDR1, LCDR2, LCDR3, each of which is associated with the following Molecules LCDR1, LCDR2, LCDR3 have at least about 80% (eg, about 85%, 90% or 95%) sequence identity to: Atezolizumab, Avelumab, BMS-936559, MPDL3280A or durvalumab.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain variable region and a light chain variable region of an antibody, wherein the heavy chain variable region comprises HCDR1, HCDR2, HCDR3, wherein the light chain can
  • the variable region comprises LCDR1, LCDR2, LCDR3, each of which has at least about 80% (eg, about 85%, 90% or 95%) sequence identity to HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, LCDR3, respectively, of the following molecules: Atezolizumab, Avelumab, BMS-936559, MPDL3280A (RG7446) or durvalumab (MEDI4736).
  • the light chain variable regions have at least about 80% (eg, about 85%, 90% or 95%) sequence identity to the light chain variable regions of atezolizumab, Avelumab, BMS-936559, MPDL3280A, or durvalumab.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain variable region and a light chain variable region of an antibody, each of which has at least about 80% (eg, about 85%, 90% or 95%) sequence identity: Atezolizumab, Avelumab, BMS-936559, MPDL3280A (RG7446) or durvalumab (MEDI4736).
  • the anti-PD-L1 antibody or antigen-binding fragment thereof further comprises a light chain of an antibody, wherein the light chain is at least about 80% (eg, about 85%, respectively) the light chain of the following molecule , 90% or 95%) sequence identity: Atezolizumab, Avelumab, BMS-936559, MPDL3280A, or durvalumab.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises heavy and light chains of an antibody that are each at least about 80% (eg, about 85%, 90% or 95%) sequence identity to: Atezolizumab, Avelumab, BMS-936559, MPDL3280A (RG7446) or durvalumab (MEDI4736).
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises: Atezolizumab, Avelumab, BMS-936559, MPDL3280A, or durvalumab, or an antigen-binding fragment thereof, or a variant or biosimilar thereof, or its combination.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain variable region of an antibody
  • the heavy chain variable region comprises one or more heavy chain selectable from the group consisting of Variable region CDRs (HCDRs): (a) HCDR1 having at least 70% sequence identity to the amino acid sequence set forth in SEQ ID NO: 1; (b) HCDR2 having at least 70% sequence identity to SEQ ID NO: 2; and (c) HCDR3 having at least 70% sequence identity to SEQ ID NO:3.
  • HCDRs Variable region CDRs
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain variable region of an antibody
  • the heavy chain variable region comprises one or more HCDRs selected from the group consisting of: ( a) HCDR1 with the amino acid sequence shown in SEQ ID NO: 1 or HCDR1 obtained by amino acid addition, elimination or substitution reaction with no more than 2 amino acid differences from the amino acid sequence shown in SEQ ID NO: 1; (b) HCDR2 having the amino acid sequence shown in SEQ ID NO: 2 or HCDR2 obtained by amino acid addition, elimination or substitution reaction and having no more than 2 amino acid differences with the amino acid sequence shown in SEQ ID NO: 2; and (c) having The HCDR3 of the amino acid sequence shown in SEQ ID NO: 3 or the HCDR3 obtained by amino acid addition, elimination or substitution reaction with the amino acid sequence shown in SEQ ID NO: 3 differs by no more than 2 amino acids.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain variable region of an antibody
  • the heavy chain variable region comprises HCDR1, HCDR2, HCDR3 comprising the
  • the amino acid sequence shown in ID NO: 1 has at least 70% sequence identity
  • the HCDR2 comprises an amino acid sequence that is at least 70% identical to the amino acid sequence shown in SEQ ID NO: 2
  • the HCDR3 comprises an amino acid sequence with at least 70% sequence identity to the amino acid sequence shown in SEQ ID NO: 2 :
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain variable region of an antibody
  • the heavy chain variable region comprises HCDR1, HCDR2, HCDR3, and the HCDR1 comprises SEQ ID
  • the HCDR2 comprises SEQ ID NO: The amino acid sequence shown in: 2 or the amino acid sequence obtained by amino acid addition, elimination or substitution reaction with the amino acid sequence shown in SEQ ID NO: 2 with no more than 2 amino acid differences
  • Described HCDR3 comprises SEQ ID NO: The amino acid sequence shown in 3 or the amino acid sequence obtained by amino acid addition, elimination or substitution reaction has no more than 2 amino acid differences with the amino acid sequence shown in SEQ ID NO: 3.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain variable region of an antibody
  • the heavy chain variable region comprises an amino acid sequence selected from the group consisting of: (a) SEQ The amino acid sequence set forth in ID NO: 4; (b) an amino acid sequence at least 85%, 90%, 95% or 99% identical to the amino acid sequence set forth in SEQ ID NO: 4; and (c) by addition , the amino acid sequence obtained by the elimination or substitution reaction with the amino acid sequence shown in SEQ ID NO: 4 has 1 or more differences.
  • TGF ⁇ inhibitor binds TGF ⁇ 1, TGF ⁇ 2, or TGF ⁇ 3.
  • TGF[beta] inhibitor binds TGF[beta] or transforming growth factor beta receptor (TGF[beta]R).
  • TGF ⁇ inhibitor binds TGF ⁇ type I receptor (TGF ⁇ RI), TGF ⁇ type II receptor (TGF ⁇ RII) or TGF ⁇ type III receptor (TGF ⁇ RIII).
  • the TGF ⁇ inhibitor comprises: 1) an anti-TGF ⁇ antibody or antigen-binding fragment thereof; 2) an anti-TGF ⁇ R antibody or antigen-binding fragment thereof; 3) a small molecule TGF ⁇ inhibitor; 4) comprising transforming growth A protein of factor beta receptor II (TGF ⁇ RII) or a functionally active fragment thereof, or a variant or biosimilar thereof, or a combination thereof.
  • TGF ⁇ RII transforming growth A protein of factor beta receptor II
  • the anti-TGF ⁇ antibody or antigen-binding fragment thereof comprises a heavy chain variable region of an antibody, wherein the heavy chain variable region comprises HCDR1, HCDR2, HCDR3, each of which is associated with HCDR1 of a Fresolimumab molecule, respectively , HCDR2, HCDR3 have at least about 80% sequence identity.
  • the heavy chain variable region has at least about 80% sequence identity with the heavy chain variable region of a Fresolimumab molecule.
  • the heavy chain has at least about 80% sequence identity with the heavy chain of a Fresolimumab molecule.
  • the anti-TGF ⁇ antibody or antigen-binding fragment thereof comprises a light chain variable region of an antibody
  • the light chain variable region comprises LCDR1, LCDR2, LCDR3, each of which is associated with LCDR1 of a Fresolimumab molecule, respectively
  • LCDR2, LCDR3 have at least about 80% sequence identity.
  • the light chain variable region has at least about 80% sequence identity with the light chain variable region of a Fresolimumab molecule.
  • the light chain has at least about 80% sequence identity with the light chain of a Fresolimumab molecule.
  • the anti-TGF ⁇ antibody or antigen-binding fragment thereof may comprise: Fresolimumab, or an antigen-binding fragment thereof, or a variant or biosimilar thereof.
  • the small molecule TGF ⁇ inhibitor comprises the following compounds: SB525334, SD-208, SB431542, LY2109761, LY2157299, GW788388, RepSox, SIS3, LDN-193189, EW-7197, LY364947, or a combination thereof .
  • the TGF ⁇ RII has a wild-type human TGF ⁇ receptor type 2 isoform A sequence (eg, the amino acid sequence of NCBI Reference Sequence (Ref Seq) Accession No. NP_001020018), or has a wild-type human A polypeptide of the TGF ⁇ receptor type 2 isoform B sequence (eg, the amino acid sequence of NCBI Ref Seq Accession No. NP_003233), or a polypeptide having a sequence substantially identical to its amino acid sequence.
  • a wild-type human TGF ⁇ receptor type 2 isoform A sequence eg, the amino acid sequence of NCBI Reference Sequence (Ref Seq) Accession No. NP_001020018
  • a wild-type human A polypeptide of the TGF ⁇ receptor type 2 isoform B sequence eg, the amino acid sequence of NCBI Ref Seq Accession No. NP_003233
  • the TGF ⁇ RII or a functionally active fragment thereof comprises: human TGF ⁇ RII extracellular domain (ECD), any portion of NCBI Ref Seq Accession No. NP_001020018 or NCBI Ref Seq Accession No. NP_003233, or substantially identical to its amino acid sequence sequence.
  • ECD human TGF ⁇ RII extracellular domain
  • TGF ⁇ RII or functionally active fragment thereof comprises:
  • (c) has an amino acid sequence with one or more amino acids added, deleted and/or substituted compared to the amino acid sequence shown in SEQ ID NO:6.
  • bifunctional immune checkpoint/TGF ⁇ inhibitor is a fusion protein.
  • the PD-L1/TGF ⁇ dual inhibitor or PD-1/TGF ⁇ dual inhibitor is a fusion protein.
  • the dual PD-L1//TGF ⁇ inhibitor comprises a PD-L1 targeting moiety and a TGF ⁇ receptor domain comprising transforming growth factor beta receptor II (TGF ⁇ RII ) or a functionally active fragment thereof.
  • TGF ⁇ RII transforming growth factor beta receptor II
  • the dual PD-L1//TGF ⁇ inhibitor comprises a polypeptide
  • the polypeptide comprises at least: (i) the heavy chain variable region of an anti-PD-L1 antibody; and (ii) TGF ⁇ RII or its functionally active fragments.
  • the anti-PD-L1 antibody heavy chain variable region comprises HCDR1, HCDR2, HCDR3; wherein the HCDR1 comprises at least about 70% sequence identity to the amino acid sequence set forth in SEQ ID NO:1 Amino acid sequence, the HCDR2 comprises an amino acid sequence with at least about 70% sequence identity to the amino acid sequence set forth in SEQ ID NO:2, and the HCDR3 comprises an amino acid sequence with at least about 70% sequence identity to the amino acid sequence set forth in SEQ ID NO:3 amino acid sequence; or
  • HCDR1, HCDR2, and HCDR3 have at least about 80%, about 85%, 90%, 95% or 99% sequence identity with HCDR1, HCDR2, and HCDR3 of the following molecules, respectively: Atezolizumab, Avelumab, BMS-936559, MPDL3280A (RG7446) or durvalumab (MEDI-4736).
  • the anti-PD-L1 antibody heavy chain variable region comprises HCDR1, HCDR2, HCDR3, and the HCDR1 comprises the amino acid sequence shown in SEQ ID NO: 1 or by amino acid addition, elimination or substitution
  • the amino acid sequence obtained by the reaction has no more than 2 amino acid differences with the amino acid sequence shown in SEQ ID NO: 1;
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO: 2 or through amino acid addition, elimination or substitution reaction
  • the obtained amino acid sequence with the amino acid sequence shown in SEQ ID NO: 2 is no more than 2 amino acid differences;
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO: 3 or is obtained by amino acid addition, elimination or substitution reaction.
  • the resulting amino acid sequence has no more than 2 amino acid differences with the amino acid sequence shown in SEQ ID NO: 3.
  • the anti-PD-L1 antibody heavy chain variable region comprises an amino acid sequence selected from the group consisting of: (a) the amino acid sequence shown in SEQ ID NO: 4; (b) the same as SEQ ID NO: : the amino acid sequence shown in 4 has at least about 85%, 90%, 95% or 99% sequence identity; (c) obtained by addition, elimination or substitution reaction with the amino acid sequence shown in SEQ ID NO: 4 An amino acid sequence with 1 or more differences in amino acid sequence; and (d) an amino acid sequence with at least about 80% sequence identity to the heavy chain variable region of Atezolizumab, Avelumab, BMS-936559, MPDL3280A, or durvalumab .
  • TGF ⁇ RII or functionally active fragment thereof comprises:
  • (c) has an amino acid sequence with one or more amino acids added, deleted and/or substituted compared to the amino acid sequence shown in SEQ ID NO:6.
  • polypeptide further comprises a linker linking the C-terminus of the heavy chain variable region of the anti-PD-L1 antibody or antigen-binding fragment thereof to the TGF ⁇ RII or its function
  • linker linking the C-terminus of the heavy chain variable region of the anti-PD-L1 antibody or antigen-binding fragment thereof to the TGF ⁇ RII or its function
  • the N-termini of the active fragments are linked.
  • the polypeptide further comprises CH2, CH3 domains, and the polypeptide comprises, from the N-terminus to the C-terminus, the heavy chain variable region (VH), CH2, CH3 domains and TGF ⁇ RII or its functionally active fragment, the C-terminus of the CH3 domain and the N-terminus of the TGF ⁇ RII or its functionally active fragment are connected by a linker.
  • VH heavy chain variable region
  • CH2, CH3 domains and TGF ⁇ RII or its functionally active fragment the C-terminus of the CH3 domain and the N-terminus of the TGF ⁇ RII or its functionally active fragment are connected by a linker.
  • CH2 domains are derived from IgG.
  • the CH2, CH3 domains are all derived from IgGl, IgG2, IgG3 or IgG4.
  • linker is a peptide linker
  • amino acid sequence of the peptide linker is (G 4 S) x , wherein x is any integer from 3-6.
  • the peptide linker comprises an amino acid sequence selected from the group consisting of: (a) the amino acid sequence set forth in SEQ ID NO:5; (b) having the amino acid sequence set forth in SEQ ID NO:5 An amino acid sequence having at least about 85%, 90%, 95% or 99% sequence identity; and (c) an amino acid sequence obtained by addition, elimination or substitution with the amino acid sequence shown in SEQ ID NO: 5 having 1 or More differential amino acid sequences.
  • polypeptide comprises an amino acid sequence selected from the group consisting of: (a) the amino acid sequence set forth in SEQ ID NO:7; (b) having at least the amino acid sequence set forth in SEQ ID NO:7 An amino acid sequence of about 85%, 90%, 95% or 99% sequence identity; (c) obtained by addition, elimination or substitution reaction with the amino acid sequence shown in SEQ ID NO: 7 has 1 or more Differential amino acid sequences.
  • the PD-L1//TGF ⁇ dual inhibitor comprises two of the aforementioned polypeptides.
  • the dual PD-L1/TGF ⁇ inhibitor comprises a first polypeptide and a second polypeptide
  • the first polypeptide comprises at least: (i) the heavy chain of an anti-PD-L1 antibody and (ii) TGF ⁇ RII or a functionally active fragment thereof
  • the second polypeptide comprises at least the light chain variable region of an anti-PD-L1 antibody
  • the heavy chain variable region of the first polypeptide and The light chain variable regions of the second polypeptide are capable of specifically binding PD-L1 when combined.
  • the first polypeptide further comprises a linker that connects the C-terminus of the heavy chain variable region of the anti-PD-L1 antibody or antigen-binding fragment thereof to the TGF ⁇ RII or the N-terminus of a functionally active fragment thereof.
  • the dual PD-L1/TGF ⁇ inhibitor comprises a first polypeptide and a second polypeptide, wherein:
  • the first polypeptide sequentially comprises the heavy chain variable region (VH) of the anti-PD-L1 antibody, the CH1 domain, and TGF ⁇ RII or a functionally active fragment thereof from the N-terminus to the C-terminus, and the C-terminus of the CH1 domain is related to the The N-terminus of the TGF ⁇ RII or its functionally active fragment is connected by a linker; and the second polypeptide comprises a light chain variable region (VL) and a light chain constant region (CL) of an anti-PD-L1 antibody from the N-terminus to the C-terminus ).
  • the dual PD-L1/TGF ⁇ inhibitor comprises a first polypeptide and a second polypeptide, wherein:
  • the first polypeptide comprises the heavy chain variable region (VH), CH1, CH2, CH3 domains and TGF ⁇ RII or functionally active fragments thereof of the anti-PD-L1 antibody in sequence from the N terminus to the C terminus, and the CH3 domain
  • the C-terminus is connected with the N-terminus of the TGF ⁇ RII or its functionally active fragment through a linker
  • the second polypeptide comprises the light chain variable region (VL), light chain of an anti-PD-L1 antibody from the N-terminus to the C-terminus constant region (CL).
  • linker is a peptide linker
  • the first polypeptide has at least about 80% sequence identity to a functionally identical polypeptide of M7824 or SHR-1701.
  • the second polypeptide has at least about 80% sequence identity to a functionally identical polypeptide of M7824 or SHR-1701.
  • the dual PD-L1/TGF ⁇ inhibitor comprises M7824, SHR-1701, or a variant or biosimilar thereof, or a combination thereof.
  • the PI3K inhibitor is a dual PI3K delta/gamma inhibitor.
  • the PI3K inhibitor comprises a compound of formula (IA-I), (IA-II), (IA-III), or (IA-IV):
  • Each occurrence of R is independently selected from hydrogen, halogen, -OR a , CN, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl, substituted or unsubstituted C 2-6 alkynyl, substituted or unsubstituted C 3-8 cycloalkyl and substituted or unsubstituted heterocyclic groups;
  • Cy 1 is selected from substituted or unsubstituted C 3-8 cycloalkyl, substituted or unsubstituted 3- to 15-membered heterocyclic group having at least one heteroatom selected from N, O and S, Substituted or unsubstituted C6-20 aryl and substituted or unsubstituted monocyclic groups of 5- to 14-membered heteroaryl groups having one or more heteroatoms selected from N, O and S;
  • Each occurrence of R a may be the same or different and independently selected from hydrogen, halogen, hydroxy, cyano, substituted or unsubstituted C 1-6 alkyl; -NR c R d , where R c , R d independently selected from hydrogen, halogen, hydroxy, cyano, substituted or unsubstituted C1-6 alkyl and C1-6 alkoxy, and -ORc , wherein Rc is substituted or unsubstituted C 1-6 alkyl;
  • n is an integer from 1 to 4.
  • q 0, 1 or 2;
  • Each occurrence of X is independently selected from CR 3 or N;
  • R is independently selected from hydrogen, hydroxy, halogen, carboxyl, cyano, nitro, substituted or unsubstituted C 1-8 alkyl, substituted or unsubstituted C 1-8 Alkoxy, substituted or unsubstituted C 2-10 alkenyl, substituted or unsubstituted C 2-12 alkynyl, substituted or unsubstituted C 6-20 aryl, substituted or unsubstituted Substituted C 6-20 aryl C 1-8 alkyl, substituted or unsubstituted C 3-20 cycloalkyl, substituted or unsubstituted C 3-20 cycloalkyl C 1-8 alkane base, substituted or unsubstituted C 3-8 cycloalkenyl C 1-8 alkyl, substituted or unsubstituted C 3-8 cycloalkenyl, substituted or unsubstituted with one or more 5- to 14-membered heteroaryl with
  • the PI3K inhibitor comprises a compound of formula (IA-V):
  • R is independently selected from hydrogen, halogen, -OR a , CN, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl, substituted or Unsubstituted C 2-6 alkynyl, substituted or unsubstituted C 3-8 cycloalkyl and substituted or unsubstituted heterocyclic groups;
  • a 3-10 membered ring including carbon atoms bound to R1 and R2, the ring may optionally include one or more heteroatoms that are the same or different and selected from O, NR a and S;
  • Each occurrence of X is independently selected from CR 3 or N;
  • Each occurrence of R is independently selected from hydrogen, hydroxy, halogen, carboxy, cyano, nitro, substituted or unsubstituted C 1-8 alkyl, substituted or unsubstituted C 1-8 alkane oxy, substituted or unsubstituted C 2-10 alkenyl, substituted or unsubstituted C 2-12 alkynyl, substituted or unsubstituted C 6-20 aryl, substituted or unsubstituted Substituted C 6-20 aryl C 1-8 alkyl, substituted or unsubstituted C 3-20 cycloalkyl, substituted or unsubstituted C 3-20 cycloalkyl C 1-8 alkyl , substituted or unsubstituted C 3-8 cycloalkenyl C 1-8 alkyl, substituted or unsubstituted C 3-8 cycloalkenyl, substituted or unsubstituted with one or more options 5- to 14-membered heteroary
  • R 5 is hydrogen, C 1-6 alkyl or halogen
  • the PI3K inhibitor comprises a compound of formula (IA-VI):
  • Each occurrence of R is independently selected from hydrogen, halogen, -OR a , CN, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl, substituted or unsubstituted C 2-6 alkynyl, substituted or unsubstituted C 3-8 cycloalkyl and substituted or unsubstituted heterocyclic groups;
  • Each occurrence of X is independently selected from CR 3 or N;
  • Each occurrence of R is independently selected from hydrogen, hydroxy, halogen, carboxy, cyano, nitro, substituted or unsubstituted C 1-8 alkyl, substituted or unsubstituted C 1-8 alkane oxy, substituted or unsubstituted C 2-10 alkenyl, substituted or unsubstituted C 2-12 alkynyl, substituted or unsubstituted C 6-20 aryl, substituted or unsubstituted Substituted C 6-20 aryl C 1-8 alkyl, substituted or unsubstituted C 3-20 cycloalkyl, substituted or unsubstituted C 3-20 cycloalkyl C 1-8 alkyl , substituted or unsubstituted C 3-8 cycloalkenyl C 1-8 alkyl, substituted or unsubstituted C 3-8 cycloalkenyl, substituted or unsubstituted with one or more options 5- to 14-membered heteroary
  • R 5 is hydrogen, C 1-6 alkyl or halogen
  • n 0, 1, 2, 3, or 4;
  • P is 0, 1, 2, 3, 4 or 5.
  • R is hydrogen, halogen, substituted or unsubstituted Ci_6 alkyl, or OR a .
  • Cy 1 is selected from:
  • R 1 and R 2 each independently represent hydrogen or substituted or unsubstituted C 1-6 alkyl.
  • R is iodo, cyano, substituted or unsubstituted alkyl, substituted or unsubstituted alkynyl, substituted or unsubstituted aryl, substituted or unsubstituted Substituted or unsubstituted heteroaryl.
  • X is CR 3 and each occurrence of R 3 is independently hydrogen, halogen, hydroxy, or NH 2 .
  • the PI3K inhibitor comprises:
  • the PI3K inhibitor comprises: 2-(1-(9H-purin-6-ylamino)propyl)-3-(3-fluorophenyl)-4H-chromene-4 - Ketones, or their enantiomers, mixtures of enantiomers, mixtures of two or more diastereomers, or isotopic variants; or pharmaceutically acceptable salts, solvates thereof , hydrate or prodrug.
  • the PI3K inhibitor is (S)-2-(1-(9H-purin-6-ylamino)propyl)-3-(3-fluorophenyl)-4H-chrome En-4-one or a pharmaceutically acceptable salt, solvate, hydrate or prodrug thereof.
  • the pharmaceutical combination includes a dual PD-L1/TGF ⁇ inhibitor and a PI3K inhibitor.
  • the dual PD-L1/TGF ⁇ inhibitor comprises an anti-PD-L1 antibody or an antigen-binding fragment thereof and TGF ⁇ RII or a functionally active fragment thereof.
  • the PD-L1/TGF ⁇ dual inhibitor comprises two polypeptides, wherein the polypeptides comprise an amino acid sequence selected from the group consisting of: (a) the amino acid sequence shown in SEQ ID NO:7; (b) an amino acid sequence having at least 85%, 90%, 95% or 99% sequence identity with the amino acid sequence shown in SEQ ID NO: 7; (c) obtained by addition, elimination or substitution reaction with SEQ ID NO: 7
  • the amino acid sequence shown in ID NO: 7 has 1 or more different amino acid sequences.
  • the PI3K inhibitor is a dual PI3K delta/gamma inhibitor.
  • the PI3K inhibitor is (S)-2-(1-(9H-purin-6-ylamino)propyl)-3-(3-fluorophenyl)-4H-chrome En-4-one or a pharmaceutically acceptable salt, solvate, hydrate or prodrug thereof.
  • the dual PD-L1/TGF ⁇ inhibitor and the PI3K inhibitor are not mixed with each other in the pharmaceutical combination.
  • the dual PD-L1/TGF ⁇ inhibitor and the PI3K inhibitor are each independently present in separate containers.
  • the pharmaceutical combination may further include a third active substance.
  • the third active substance can be selected from vincristine, vinblastine, vindesine, etoposide, docetaxel, paclitaxel, irinotecan, vinorelbine, mitoxantre Any one of quinone, vinflunine, and topotecan or any combination thereof.
  • the present application provides a pharmaceutical composition
  • a pharmaceutical composition comprising the aforementioned PI3K inhibitor and a second therapeutic agent, and optionally one or more pharmaceutically acceptable carriers or excipients.
  • the second therapeutic agent is the aforementioned dual PD-L1/TGF ⁇ inhibitor.
  • the PI3K inhibitor is the aforementioned dual PI3K delta/gamma inhibitor.
  • the second therapeutic agent and the PI3K inhibitor are present in a single or separate dosage form.
  • the present application provides a method for treating and/or preventing tumors, comprising administering to a subject in need thereof: an effective amount of the aforementioned pharmaceutical combination or the aforementioned pharmaceutical composition.
  • the method comprises administering to a subject in need thereof: an effective amount of the aforementioned PI3K inhibitor and the aforementioned inhibitor of immune checkpoints. In certain embodiments, the method comprises administering to a subject in need thereof: an effective amount of the aforementioned PI3K inhibitor and the aforementioned TGF ⁇ inhibitor. In certain embodiments, the method comprises administering to a subject in need thereof: an effective amount of the aforementioned PI3K inhibitor, the aforementioned TGF ⁇ inhibitor, and the aforementioned inhibitor of immune checkpoints.
  • the method comprises administering to a subject in need thereof: an effective amount of the aforementioned PI3K inhibitor and the aforementioned dual PD-L1/TGF ⁇ inhibitor.
  • the PI3K inhibitor is administered concurrently with the dual PD-L1/TGF ⁇ inhibitor.
  • the PI3K inhibitor is administered after the dual PD-L1/TGF ⁇ inhibitor.
  • the PI3K inhibitor is administered before the dual PD-L1/TGF ⁇ inhibitor.
  • it also includes administering to a subject in need thereof an effective amount of the aforementioned small molecule TGF ⁇ inhibitor.
  • the tumor includes solid tumors and non-solid tumors.
  • the tumor comprises a tumor with abnormal PI3K expression.
  • the tumor comprises a tumor with abnormal PD-L1 expression.
  • the tumor comprises a tumor with aberrant expression of TGF[beta].
  • the tumor comprises a gastrointestinal tumor, melanoma, or lymphoma.
  • the tumor is delayed in resistance to the PI3K inhibitor.
  • MRD minimal residual disease
  • the present application provides the use of the aforementioned pharmaceutical combination or the aforementioned pharmaceutical composition in the preparation of a medicament for treating and/or preventing tumors.
  • the use of the aforementioned PI3K inhibitors and the aforementioned inhibitors of immune checkpoints in the preparation of medicaments for treating and/or preventing tumors For example, the use of the aforementioned PI3K inhibitor and the aforementioned TGF ⁇ inhibitor in the preparation of a medicament for treating and/or preventing tumors.
  • the present application provides the aforementioned pharmaceutical combination or the aforementioned pharmaceutical composition for treating and/or preventing tumors.
  • the present application provides a method of inhibiting tumor growth, the method comprising contacting the tumor with the aforementioned pharmaceutical combination or the aforementioned pharmaceutical composition.
  • the contacting is in an in vitro or ex vivo environment.
  • the method comprises contacting the tumor with the aforementioned PI3K inhibitor and the aforementioned inhibitor of immune checkpoints. In certain embodiments, the method comprises contacting the tumor with the aforementioned PI3K inhibitor and the aforementioned TGF ⁇ inhibitor. In certain embodiments, the method comprises contacting the tumor with the aforementioned PI3K inhibitor, the aforementioned inhibitor of an immune checkpoint, and the aforementioned TGF ⁇ inhibitor.
  • the present application provides a method of inhibiting tumor growth, the method comprising contacting the tumor with the aforementioned PI3K inhibitor and the aforementioned dual PD-L1/TGF ⁇ inhibitor.
  • the present application provides a kit comprising: (1) a first container, and the aforementioned PI3K inhibitor located in the first container; (2) a second container, and located in the first container The aforementioned dual PD-L1/TGF ⁇ inhibitor in two containers.
  • the present application provides a kit, which may include: (1) a first container, and the aforementioned PI3K inhibitor located in the first container; (2) a second container, and located in the first container The aforementioned inhibitor of immune checkpoint or the aforementioned TGF ⁇ inhibitor in the second container.
  • the kit further comprises (3) a third container, and the aforementioned inhibitor of immune checkpoint or the aforementioned TGF ⁇ inhibitor located in the third container, the agent in the third container Different from the medicament in the second container.
  • the dosage form of the drug in the kit is an oral dosage form or an injection dosage form.
  • the kit also contains instructions.
  • Figures 1A-1F show the results of combined administration of CN401 (Tenalisib) (150 mg/kg) and CN202 (WBP1126) (5 mg/kg) in an A20 mouse B-cell lymphoma animal model.
  • Figures 2A-2F show the results of combined administration of CN401 (Tenalisib) (150 mg/kg) and CN202 (WBP1126) (15 mg/kg) in an A20 mouse B-cell lymphoma animal model.
  • Figures 3A-B show the results of combined administration of CN401 (Tenalisib) (150 mg/kg) and CN202 (WBP1126) (5 mg/kg)/(15 mg/kg) in an A20 mouse B-cell lymphoma animal model.
  • Figures 4A-4F show the results of combined administration of CN202 (WBP1126) (5 mg/kg) and CN401 (Tenalisib) (150 mg/kg) in an A20 mouse B-cell lymphoma animal model.
  • Figures 5A-5F show the results of combined administration of CN202 (WBP1126) (5 mg/kg) and Alpelisib (50 mg/kg) in an A20 mouse B-cell lymphoma animal model.
  • Figures 6A-6F show the results of combined administration of CN401 (Tenalisib) (150 mg/kg) and anti-PD-L1 antibody (Atezolizumab) (5 mg/kg) in an A20 mouse B-cell lymphoma animal model.
  • Figures 7A-7B show CN401 (Tenalisib) (150 mg/kg), CN202 (WBP1126) (15 mg/kg) and nab-pac (10 mg/kg) in mouse EMT-60 (mouse breast cancer) cell) tumor growth curve and body weight growth curve.
  • Figures 8A-8G show tumor growth curves of mouse EMT-60 (mouse breast cancer cells) in different administration groups.
  • PI3K inhibitor or “PI3K inhibitor” generally refers to any inhibitor of PI3K.
  • PI3Ks are members of a unique and conserved family of intracellular lipid kinases that phosphorylate phosphatidylinositol or the 3'-OH group on phosphoinositides.
  • the PI3K family includes kinases with different substrate specificities, expression patterns, and regulatory patterns (see, eg, Katso et al, 2001, Annu. Rev. Cell Dev. Biol. 17, 615-675; Foster, F.M. et al, 2003, J Cell Sci 116, 3037-3040).
  • Class I PI3Ks (eg, p110 ⁇ , p110 ⁇ , p110 ⁇ , and p110 ⁇ are typically activated by tyrosine kinases or G-protein coupled receptors to generate PIP3, which binds downstream mediators such as those in the Akt/PDK1 pathway, mTOR, the Tec family Kinases and Rho family GTPases.
  • Class II PI3Ks (eg, PI3K-C2 ⁇ , PI3K-C2 ⁇ , PI3K-C2 ⁇ ) and class III PI3Ks (eg, Vps34) are synthesized in PI(3)P and PI(3,4)P2 in It plays a key role in intracellular trafficking.
  • PI3K inhibitors inhibit PI3K-alpha, PI3K-beta, PI3K-gamma, and PI3K-delta isoforms or combinations thereof.
  • the PI3K inhibitor can be the dual PI3K delta/gamma inhibitor Tenalisib, 2-(1-(9H-purin-6-ylamino)propyl)-3-(3-fluorophenyl)-4H-chromene-4 - Ketones, the structural formula of which is as follows:
  • the PI3K inhibitor can be the PI3K inhibitor Alpelisib, (2S)-1-N-[4-methyl-5-[2-(1,1,1-trifluoro-2-methylpropan-2-yl ) pyridin-4-yl]-1,3-thiazol-2-yl]pyrrolidine-1,2-dicarboxamide, its structural formula is as follows:
  • a PI3K inhibitor generally refers to a compound that inhibits one or more PI3K isoforms with an IC50 of less than about 1000 nM, less than about 900 nM, less than about 800 nM, less than about 700 nM, less than about 600 nM, less than about 500 nM, Less than about 400 nM, less than about 300 nM, less than about 200 nM, less than about 100 nM, less than about 75 nM, less than about 50 nM, less than about 25 nM, less than about 20 nM, less than about 15 nM, less than about 10 nM, less than about 10 nM, less than about 5 nM, or less than about 1nM.
  • alkyl generally refers to a straight or branched chain consisting only of carbon atoms and hydrogen atoms, free of unsaturation, having 1-8 carbon atoms and connected to the remainder of the molecule by a single bond Hydrocarbon radicals such as methyl, ethyl, n-propyl, 1-methylethyl (isopropyl), n-butyl, n-pentyl and 1,1-dimethylethyl (tert-butyl).
  • (C 1-6 )alkyl refers to an alkyl group as defined above having up to 6 carbon atoms.
  • alkenyl generally refers to a straight or branched aliphatic hydrocarbon group containing a carbon-carbon double bond and possibly having from about 2 to about 10 carbon atoms, such as vinyl, 1-propenyl , 2-propenyl (allyl), isopropenyl, 2-methyl-1-propenyl, 1-butenyl and 2-butenyl.
  • ( C2-6 )alkenyl refers to an alkenyl group as defined above having up to 6 carbon atoms.
  • alkynyl generally refers to a straight or branched chain hydrocarbon group having at least one carbon-carbon triple bond and having 2 to 12 carbon atoms (currently preferred are groups having 2 to 10 carbon atoms). groups), such as ethynyl, propynyl and butynyl.
  • groups such as ethynyl, propynyl and butynyl.
  • ( C2-6 )alkynyl refers to an alkynyl group as defined above having up to 6 carbon atoms.
  • alkoxy generally refers to an alkyl, cycloalkyl, cycloalkylalkyl group attached to the rest of the molecule through an oxygen bond as defined above.
  • substituted alkoxy refers to an alkoxy group wherein the alkyl group is substituted (ie, -O-(substituted alkyl)), wherein the term “substituted alkyl” is the same as the term “substituted alkyl” above for " Alkyl” has the same definition.
  • alkoxy refers to the group -O-alkyl, including 1-8 carbon atoms in straight chain, branched chain, cyclic configurations, and combinations thereof attached to the parent structure through an oxygen. Examples include methoxy, ethoxy, propoxy, isopropoxy, cyclopropoxy and cyclohexyloxy.
  • cycloalkyl generally refers to non-aromatic mono- or polycyclic ring systems of about 3-12 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
  • polycyclic cycloalkyl groups include perhydronaphthyl, adamantyl and norbornyl groups, bridged ring groups and spiro groups such as spiro(4,4)nonan-2-yl.
  • ( C3-8 )cycloalkyl refers to a cycloalkyl group as defined above having up to 8 carbon atoms.
  • cycloalkylalkyl generally refers to a group containing about 3 to 8 carbon atoms directly attached to an alkyl group which in turn results in a stable structure formed from the alkyl group. Ring-containing groups attached to the main structure at any carbon atom, such as cyclopropylmethyl, cyclobutylethyl, and cyclopentylethyl.
  • cycloalkenyl generally refers to ring-containing groups containing about 3 to 8 carbon atoms with at least one carbon-carbon double bond, such as cyclopropenyl, cyclobutenyl, and cyclopentene base.
  • cycloalkenylalkyl refers to a ring-containing group attached directly to an alkyl group, which in turn is attached to the primary structure at any carbon atom from the alkyl group that results in the formation of a stable structure.
  • aryl generally refers to aromatic groups having 6-20 carbon atoms, such as phenyl, naphthyl, tetrahydronaphthyl, indanyl, and biphenyl.
  • arylalkyl generally refers to an aryl group as defined above attached directly to an alkyl group as defined above, eg -CH2C6H5 and -C2H5C6 H5 .
  • heterocycle generally refers to a non-aromatic 3-15 membered ring group consisting of carbon atoms and at least one heteroatom selected from nitrogen, phosphorus, oxygen and sulfur.
  • a heterocyclyl group may be a mono-, bi-, tri- or tetracyclic ring system, which may include fused, bridged, or spiro ring systems, and the nitrogen, phosphorus, carbon, oxygen, or Sulfur atoms can be optionally oxidized to various oxidation states.
  • nitrogen atoms can be optionally quaternized.
  • a heterocyclyl group can be attached to the main structure at any heteroatom or carbon atom that results in the formation of a stable structure.
  • heterocyclyl generally refers to a heterocyclic-type cyclyl group as defined above. Heterocyclic-type ring groups can be attached to the main structure at any heteroatom or carbon atom that results in the formation of a stable structure.
  • heterocyclylalkyl generally refers to a heterocyclic-type ring group as defined above directly bonded to an alkyl group.
  • a heterocyclylalkyl group can be attached to the primary structure at a carbon atom in the alkyl group that results in the formation of a stable structure.
  • heterocycloalkyl groups include, but are not limited to, dioxolanyl, thienyl[l,3]dithianyl, decahydroisoquinolinyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, Isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidyl, 2-oxopyrrolidinyl, oxazolidine base, piperidinyl, piperazinyl, 4-piperidinyl, pyrrolidinyl, pyrazolidine, quinuclidinyl, thiazolidinyl, tetrahydrofuranyl, trithianyl, tetrahydropyranyl, sulfur morpholinyl, thiamorpholinyl, 1-oxo-
  • heteroaryl generally refers to an optionally substituted 5-14 membered aromatic ring having one or more heteroatoms selected from N, O, and S as ring atoms.
  • Heteroaryl groups can be mono-, bi- or tricyclic ring systems.
  • heterocycle or “heteroaryl” groups include, but are not limited to, oxazolyl, thiazolyl, imidazolyl, pyrrolyl, furyl, pyridyl, pyrimidinyl, pyrazinyl, benzofuranyl , indolyl, benzothiazolyl, benzoxazolyl, carbazolyl, quinolinyl, isoquinolinyl, azetidinyl, acridinyl, benzodioxolyl, benzodi oxanyl, benzofuranyl, carbazolyl, cinnolinyl, dioxolane, indolizinyl, naphthyridinyl, perhydroazepinyl, phenazinyl, phenothiazinyl, phenoxazine base, phthalazinyl, pteridyl, purinyl, quina
  • a heteroaryl ring group can be attached to the main structure at any heteroatom or carbon atom that results in the formation of a stable structure.
  • substituted heteroaryl also includes ring systems substituted with one or more oxide (-O-) substituents (eg, pyridyl N-oxide).
  • heteroarylalkyl generally refers to a heteroaryl ring group as defined above bonded directly to an alkyl group.
  • a heteroarylalkyl group can be attached to the primary structure at any carbon atom from the alkyl group that results in the formation of a stable structure.
  • ring generally refers to a ring containing 3-10 carbon atoms.
  • Immune checkpoints generally refers to a group of molecules on the cell surface of CD4 cells and CD8 T cells. These molecules can effectively act as “brakes” to downregulate or suppress antitumor immune responses.
  • Immune checkpoint molecules include but are not limited to programmed cell death 1 (PD-1), programmed cell death ligand 1 (PD-L1), programmed cell death ligand 2 (PD-L2), lymphocyte activation gene- 3 (LAG-3; also known as CD223), Galectin-3, B and T lymphocyte attenuator (BTLA), T-cell membrane protein 3 (TIM3), Galectin-9 (GAL9), B7 -H1, B7-H3, B7-H4, T-cell immunoreceptors with Ig and ITIM domains (TIGIT/Vstm3/WUCAM/VSIG9), V-domain Ig inhibitor of T-cell activation (VISTA), Glucocorticoid-induced tumor necrosis factor receptor-related (GITR) protein,
  • PD-1 programme
  • inhibitor of an immune checkpoint generally refers to a molecule that inhibits, reduces or interferes with the activity of inhibitory checkpoint molecules.
  • inhibitory checkpoint molecules downregulate immune responses (eg, T-cell activation) by delivering negative signals to T cells upon their binding by ligands or counterreceptors.
  • the checkpoint inhibitors used with the methods and compositions provided herein can directly inhibit the activity of the inhibitory checkpoint molecule or reduce the expression of the inhibitory checkpoint molecule or interfere with the inhibitory checkpoint molecule and Interaction of binding partners (eg, ligands).
  • inhibitors of immune checkpoints include, but are not limited to, proteins, polypeptides, peptides, antisense oligonucleotides, antibodies, antibody fragments, or RNA molecules (eg, targeting the expression of inhibitory checkpoint molecules) inhibitory RNA molecules). Its inhibition can be performed at the DNA, RNA or protein level.
  • inhibitory nucleic acids eg, dsRNA, siRNA, or shRNA
  • dsRNA dsRNA, siRNA, or shRNA
  • the inhibitor of the inhibitory signal is a polypeptide that binds to an immune checkpoint, eg, a soluble ligand (eg, PD-1-Ig), an antibody or antigen-binding fragment thereof; - Antibodies or fragments thereof that bind to L1, PD-L2, CTLA4, TIM3, LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 and/or TGFR ⁇ or a combination thereof.
  • an immune checkpoint eg, a soluble ligand (eg, PD-1-Ig), an antibody or antigen-binding fragment thereof; - Antibodies or fragments thereof that bind to L1, PD-L2, CTLA4, TIM3, LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 and/or TGFR ⁇ or a combination thereof.
  • TGF ⁇ RII or "TGF ⁇ receptor II” generally refers to a protein having a wild-type human TGF ⁇ receptor type 2 isoform A sequence (eg, the amino acid sequence of NCBI Reference Sequence (Ref Seq) Accession No. NP_001020018).
  • Polypeptides, or polypeptides having a wild-type human TGF ⁇ receptor type 2 isoform B sequence (such as the amino acid sequence of NCBI Ref Seq Accession No. NP_003233) or a polypeptide having a sequence substantially identical to their amino acid sequence.
  • TGF ⁇ RII can retain at least 0.1%, 0.5%, 1%, 5%, 10%, 25%, 35%, 50%, 75%, 90%, 95%, or 99% of the binding activity of the wild-type sequence TGF ⁇ .
  • TGF ⁇ RII fragment capable of binding TGF ⁇ generally refers to NCBI Ref Seq Accession No. NP_001020018 or NCBI Ref Seq Accession No. NP_003233 or any portion of substantially identical sequence thereto, the fragment length is at least 20 (eg at least 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 175, or 200) amino acids and retain at least a portion of the TGF ⁇ binding activity of the wild-type receptor or wild-type fragment thereof (e.g., at least 0.1%, 0.5%, 1%, 5%, 10%, 25%, 35%, 50%, 75%, 90%, 95% or 99%). Typically, these fragments are soluble fragments.
  • One of the exemplary fragments is the TGF ⁇ RII ectodomain having the sequence of SEQ ID NO:6.
  • substantially identical generally means that the polypeptide exhibits at least 50%, preferably 60%, 70%, 75% or 80%, more preferably 85%, 90% or 95%, and most preferably 99% of the reference amino acid sequence % amino acid sequence identity (identity).
  • the length of the comparison sequences is generally at least 10 amino acids, preferably at least 15 contiguous amino acids, at least 20, 25, 50, 75, 90, 100, 150, 200, 250, 300 or 350 contiguous amino acids, more preferably, full length amino acid sequence.
  • algorithms suitable for determining percent sequence identity and percent sequence similarity may be the BLAST and BLAST 2.0 algorithms, see Altschul et al. (1977) Nucleic Acids Res. 25:3389 and Altschul et al. (1990) J. Mol. Biol. 215, respectively : 403.
  • TGF ⁇ inhibitor generally refers to a substance that inhibits the production and signal transduction of TGF ⁇ signaling molecules, and the inhibitor can be a small molecule compound or a macromolecule compound (such as an antibody).
  • exemplary small molecule TGF ⁇ inhibitors include SB525334, SD-208, SB431542, LY2109761, LY2157299 (Galunisertib), GW788388, RepSox, SIS3, LDN-193189, EW-7197, and LY364947, among others.
  • antibody generally refers to an immunoglobulin reactive against a specified protein or peptide or fragment thereof.
  • Antibodies can be antibodies from any class, including but not limited to IgG, IgA, IgM, IgD, and IgE, and antibodies from any subclass (eg, IgGl, IgG2, IgG3, and IgG4).
  • the antibody may have a heavy chain constant region selected from, eg, IgGl, IgG2, IgG3, or IgG4.
  • the antibody may also have a light chain selected from, for example, kappa ( ⁇ ) or lambda ( ⁇ ).
  • the antibodies of the present application can be derived from any species.
  • an antigen binding domain generally refers to a portion of an antibody molecule comprising the amino acids responsible for specific binding between the antibody and an antigen.
  • the portion of an antigen that is specifically recognized and bound by an antibody is referred to as an "epitope" as described above.
  • an antigen binding domain may typically comprise an antibody light chain variable region (VL) and an antibody heavy chain variable region (VH); however, it need not comprise both.
  • Fd fragments for example, have two VH regions and generally retain some antigen-binding function of the intact antigen-binding domain.
  • antigen-binding fragments of antibodies include (1) Fab fragments, monovalent fragments having VL, VH, constant light chain (CL) and CH1 domains; (2) F(ab')2 fragments, Bivalent fragment of two Fab fragments linked by a sulfur bridge; (3) Fd fragment with two VH and CH1 domains; (4) Fv fragment with VL and VH domains of the antibody one-arm, (5) dAb fragment (Ward et al., "Binding Activities of a Repertoire of Single Immunoglobulin Variable DomainsSecreted From Escherichia coli," Nature 341:544-546 (1989), which is incorporated by reference in its entirety), which has a VH domain; ( 6) Isolated Complementarity Determining Regions (CDRs); (7) Single-chain Fvs (scFvs), eg from scFV-libraries.
  • Fab fragments monovalent fragments having VL, VH, constant light chain (CL) and CH
  • the two domains of the Fv fragment, VL and VH are encoded by separate genes, they can be joined using recombinant methods by a synthetic linker that allows it to be prepared as a single protein in which the VL and VH domains are paired to form a monovalent molecule chain (referred to as single-chain Fv (scFv))
  • scFv single-chain Fv
  • Huston et al. "Protein Engineering of AntibodyBinding Sites: Recovery of Specific Activity in an Anti-Digoxin Single-ChainFv Analogue Produced in Escherichia coli," Proc. Natl. Acad . Sci.
  • VHH relates to variable antigen binding domains of heavy chain antibodies from the family Camelidae (camelids, dromedaries, llamas, alpacas, etc.) (see Nguyen V.K. et al., 2000, The EMBO Journal, 19, 921-930; Muyldermans S., 2001, J Biotechnol., 74, 277-302 and review Vanlandschoot P. et al., 2011, Antiviral Research 92, 389-407) . VHHs may also be referred to as Nanobodies (Nb).
  • variable region or “variable domain” generally refers to the domain of an antibody heavy or light chain that is involved in the binding of an antibody to an antigen.
  • variable generally refers to certain portions of the sequence of the variable domains of antibodies that vary strongly, resulting in the binding and specificity of each particular antibody for its particular antigen. Variability is not evenly distributed throughout the variable region of an antibody. It is concentrated in three segments in the light and heavy chain variable regions, called complementarity determining regions (CDRs) or hypervariable regions (HVRs), LCDR1, LCDR2, LCDR3, HCDR1, HCDR2 and HCDR3. The more highly conserved portions of the variable domains are referred to as framework regions (FRs).
  • CDRs complementarity determining regions
  • HVRs hypervariable regions
  • FRs framework regions
  • variable domains of native heavy and light chains each comprise four FR regions (H-FR1, H-FR2, H-FR3, H-FR4, L-FR1, L-FR2, L-FR3, L-FR4) , mostly adopting a ⁇ -sheet configuration, connected by three loop regions of the CDR structure.
  • the CDRs in each chain are brought together in close proximity by the FR regions, and together with the CDRs from the other chain form the antigen-binding site of the antibody.
  • variable regions of an antibody or the CDRs of an antibody can be encoded by a variety of methods, such as the Kabat numbering scheme and definition rules based on sequence variability (see, Kabat et al., Protein Sequences in Immunology, 5.
  • peptide linker generally refers to the amino acid sequence by which the amino acid sequences of different domains in the PD-L1/TGF ⁇ dual inhibitor of the present application are connected to each other.
  • An essential technical feature of such a peptide linker is that the peptide linker does not contain any polymerization activity.
  • Preferred amino acid residues for peptide linkers include Gly, Ser and Thr, and are characterized by a length of 5 to 25 amino acid residues.
  • Suitable peptide linkers include those described in US Pat. Nos. 4,751,180 and 4,935,233 or WO88/09344.
  • a preferred embodiment of the peptide linker is characterized by the amino acid sequence Gly-Gly-Gly-Gly - Ser, ie Gly4Ser, or a polymer thereof, ie (Gly4Ser)x, where x is an integer 1 or greater.
  • peptide linkers including non-promoting secondary structure, are known in the art and described, for example, in Dall'Acqua et al. (Biochem. (1998) 37, 9266-9273), Cheadle et al. (Mol Immunol ( 1992) 29, 21-30) and Raag and Whitlow (FASEB (1995) 9(1), 73-80).
  • Peptide linkers that also do not contribute to any secondary structure are preferred.
  • the domains are provided in connection with each other, eg by genetic engineering, as described in the Examples.
  • Methods for preparing fusion and operably linked bispecific single chain constructs and expressing them in mammalian cells or bacteria are well known in the art (e.g. WO99/54440 or Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 2001).
  • the terms "antagonist” and “inhibitor” are used interchangeably and generally refer to compounds that are capable of reducing or inhibiting the biological function of a target protein or polypeptide, such as reducing or inhibiting the activity or expression of the target protein or polypeptide or Pharmacy. Inhibitors need not completely eliminate the biological function of the target protein or polypeptide, and in some embodiments, reduce activity by at least 50%, 60%, 70%, 80%, 90%, 95%, or 99%. While some of the antagonists of the present application specifically interact with (eg, bind to) the target, they inhibit the biological activity of the target protein or polypeptide by interacting with other members of the signal transduction pathway that includes the target protein or polypeptide The compounds of are also specifically included in this definition. Non-limiting examples of biological activities that are inhibited by antagonists include those activities associated with the development, growth or spread of tumors or undesired immune responses manifested in autoimmune diseases.
  • the term "effective amount” or “therapeutically effective amount” generally refers to an amount of a compound or pharmaceutical composition described herein sufficient to achieve the intended use described below, including but not limited to disease treatment .
  • a therapeutically effective amount may vary according to: the intended application (in vivo or in vitro); or the subject being treated and the disease condition, eg, the subject's weight and age, the severity of the disease condition; the mode of administration, etc., which can be determined by one of ordinary skill in the art Personnel are easily identified.
  • the term also applies to doses that will induce a specific response in target cells, such as platelet adhesion and/or cell migration.
  • the specific dosage will vary depending on, for example, the specific compound chosen, the dosing regimen followed, whether it is administered in combination with other agents, the time of administration, the tissue to which it is administered, and the physical delivery system by which it is delivered.
  • in vivo generally refers to an event that occurs in a subject.
  • an in vitro assay generally refers to events that occur outside the body of a subject.
  • an in vitro assay includes any assay performed outside the subject.
  • In vitro assays include cell-based assays in which live or dead cells are employed.
  • In vitro assays also include cell-free assays in which intact cells are not employed.
  • combination therapy or “combination” generally refers to the use of more than one compound or agent to treat a particular disorder or condition.
  • a PI3K inhibitor can be administered in combination with at least one additional therapeutic agent.
  • “Combination” is not intended to imply that the other therapy and the PI3K inhibitor must be administered and/or formulated for delivery together, but such delivery methods are within the scope of this application.
  • the PI3K inhibitor can be administered concurrently with one or more additional agents prior to (eg, 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, 12 weeks, or 16 weeks) or after (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks , 6 weeks, 8 weeks, 12 weeks, or 16 weeks).
  • additional agents eg., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks , 6 weeks, 8 weeks, 12 weeks, or 16 weeks.
  • each therapeutic agent will be administered at a dose and/or on a schedule determined for that particular agent.
  • the other therapeutic agents can be administered in a single composition or in separate compositions with the PI3K inhibitors provided herein.
  • the present application also contemplates higher combinations, such as triple therapy of PI3K inhibitor + PD-L1 inhibitor + TGF ⁇ inhibitor.
  • administering generally refers to introducing the drug combination into a subject's body by any route of introduction or delivery. Any method known to those of skill in the art for contacting cells, organs or tissues with the drug combination can be employed. Including, but not limited to, intraarterial, intranasal, intraperitoneal, intravenous, intramuscular, subcutaneous transdermal or oral.
  • the daily dose may be divided into one, two or more doses in suitable form to be administered at one, two or more times during a certain period of time.
  • the term "administering” includes administering to a subject two or more agents such that the agents and/or their metabolites are present in the subject at the same or substantially the same time.
  • co-administration of a PI3K inhibitor with an additional anti-cancer agent refers to any administration of the two active agents, separate or together, Two of the active agents are administered as part of an appropriate dosage regimen aimed at obtaining the benefits of the combination therapy.
  • the two active agents can be administered as part of the same pharmaceutical composition or in separate pharmaceutical compositions.
  • the additional agent may be administered before, concurrently with, or after administration of the PI3K inhibitor, or some combination thereof.
  • the additional agent may be administered before, concurrently with, or after each administration of the PI3K inhibitor, or some combination thereof, or at a relative level relative to the PI3K inhibitor
  • the agents are administered at different intervals for treatment, or administered in a single dose prior to a course of treatment with a PI3K inhibitor, at any time during a course of treatment with a PI3K inhibitor, or after a course of treatment with a PI3K inhibitor.
  • the first agent may be administered prior to administration of the second therapeutic agent (eg, 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours) , 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks ago), substantially simultaneously or thereafter (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks , 8 weeks or 12 weeks later).
  • the second therapeutic agent eg., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks , 8 weeks or 12 weeks later.
  • monotherapy refers to the use of an agent alone (also referred to herein as alone) (eg, as a single compound or agent), eg, to treat the same indication without a second active ingredient disease, such as cancer.
  • monotherapy includes the use of a PI3K inhibitor or a second agent alone to treat cancer.
  • synergy or synergy refers to the beneficial effect of combining two or more agents, eg, in a pharmaceutical composition or in a method of treatment.
  • one or more beneficial effects are achieved through the use of a PI3K inhibitor in combination with a second therapeutic agent described herein (eg, one or more second therapeutic agents).
  • a synergistic effect refers to a lower dose of one or both agents required to achieve the effect.
  • a combination may provide a selected effect, eg, a therapeutic effect, when at least one agent is administered at a lower dose than that required to achieve the same therapeutic effect when the agent is administered as a monotherapy.
  • the combination of a PI3K inhibitor (eg, a PI3K inhibitor) and a second agent (as described herein) allows the PI3K inhibitor to achieve the same therapeutic effect as when the PI3K inhibitor is administered as monotherapy administered at lower doses as required.
  • the synergistic effect reduces, prevents, delays or reduces the occurrence or likelihood of occurrence of one or more side effects, toxicity, resistance otherwise associated with administration of at least one of the agents.
  • the synergistic effect reduces resistance to at least one of the agents (eg, a measure of resistance is reduced or the likelihood of developing resistance is reduced) or delays the development of resistance to at least one of the agents produce.
  • the synergistic effect is a reduction in minimal residual disease (MRD).
  • the combination of a PI3K inhibitor (eg, a PI3K inhibitor described herein) and a second agent (eg, a second agent described herein) is effective to reduce MRD in a subject, eg, low at levels previously measured in the subject (eg, levels measured prior to administration of the combination).
  • the combination of a PI3K inhibitor and a second agent is effective to reduce MRD in a subject below levels observed during or after treatment with a monotherapy, eg, comprising a PI3K inhibitor or a second agent Monotherapy of any of the medicaments.
  • the MRD is reduced below the level observed during treatment with a monotherapy comprising a PI3K inhibitor. In certain embodiments, the MRD is reduced below the level observed during treatment with a monotherapy comprising the second agent. In certain embodiments, the combination is effective to reduce the level of MRD to a preselected cutoff value (eg, 1 malignant cell in 100 normal cells, 1 malignant cell in 1000 normal cells, or 10,000 normal cells 1 malignant cell in cells, or 1 malignant cell in 100,000 normal cells) or less. In certain embodiments, the preselected cutoff value is 1 malignant cell in 1000 normal cells. In certain embodiments, the preselected cutoff value is 1 malignant cell in 100,000 normal cells.
  • a preselected cutoff value eg, 1 malignant cell in 100 normal cells, 1 malignant cell in 1000 normal cells, or 10,000 normal cells 1 malignant cell in cells, or 1 malignant cell in 100,000 normal cells
  • synergy refers to a PI3K inhibitor (eg, a PI3K inhibitor or a pharmaceutically acceptable form thereof) and a second therapeutic agent (eg, one or more additional therapeutic agents or a pharmaceutically acceptable form thereof)
  • a PI3K inhibitor eg, a PI3K inhibitor or a pharmaceutically acceptable form thereof
  • a second therapeutic agent eg, one or more additional therapeutic agents or a pharmaceutically acceptable form thereof
  • pharmaceutically acceptable generally refers to one or more nontoxic substances that do not interfere with the effectiveness of the biological activity of the active ingredient.
  • Such formulations may generally contain salts, buffers, preservatives, compatible carriers and optionally other therapeutic agents.
  • “pharmaceutically acceptable forms” of compounds include, but are not limited to, pharmaceutically acceptable salts, hydrates, solvates, isomers, prodrugs, and isotopically labeled derivatives of the disclosed compounds.
  • “pharmaceutically acceptable forms” include, but are not limited to, pharmaceutically acceptable salts, isomers, prodrugs, and isotopically labeled derivatives of the disclosed compounds.
  • the term "pharmaceutically acceptable salt” generally refers to a salt suitable for use in contact with a subject's tissue without undue toxicity, irritation, allergic reaction, etc., within the scope of sound medical judgment, and with reasonable benefit / hazard ratios that match those salts.
  • Pharmaceutically acceptable salts are well known in the art. For example, Berge et al. describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences (1977) 66:1-19.
  • Pharmaceutically acceptable salts of the compounds provided herein include salts derived from suitable inorganic and organic acids and bases.
  • non-toxic acid addition salts are with inorganic acids such as hydrochloric, hydrobromic, phosphoric, sulfuric and perchloric or organic acids such as acetic, oxalic, maleic, tartaric , citric acid, succinic acid or malonic acid), or by using other methods used in the art such as ion exchange.
  • inorganic acids such as hydrochloric, hydrobromic, phosphoric, sulfuric and perchloric or organic acids
  • acetic, oxalic, maleic, tartaric , citric acid, succinic acid or malonic acid such as ion exchange.
  • salts include adipate, alginate, ascorbate, aspartate, besylate, besylate, benzoate, bisulfate, boric acid Salt, butyrate, camphorate, camphorsulfonate, citrate, cyclopentane propionate, digluconate, lauryl sulfate, ethanesulfonate, formate, fumarate enedioate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodate, 2-hydroxy-ethanesulfonate, lactobionate , lactate, laurate, lauryl sulfate, malate, maleate, malonate, mesylate, 2-naphthalenesulfonate, nicotinate, nitrate, oil acid salt, oxalate, palmitate, pamoate, pectate,
  • organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid , citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, etc.
  • Pharmaceutically acceptable salts derived from suitable bases include alkali metal, alkaline earth metal, ammonium and N + (C 1-4 alkyl) 4 salts.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like.
  • Additional pharmaceutically acceptable salts include, where appropriate, non-toxic ammonium, quaternary ammonium and amine cations formed using counter ions such as halides, hydroxides, carboxylates, sulfates, phosphates, nitric acid root, lower alkyl sulfonate and aryl sulfonate.
  • Organic bases from which salts can be derived include, for example, primary, secondary and tertiary amines, substituted amines (including naturally occurring substituted amines), cyclic amines, basic ion exchange resins, and the like, such as isopropylamine, Trimethylamine, diethylamine, triethylamine, tripropylamine and ethanolamine.
  • the pharmaceutically acceptable base addition salt is selected from the group consisting of ammonium, potassium, sodium, calcium and magnesium salts.
  • solvate generally refers to a compound that also includes a stoichiometric or non-stoichiometric amount of a solvent bound by non-covalent intermolecular forces.
  • a solvate can be a disclosed compound or a pharmaceutically acceptable salt thereof.
  • the solvent is water
  • the solvate is a "hydrate”.
  • Pharmaceutically acceptable solvents and hydrates are, for example, complexes that may include 1 to about 100, or 1 to about 10, or 1 to about 2, about 3, or about 4 molecules of solvent or water . It is to be understood that the term “compound” in this application includes compounds and solvates of compounds, as well as mixtures thereof.
  • prodrug generally refers to a compound that is transformed in vivo to yield the disclosed compound or a pharmaceutically acceptable form of the compound.
  • a prodrug may be inactive when administered to a subject, but is converted to an active compound in vivo, eg, by hydrolysis (eg, in blood).
  • prodrugs have improved physical and/or delivery properties over the parent compound.
  • Prodrugs are typically designed to enhance pharmacological and/or pharmacokinetic-based properties relative to the parent compound.
  • Prodrug compounds often offer the advantages of solubility, histocompatibility, or delayed release in mammalian organisms (see, eg, Bundgard, H., Design of Prodrugs (1985), pp.
  • prodrugs may include, but are not limited to, their physical properties, such as increased water solubility over the parent compound for parenteral administration at physiological pH, or they may enhance absorption from the digestive tract, or they may enhance long-term storage of the drug stability.
  • prodrug is also intended to include any covalently bonded carrier that releases the active compound in vivo when the prodrug is administered to a subject.
  • Prodrugs of active compounds as described herein can be prepared by modifying groups present in the active compound in a manner that cleaves into the parent active compound in routine practice or in vivo.
  • Prodrugs include compounds in which the hydroxyl, amino, or sulfhydryl groups are bonded to any group that, when a prodrug of the active compound is administered to a subject, cleaved to form a free hydroxyl, free amino, or free sulfhydryl group, respectively .
  • prodrugs examples include, but are not limited to, acetate, formate, and benzoate derivatives of alcohols in the active compound or acetamide, formamide, and benzamide derivatives of amine functional groups, and the like.
  • Other examples of prodrugs include compounds comprising -NO, -NO2, -ONO or -ONO2 moieties.
  • Prodrugs can generally be prepared using well-known methods, as in Burger's Medicinal Chemistry and Drug Discovery, 172-178, 949-982 (Manfred E. Wolff ed., 5th ed., 1995) and Design of Prodrugs (H. Bundgaard ed., Elsevier, Those described in New York, 1985).
  • the prodrug may comprise a pharmaceutically acceptable ester formed by replacing the hydrogen atom of the acid group with a group that Groups such as (C1-C8) alkyl, (C2-C12) acyloxymethyl, 1-(acyloxy)ethyl having 4 to 9 carbon atoms, 1-methyl having 5 to 10 carbon atoms yl-1-(acyloxy)-ethyl, alkoxycarbonyloxymethyl having 3 to 6 carbon atoms, 1-(alkoxycarbonyloxy)ethyl having 4 to 7 carbon atoms , 1-methyl-1-(alkoxycarbonyloxy)ethyl having 5 to 8 carbon atoms, N-(alkoxycarbonyl)aminomethyl having 3 to 9 carbon atoms, 4 to 1-(N-(alkoxycarbonyl)amino)ethyl, 3-phthaloyl, 4-crotonyllactone
  • a prodrug can be formed by replacing the hydrogen atom of the alcohol group with a group such as (C1-C6) Acyloxymethyl, 1-((C1-C6)acyloxy)ethyl, 1-methyl-1-((C1-C6)acyloxy)ethyl, (C1-C6)alkoxycarbonyl Oxymethyl, N-(C1-C6)alkoxycarbonylaminomethyl, succinyl, (C1-C6)acyl, ⁇ -amino(C1-C4)acyl, arylacyl and ⁇ -aminoacyl or ⁇ -aminoacyl- ⁇ -aminoacyl, wherein each ⁇ -aminoacyl is independently selected from naturally occurring L-amino acids, P(O)(OH)2, -P(O)(O(C1-C6)alkanes group) 2 and a sugar group (a group generated by
  • the prodrug can be formed by replacing the hydrogen atom in the amine group with a group such as R-carbonyl, RO- Carbonyl, NRR'-carbonyl, where R and R' are each independently (C1-C10)alkyl, (C3-C7)cycloalkyl, benzyl, natural alpha-aminoacyl or natural alpha-aminoacyl-natural alpha -aminoacyl; -C(OH)C(O)OY1, where Y1 is H, (C1-C6)alkyl or benzyl; -C(OY2)Y3, where Y2 is (C1-C4)alkyl and Y3 is (C1-C6)alkyl, carboxy(C1-C6)alkyl, amino(C1-C4)alkyl or mono-N- or di-N,N-(C1-C6)alkylaminoalkyl;- C
  • isomers generally refer to different compounds having the same molecular formula.
  • Stepoisomer generally refers to isomers that differ only in the arrangement of the atoms in space.
  • the term “isomer” includes any and all geometric and stereoisomers.
  • isomers include geometric double bond cis and trans isomers, also known as E- and Z- isomers; R- and S- enantiomers; diastereomers, (d)-isomers and (l)-isomers, racemic mixtures thereof; and other mixtures thereof falling within the scope of the present disclosure.
  • enantiomers generally refer to a pair of stereoisomers that are non-superimposable mirror images of each other.
  • a 1:1 mixture of a pair of enantiomers is a “racemic” mixture.
  • the term “( ⁇ )” is used to denote a racemic mixture.
  • “Diastereomers” are stereoisomers that have at least two asymmetric atoms, but are not mirror images of each other. Absolute stereochemistry is specified according to the Cahn-Ingold-Prelog R-S system. When the compounds are pure enantiomers, the stereochemistry at each chiral carbon can be designated by R or S.
  • Resolved compounds of unknown absolute configuration can be assigned (+) or (-) depending on the direction (right or left) that they rotate plane polarized light at the wavelength of the sodium D line.
  • Certain compounds described in this application contain one or more asymmetric centers and thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that can be defined in terms of absolute stereochemistry, such as (R)- or (S)-.
  • the chemical entities, pharmaceutical compositions and methods of the present application are intended to include all such possible isomers, including racemic mixtures, optically pure forms and intermediate mixtures.
  • Optically active (R)- and (S)-isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless otherwise specified, the compounds are intended to include both E and Z geometric isomers.
  • enantiomeric purity generally refers to the relative amount of the presence of a particular enantiomer relative to another enantiomer, expressed as a percentage. For example, if a compound that may have an (R)- or (S)-isomeric configuration exists as a racemic mixture, then with respect to either the (R)- or (S)-isomer, the The enantiomeric purity was about 50%. If one conformational form of the compound is superior to the other, for example, 80% (S)- and 20% (R)-, the compound has an enantiopurity of 80 for the (S)-isomeric form %.
  • the enantiomeric purity of a compound can be determined by a variety of means known in the art, including but not limited to chromatography using chiral supports, polarization measurement of polarized light rotation, using chiral shifting reagents (including but not limited to containing NMR spectroscopy of chiral complexes of lanthanides or Pirkle alcohols), or derivatization of compounds using chiral compounds such as Mosher acids followed by chromatography or NMR spectroscopy.
  • tautomer generally refers to a type of isomer that includes migration from at least one form of hydrogen atom and at least one change in valence (eg, a single bond to a double bond). , triple bond to double bond or triple bond to single bond, or vice versa) resulting in two or more interconvertible compounds.
  • Teautomerism includes protonation or protonation tautomerism, which is considered a subset of acid-base chemistry.
  • Proton shift tautomerism or “proton shift tautomerism” involves the migration of protons with a concomitant change in bond order. The exact ratio of tautomers depends on a variety of factors, including temperature, solvent and pH.
  • tautomers Chemical equilibrium of tautomers can be achieved when tautomerization is possible (eg, in solution).
  • Tautomerism ie, reactions that provide tautomeric pairs
  • Exemplary tautomers include, but are not limited to, keto-enol; amide-imide; lactam-lactam; enamine-imine; and enamine-(different) enamine tautomerism structure.
  • keto-enol tautomerism is the interconversion of pentane-2,4-dione and 4-hydroxypent-3-en-2-one tautomers.
  • tautomerism is phenol-ketone tautomerism.
  • a specific example of phenol-ketone tautomerism is the interconversion of pyridin-4-ol and pyridin-4(1H)-one tautomers.
  • structures depicted herein are also intended to include compounds that differ only by the presence of one or more isotopically enriched atoms. For example, except that hydrogen is replaced or enriched with deuterium or tritium at one or more atoms in the molecule, or carbon is replaced or enriched with13C or14C at one or more atoms in the molecule or 14 C, compounds having the structures of the present application are within the scope of this disclosure.
  • the compounds of this application also include isotopically-labeled compounds, which are the same as those described in this application, except that one or more atoms are separated by an atomic mass or mass number that is different from the atomic mass or mass number usually found in nature different atoms instead.
  • isotopes that may be added to the disclosed compounds include isotopes of hydrogen , carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, and chlorine, such as 2H, 3H , 13C , 14C , 15N , 18O , 17O , 31P , 32P , 35S , 18F and 36Cl .
  • isotopically-labeled disclosed compounds are useful in compound and/or matrix tissue distribution analysis.
  • Tritium (ie, 3 H) and carbon-14 (ie, 14 C) isotopes can allow for easy and detectable preparation.
  • substitution with heavier isotopes such as deuterium (ie, 2 H) may provide certain therapeutic advantages (eg, increased in vivo half-life or reduced dosage requirements) due to greater metabolic stability.
  • Isotopically labeled disclosed compounds can generally be prepared by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
  • the application provides compounds that also contain unnatural proportions of atomic isotopes at one or more of the atoms that make up the compound. All isotopic variations disclosed in this application, whether radioactive or not, are included within the scope of this disclosure.
  • pharmaceutically acceptable carrier or “pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic agents and absorption delaying agents agent, etc.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic agents and absorption delaying agents agent, etc.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Unless any conventional medium or agent is incompatible with the active ingredient, its use in the experimental compositions disclosed herein is contemplated. Supplementary active ingredients can also be incorporated into the pharmaceutical compositions.
  • tumor generally refers to any neoplastic cell growth and proliferation, malignant or benign, as well as any precancerous and cancerous cells and tissues.
  • neoplastic refers to any form of abnormally regulated or unregulated cell growth, malignant or benign, resulting in abnormal tissue growth.
  • neoplastic cells include malignant and benign cells with abnormally regulated or unregulated cell growth.
  • cancer cancer, “cancerous”, “cell proliferative disorder”, “proliferative disorder” and “tumor” are not mutually exclusive when referred to in this application.
  • a tumor can refer to a physical mass containing a majority of cancer cells, eg, cells that display any of the cancers described herein.
  • a tumor can be a solid tumor or a non-solid tumor (eg, hematological tumor, lymphoma).
  • the term "solid tumor” generally refers to an abnormal tissue growth or mass that usually does not contain cysts or fluid areas. Solid tumors can be benign (noncancerous) or malignant (cancerous).
  • the tumor may be a tumor with abnormal expression of PD-1 or PD-L1 in cells and tissues.
  • the tumor may be a tumor with abnormal expression or activity of TGF ⁇ or TGF ⁇ R in cells and tissues.
  • the tumor can be a tumor with abnormal expression or activity of PI3K in cells and tissues.
  • the term “resistance” generally refers to when the cancer has a reduced response to treatment, eg, to the extent that the cancer does not respond to treatment.
  • the cancer may be resistant at the start of treatment or it may develop resistance during treatment.
  • a subject with cancer may have one or more mutations that render it resistant to treatment, or the subject may develop such mutations during treatment.
  • the term “refractory” can refer to cancers that have proven ineffective with treatment (eg, chemotherapeutic drugs, biologics, and/or radiation therapy). Refractory cancer tumors can shrink, but not to the point where treatment is known to be effective. Often, however, tumors remain the same size (stable disease), or grow (progressive disease) as they were before treatment.
  • treatment and “treating” generally refer to methods of obtaining beneficial or desired results, including but not limited to therapeutic benefit.
  • Therapeutic benefit includes, but is not limited to, eradication, inhibition, reduction or amelioration of the underlying disorder being treated. Additionally, therapeutic benefit is achieved by eradicating inhibition, reduction or amelioration of one or more physiological symptoms associated with the underlying disorder, whereby improvement is observed in the patient, but the patient may still have the underlying disorder.
  • prevention and preventing generally refer to methods of obtaining beneficial or desired results, including but not limited to preventive benefits.
  • a pharmaceutical composition can be administered to a patient at risk of developing a particular disease or to a patient reporting one or more physical symptoms of a disease, even if the disease has not been diagnosed.
  • the term "subject” or “patient” generally refers to a human (ie, male or female of any age group, eg, a pediatric subject (eg, infant, child, adolescent) or an adult subject (eg, young human, middle-aged or elderly)) and/or other primates (eg, cynomolgus monkeys, rhesus monkeys); mammals, including commercially related mammals such as cattle, pigs, horses, sheep, goats, cats and/or dogs; and/or birds, including commercially related birds such as chickens, ducks, geese, quail and/or turkeys.
  • a human ie, male or female of any age group, eg, a pediatric subject (eg, infant, child, adolescent) or an adult subject (eg, young human, middle-aged or elderly)) and/or other primates (eg, cynomolgus monkeys, rhesus monkeys); mammals, including commercially related mammals such
  • the term “about” or “approximately” generally refers to an acceptable error in a particular value determined by one of ordinary skill in the art, which depends in part on how the value is measured or determined. In certain embodiments, the term “about” or “approximately” generally refers to 1, 2, 3, or 4 standard deviations. In certain embodiments, the term “about” or “approximately” generally means 50%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5% of a given value or range , 4%, 3%, 2%, 1%, 0.5% or 0.05%.
  • the term “comprising” or “comprising” generally refers to an open-ended form, and it should be understood that it may also contain other substances not mentioned.
  • a drug combination comprising a phosphoinositide 3-kinase (PI3K) inhibitor and a second therapeutic agent, wherein the second therapeutic agent is an inhibitor of an immune checkpoint, a TGF ⁇ inhibitor, a dual function immune checkpoint/ TGF ⁇ inhibitors or combinations thereof.
  • PI3K phosphoinositide 3-kinase
  • “comprising” should be understood as containing other substances in addition to the phosphoinositide 3-kinase (PI3K) inhibitor and the second therapeutic agent.
  • the application provides a pharmaceutical combination comprising a phosphoinositide 3-kinase (PI3K) inhibitor and a second therapeutic agent, wherein the second therapeutic agent is an immune checkpoint inhibitor, a TGF ⁇ inhibitor, a dual function Immune checkpoint/TGF ⁇ inhibitors or combinations thereof.
  • PI3K phosphoinositide 3-kinase
  • the inhibitor of the immune checkpoint may comprise an agent capable of blocking the interaction of programmed death 1 (PD-1) with programmed death ligand 1 (PD-L1).
  • PD-1 programmed death 1
  • PD-L1 programmed death ligand 1
  • the inhibitor of the immune checkpoint may comprise an inhibitor of programmed death ligand 1 (PD-L1) and/or programmed death 1 (PD-1).
  • an inhibitor of human PD-1 such as an inhibitor of human PD-L1.
  • the inhibitor of the immune checkpoint may comprise an anti-PD-L1 antibody or an antigen-binding fragment thereof, or an anti-PD-1 antibody or an antigen-binding fragment thereof.
  • the inhibitor of PD-1 or PD-L1 can be an antibody molecule against PD-1 or PD-L1.
  • PD-1 or PD-L1 inhibitors can be administered alone or in combination with other immune checkpoint modulators, eg, in combination with inhibitors of LAG-3, TIM-3, or CTLA-4.
  • the pharmaceutical combination may include:
  • the inhibitor of the immune checkpoint can comprise nucleic acid (eg, dsRNA, siRNA or shRNA), polypeptide (eg, soluble ligand, antibody or antigen-binding fragment thereof, immunoadhesin, fusion proteins, oligopeptides and other molecules), or compounds.
  • nucleic acid eg, dsRNA, siRNA or shRNA
  • polypeptide eg, soluble ligand, antibody or antigen-binding fragment thereof, immunoadhesin, fusion proteins, oligopeptides and other molecules
  • the antibody can be selected from the group consisting of human antibodies, humanized antibodies, chimeric antibodies, multispecific antibodies, monoclonal antibodies and polyclonal antibodies.
  • the antigen-binding fragment can be selected from the group consisting of: Fab, Fab', F(ab') 2 , Fv, scFv, diabody, Fd, dAb, VHH, large antibody and complementarity determining region (CDR) fragments.
  • the antibody or antigen-binding fragment thereof may comprise a heavy chain variable region (VH) comprising HCDR1, HCDR2 and HCDR3.
  • VH heavy chain variable region
  • the antibody or antigen-binding fragment thereof may or may not also comprise a CH1 domain.
  • the antibody or antigen-binding fragment thereof may or may not also comprise CH2 and CH3 domains.
  • the antibody or antigen-binding fragment thereof comprises the domain of VH-CH2-CH3.
  • the antibody or antigen-binding fragment thereof may further comprise an antibody heavy chain constant region.
  • the antibody heavy chain constant region may be a human antibody heavy chain constant region.
  • the human antibody heavy chain constant region may be selected from constant regions derived from the group of IgGl, IgG2, IgG3, IgG4 and variants thereof.
  • the antibody or antigen-binding fragment thereof may further comprise a light chain variable region comprising LCDR1, LCDR2, LCDR3.
  • the antibody or antigen-binding fragment thereof may comprise a heavy chain variable region (VH), which may contain HCDRl, HCDR2, and HCDR3, and a light chain variable region, which may contain LCDRl, LCDR2, LCDR3.
  • VH heavy chain variable region
  • LCDRl LCDR2, LCDR3.
  • the antibody or antigen-binding fragment thereof may further comprise a light chain constant region (CL).
  • CL light chain constant region
  • the antibody or antigen-binding fragment thereof may comprise a heavy chain comprising VH and CH1, and an antibody light chain comprising VL and CL.
  • the antibody or antigen-binding fragment thereof may comprise a heavy chain comprising VH, CH1, CH2 and CH3, and an antibody light chain comprising VL and CL.
  • the inhibitor of the immune checkpoint can be a PD-1/PD-L1 inhibitor.
  • PD-1/PD-L1 inhibitors include, but are not limited to, US 7,488,802, US 7,943,743, US 8,008,449, US 8,168,757, US 8,217,149, US 8,609,089, US 2010/028330, US 2012/0114649, WO 2003/042 /156712, WO 2010/089411, WO 2010/036959, WO 2011/066342, WO 2011/159877, WO 2011/082400 and WO 2011/161699, all of which are incorporated herein by reference in their entirety.
  • the checkpoint modulator can be a PD-1 inhibitor.
  • the checkpoint modulator can be an anti-PD-1 antibody.
  • the anti-PD-1 antibody can be nivolumab.
  • Alternative names for nivolumab include MDX-1106, MDX-1106-04, ONO-4538, or BMS-936558, and the CAS Registry Number is: 946414-94-4.
  • Nivolumab is a full-length human IgG4 monoclonal antibody that specifically blocks PD-1.
  • Nivolumab (clone 5C4) and other human monoclonal antibodies that specifically bind to PD-1 are disclosed in US 8,008,449 and WO 2006/121168.
  • the anti-PD-1 antibody can be Pembrolizumab.
  • Pembrolizumab (trade name KEYTRUDA, formerly known as Lambrolizumab, also known as Merck 3745, MK-3475 or SCH-900475) is a humanized IgG4 monoclonal antibody that binds to PD-1.
  • Pembrolizumab is disclosed, for example, in Hamid, O. et al. (2013) New England Journal of Medicine 369(2):134-44, WO 2009/114335, and US 8,354,509.
  • the anti-PD-1 antibody can be pidilizumab.
  • Pidilizumab (CT-011; CureTech) is a humanized IgG1k monoclonal antibody that binds to PD-1.
  • Pidilizumab and other humanized anti-PD-1 monoclonal antibodies are disclosed in WO 2009/101611.
  • Other anti-PD1 antibodies are disclosed in US 8,609,089, US 2010/028330 and/or US 2012/0114649.
  • the anti-PD-1 antibody can be AMP-514 (Amplimmune).
  • the PD-1 inhibitor can be an immunoadhesin (eg, comprising an extracellular or PD-1 binding portion of PD-L1 or PD-L2 fused to a constant region (eg, the Fc region of an immunoglobulin sequence) immunoadhesins).
  • an immunoadhesin eg, comprising an extracellular or PD-1 binding portion of PD-L1 or PD-L2 fused to a constant region (eg, the Fc region of an immunoglobulin sequence) immunoadhesins).
  • the checkpoint modulator is a PD-L1 inhibitor.
  • the checkpoint modulator can be an anti-PD-L1 antibody or antigen-binding fragment thereof.
  • PD-L1 antibody or antigen-binding fragment thereof can include any anti-PD-L1 antibody or antigen-binding fragment thereof described in the art.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain variable region of an antibody
  • the heavy chain variable region comprises HCDR1, HCDR2, HCDR3, each of which is associated with the following molecules, respectively HCDR1, HCDR2, HCDR3 have at least about 80% (eg, about 85%, 90% or 95%) sequence identity to: Atezolizumab, Avelumab, BMS-936559, MPDL3280A (RG7446) or durvalumab (MEDI4736).
  • the heavy chain variable regions have at least about 80% (eg, about 85%, 90% or 95%) sequence identity to the heavy chain variable regions of the following molecules, respectively: Atezolizumab, Avelumab, BMS-936559, MPDL3280A, or durvalumab.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain of an antibody, wherein the heavy chain is at least about 80% (e.g., about 85%, e.g., about 85%, respectively,) 90% or 95%) sequence identity: Atezolizumab, Avelumab, BMS-936559, MPDL3280A, or durvalumab.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof further comprises a light chain variable region of an antibody, wherein the light chain variable region comprises LCDR1, LCDR2, LCDR3, each of which is associated with the following Molecules LCDR1, LCDR2, LCDR3 have at least about 80% (eg, about 85%, 90% or 95%) sequence identity to: Atezolizumab, Avelumab, BMS-936559, MPDL3280A or durvalumab.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain variable region and a light chain variable region of an antibody, wherein the heavy chain variable region comprises HCDR1, HCDR2, HCDR3, wherein the light chain can
  • the variable region comprises LCDR1, LCDR2, LCDR3, each of which has at least about 80% (eg, about 85%, 90% or 95%) sequence identity to HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, LCDR3, respectively, of the following molecules: Atezolizumab, Avelumab, BMS-936559, MPDL3280A (RG7446) or durvalumab (MEDI4736).
  • the light chain variable regions have at least about 80% (eg, about 85%, 90% or 95%) sequence identity to the light chain variable regions of atezolizumab, Avelumab, BMS-936559, MPDL3280A, or durvalumab.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain variable region and a light chain variable region of an antibody, each of which has at least about 80% (eg, about 85%, 90% or 95%) sequence identity: Atezolizumab, Avelumab, BMS-936559, MPDL3280A (RG7446) or durvalumab (MEDI4736).
  • the anti-PD-L1 antibody or antigen-binding fragment thereof further comprises a light chain of an antibody, wherein the light chain is at least about 80% (eg, about 85%, respectively) the light chain of the following molecule , 90% or 95%) sequence identity: Atezolizumab, Avelumab, BMS-936559, MPDL3280A, or durvalumab.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises heavy and light chains of an antibody that are each at least about 80% (eg, about 85%, 90% or 95%) sequence identity to: Atezolizumab, Avelumab, BMS-936559, MPDL3280A (RG7446) or durvalumab (MEDI4736).
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises: Atezolizumab, Avelumab, BMS-936559, MPDL3280A, or durvalumab, or an antigen-binding fragment thereof, or a variant or biosimilar thereof, or its combination.
  • the anti-PD-L1 antibody may also be a commercially available or published PD-L1 antibody in the literature. Including but not limited to, such as PD-L1 antibody BMS-936559, MPDL3280A, MEDI4736, MSB0010718C (see US2014341917, US20130034559, US8779108) and the like.
  • an anti-PD-L1 antibody is MDX-1105.
  • MDX-1105 also known as BMS-936559, is an anti-PD-L1 antibody as described in WO 2007/005874.
  • the anti-PD-L1 antibody is YW243.55.S70.
  • the YW243.55.S70 antibody is an anti-PD-L1 antibody as described in WO 2010/077634.
  • the heavy and light chain variable region sequences of YW243.55.S70 are also described in WO2010/077634.
  • the anti-PD-L1 antibody is MDPL3280A (Genentech/Roche).
  • MDPL3280A is a human Fc-optimized IgG1 monoclonal antibody that binds to PD-L1.
  • Human monoclonal antibodies to MDPL3280A and other PD-L1 are described in US 7,943,743 and US 2012/0039906.
  • the anti-PD-L1 antibody is Avelumab (MSB0010718C).
  • MSB0010718C also known as A09-246-2; Merck Serono
  • Other humanized anti-PD-L1 antibodies are disclosed in WO 2013/079174.
  • the anti-PD-L1 antibody is durvalumab (CAS Number: 1428935-60-7).
  • the anti-PD-L1 antibody is Atezolizumab (CAS Number: 1380723-44-3).
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain variable region of an antibody
  • the heavy chain variable region comprises one or more heavy chain selectable from the group consisting of Variable region CDRs (HCDRs): (a) HCDR1 having at least about 70% sequence identity to the amino acid sequence set forth in SEQ ID NO:1; (b) HCDR2 having at least about 70% sequence identity to SEQ ID NO:2 and (c) a HCDR3 having at least about 70% sequence identity to SEQ ID NO:3.
  • HCDRs Variable region CDRs
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain variable region of an antibody
  • the heavy chain variable region comprises one or more HCDRs selected from the group consisting of: ( a) HCDR1 with the amino acid sequence shown in SEQ ID NO: 1 or HCDR1 obtained by amino acid addition, elimination or substitution reaction with no more than 2 amino acid differences from the amino acid sequence shown in SEQ ID NO: 1; (b) HCDR2 having the amino acid sequence shown in SEQ ID NO: 2 or HCDR2 obtained by amino acid addition, elimination or substitution reaction and having no more than 2 amino acid differences with the amino acid sequence shown in SEQ ID NO: 2; and (c) having The HCDR3 of the amino acid sequence shown in SEQ ID NO: 3 or the HCDR3 obtained by amino acid addition, elimination or substitution reaction with the amino acid sequence shown in SEQ ID NO: 3 differs by no more than 2 amino acids.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain variable region of an antibody
  • the heavy chain variable region comprises HCDR1, HCDR2, HCDR3 comprising the
  • the amino acid sequence set forth in ID NO: 1 is at least about 70% identical in sequence
  • the HCDR2 comprises an amino acid sequence at least about 70% identical to the amino acid sequence set forth in SEQ ID NO: 2
  • the HCDR3 comprises an amino acid sequence at least about 70% identical to the amino acid sequence set forth in SEQ ID NO: 2
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain variable region of an antibody
  • the heavy chain variable region comprises HCDR1, HCDR2, HCDR3, and the HCDR1 comprises SEQ ID
  • the HCDR2 comprises SEQ ID NO: The amino acid sequence shown in: 2 or the amino acid sequence obtained by amino acid addition, elimination or substitution reaction with the amino acid sequence shown in SEQ ID NO: 2 with no more than 2 amino acid differences
  • Described HCDR3 comprises SEQ ID NO: The amino acid sequence shown in 3 or the amino acid sequence obtained by amino acid addition, elimination or substitution reaction has no more than 2 amino acid differences with the amino acid sequence shown in SEQ ID NO: 3.
  • the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain variable region of an antibody
  • the heavy chain variable region comprises an amino acid sequence selected from the group consisting of: (a) SEQ The amino acid sequence set forth in ID NO: 4; (b) an amino acid sequence at least about 85%, 90%, 95% or 99% identical to the amino acid sequence set forth in SEQ ID NO: 4; and (c) by adding The amino acid sequence obtained by the formation, elimination or substitution reaction with the amino acid sequence shown in SEQ ID NO: 4 has 1 or more differences.
  • TGF ⁇ inhibitor binds TGF ⁇ 1, TGF ⁇ 2, or TGF ⁇ 3.
  • the TGF[beta] inhibitor binds TGF[beta] or transforming growth factor beta receptor (TGF[beta]R).
  • TGF[beta]R transforming growth factor beta receptor
  • the TGF[beta] inhibitor can bind to TGF[beta] type I receptor (TGF[beta]RI), TGF[beta] type II receptor (TGF[beta]RII) or TGF[beta] type III receptor (TGF[beta]RIII).
  • the TGF ⁇ inhibitor comprises: 1) an anti-TGF ⁇ antibody or antigen-binding fragment thereof; 2) an anti-TGF ⁇ R antibody or antigen-binding fragment thereof; 3) a small molecule TGF ⁇ inhibitor; 4) comprising transforming growth A protein of factor beta receptor II (TGF ⁇ RII) or a functionally active fragment thereof, or a variant or biosimilar thereof, or a combination thereof.
  • TGF ⁇ RII transforming growth A protein of factor beta receptor II
  • the anti-TGF ⁇ antibody or antigen-binding fragment thereof comprises a heavy chain variable region of an antibody, wherein the heavy chain variable region comprises HCDR1, HCDR2, HCDR3, each of which is associated with a Fresolimumab molecule (CAS), respectively.
  • HCDR1, HCDR2, HCDR3 of Number: 948564-73-6 have at least about 80% (eg, about 85%, 90% or 95%) sequence identity.
  • the heavy chain variable region can have at least about 80% (eg, about 85%, 90% or 95%) sequence identity to the heavy chain variable region of a Fresolimumab molecule.
  • the heavy chain can have at least about 80% (eg, about 85%, 90% or 95%) sequence identity to the heavy chain of a Fresolimumab molecule.
  • the anti-TGF ⁇ antibody or antigen-binding fragment thereof comprises a light chain variable region of an antibody
  • the light chain variable region comprises LCDR1, LCDR2, LCDR3, each of which is associated with LCDR1 of a Fresolimumab molecule, respectively
  • LCDR2, LCDR3 have at least about 80% (eg, 85%, 90% or 95%) sequence identity.
  • the anti-TGF ⁇ antibody or antigen-binding fragment thereof comprises a heavy chain variable region and a light chain variable region of an antibody, wherein the heavy chain variable region comprises HCDR1, HCDR2, HCDR3, wherein the light chain variable region Comprising LCDR1, LCDR2, LCDR3, each of which may have at least about 80% (eg, 85%, 90% or 95%) sequence identity to HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, LCDR3 of a Fresolimumab molecule, respectively.
  • the light chain variable region can have at least about 80% sequence identity with the light chain variable region of a Fresolimumab molecule.
  • the anti-TGF ⁇ antibody or antigen-binding fragment thereof comprises a heavy chain variable region and a light chain variable region of an antibody, each of which may have at least about 80% of the heavy chain variable region and light chain variable region, respectively, of a Fresolimumab molecule. % (eg, about 85%, 90% or 95%) sequence identity.
  • the light chain has at least about 80% sequence identity with the light chain of a Fresolimumab molecule.
  • the anti-TGF ⁇ antibody or antigen-binding fragment thereof comprises heavy and light chains of the antibody, each of which may be at least about 80% (eg, 85%, 90%, or 95%), respectively, of the heavy and light chains of a Fresolimumab molecule. ) sequence identity.
  • the anti-TGF ⁇ antibody or antigen-binding fragment thereof may comprise: Fresolimumab, or an antigen-binding fragment thereof, or a variant or biosimilar thereof.
  • the small molecule TGF ⁇ inhibitor can include the following compounds: SB525334, SD-208, SB431542, LY2109761, LY2157299, GW788388, RepSox, SIS3, LDN-193189, EW-7197, LY364947, or the same combination.
  • the TGF ⁇ RII can be a polypeptide having a wild-type human TGF ⁇ receptor type 2 isoform A sequence (eg, the amino acid sequence of NCBI Reference Sequence (Ref Seq) Accession No. NP_001020018), or a wild-type Polypeptides of human TGF ⁇ receptor type 2 isoform B sequence (e.g., the amino acid sequence of NCBI Ref Seq Accession No. NP_003233), or substantially identical (e.g., about 80%, 85%, 90%, or 95% identical to their amino acid sequence) sex) polypeptides.
  • a wild-type human TGF ⁇ receptor type 2 isoform A sequence eg, the amino acid sequence of NCBI Reference Sequence (Ref Seq) Accession No. NP_001020018
  • a wild-type Polypeptides of human TGF ⁇ receptor type 2 isoform B sequence e.g., the amino acid sequence of NCBI Ref Seq Accession No. NP_003233
  • the functionally active fragment can comprise: human TGF ⁇ RII extracellular domain (ECD), any portion of NCBI Ref Seq Accession No. NP_001020018 or NCBI Ref Seq Accession No. NP_003233, or substantially identical to its amino acid sequence (e.g. , about 80%, 85%, 90% or 95% identical) sequences.
  • ECD human TGF ⁇ RII extracellular domain
  • TGF ⁇ RII or functionally active fragment thereof may comprise:
  • (c) has an amino acid sequence with one or more amino acids added, deleted and/or substituted compared to the amino acid sequence shown in SEQ ID NO:6.
  • bifunctional immune checkpoint/TGF ⁇ inhibitor is a fusion protein.
  • the PD-L1/TGF ⁇ dual inhibitor or PD-1/TGF ⁇ dual inhibitor is a fusion protein.
  • the dual PD-L1//TGF ⁇ inhibitor comprises a PD-L1 targeting moiety and a TGF ⁇ receptor domain comprising transforming growth factor beta receptor II (TGF ⁇ RII ) or a functionally active fragment thereof.
  • TGF ⁇ RII transforming growth factor beta receptor II
  • the dual PD-L1//TGF ⁇ inhibitor comprises a polypeptide
  • the polypeptide comprises at least: (i) the heavy chain variable region of an anti-PD-L1 antibody; and (ii) TGF ⁇ RII or its functionally active fragments.
  • the anti-PD-L1 antibody heavy chain variable region comprises HCDR1, HCDR2, HCDR3, and the HCDR1 comprises an amino acid sequence with at least 70% sequence identity to the amino acid sequence set forth in SEQ ID NO: 1
  • the HCDR2 comprises an amino acid sequence having at least 70% sequence identity with the amino acid sequence shown in SEQ ID NO: 2
  • the HCDR3 comprises an amino acid sequence having at least 70% sequence identity with the amino acid sequence shown in SEQ ID NO: 3.
  • the anti-PD-L1 antibody heavy chain variable region comprises HCDR1, HCDR2, HCDR3, and the HCDR1 comprises the amino acid sequence shown in SEQ ID NO: 1 or by amino acid addition, elimination or substitution
  • the amino acid sequence obtained by the reaction has no more than 2 amino acid differences with the amino acid sequence shown in SEQ ID NO: 1;
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO: 2 or through amino acid addition, elimination or substitution reaction
  • the obtained amino acid sequence with the amino acid sequence shown in SEQ ID NO: 2 is no more than 2 amino acid differences;
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO: 3 or is obtained by amino acid addition, elimination or substitution reaction.
  • the resulting amino acid sequence has no more than 2 amino acid differences with the amino acid sequence shown in SEQ ID NO: 3.
  • the anti-PD-L1 antibody heavy chain variable region comprises an amino acid sequence selected from the group consisting of: (a) the amino acid sequence shown in SEQ ID NO: 4; (b) the same as SEQ ID NO: : an amino acid sequence with at least 85%, 90%, 95% or 99% sequence identity to the amino acid sequence shown in 4; Amino acid sequences have 1 or more differing amino acid sequences.
  • TGF ⁇ RII or functionally active fragment thereof comprises:
  • (c) has an amino acid sequence with one or more amino acids added, deleted and/or substituted compared to the amino acid sequence shown in SEQ ID NO:6.
  • polypeptide further comprises a linker linking the C-terminus of the heavy chain variable region of the anti-PD-L1 antibody or antigen-binding fragment thereof to the TGF ⁇ RII or its function
  • linker linking the C-terminus of the heavy chain variable region of the anti-PD-L1 antibody or antigen-binding fragment thereof to the TGF ⁇ RII or its function
  • the N-termini of the active fragments are linked.
  • the polypeptide further comprises CH2, CH3 domains, and the polypeptide comprises, from the N-terminus to the C-terminus, the heavy chain variable region (VH), CH2, CH3 domains and TGF ⁇ RII or its functionally active fragment, the C-terminus of the CH3 domain and the N-terminus of the TGF ⁇ RII or its functionally active fragment are connected by a linker.
  • VH heavy chain variable region
  • CH2, CH3 domains and TGF ⁇ RII or its functionally active fragment the C-terminus of the CH3 domain and the N-terminus of the TGF ⁇ RII or its functionally active fragment are connected by a linker.
  • CH2 domains are derived from IgG.
  • linker is a peptide linker
  • amino acid sequence of the peptide linker is G( G4S ) x , wherein x is 3-6.
  • the peptide linker comprises an amino acid sequence selected from the group consisting of: (a) the amino acid sequence set forth in SEQ ID NO:5; (b) having the amino acid sequence set forth in SEQ ID NO:5 An amino acid sequence having at least 85%, 90%, 95% or 99% sequence identity; and (c) an amino acid sequence obtained by addition, elimination or substitution reaction with 1 or more amino acid sequences shown in SEQ ID NO:5 Multiple differential amino acid sequences.
  • polypeptide comprises an amino acid sequence selected from the group consisting of: (a) the amino acid sequence set forth in SEQ ID NO:7; (b) having at least the amino acid sequence set forth in SEQ ID NO:7 An amino acid sequence of 85%, 90%, 95% or 99% sequence identity; (c) the amino acid sequence obtained by addition, elimination or substitution reaction has 1 or more differences with the amino acid sequence shown in SEQ ID NO: 7 amino acid sequence.
  • the PD-L1//TGF ⁇ dual inhibitor comprises two of the aforementioned polypeptides.
  • the dual PD-L1/TGF ⁇ inhibitor comprises a first polypeptide and a second polypeptide
  • the first polypeptide comprises at least: (i) the heavy chain of an anti-PD-L1 antibody and (ii) TGF ⁇ RII or a functionally active fragment thereof
  • the second polypeptide comprises at least the light chain variable region of an anti-PD-L1 antibody
  • the heavy chain variable region of the first polypeptide and The light chain variable regions of the second polypeptide are capable of specifically binding PD-L1 when combined.
  • the first polypeptide further comprises a linker that connects the C-terminus of the heavy chain variable region of the anti-PD-L1 antibody or antigen-binding fragment thereof to the TGF ⁇ RII or the N-terminus of a functionally active fragment thereof.
  • the dual PD-L1/TGF ⁇ inhibitor comprises a first polypeptide and a second polypeptide, wherein:
  • the first polypeptide sequentially comprises the heavy chain variable region (VH) of the anti-PD-L1 antibody, the CH1 domain, and TGF ⁇ RII or a functionally active fragment thereof from the N-terminus to the C-terminus, and the C-terminus of the CH1 domain is related to the The N-terminus of the TGF ⁇ RII or its functionally active fragment is connected by a linker; and the second polypeptide comprises a light chain variable region (VL) and a light chain constant region (CL) of an anti-PD-L1 antibody from the N-terminus to the C-terminus ).
  • the dual PD-L1/TGF ⁇ inhibitor comprises a first polypeptide and a second polypeptide, wherein:
  • the first polypeptide comprises the heavy chain variable region (VH), CH1, CH2, CH3 domains of the anti-PD-L1 antibody and TGF ⁇ RII or a functionally active fragment thereof in sequence from the N-terminus to the C-terminus, and the CH3 domain is The C-terminus is connected with the N-terminus of the TGF ⁇ RII or its functionally active fragment through a linker; and the second polypeptide comprises the light chain variable region (VL), light chain of an anti-PD-L1 antibody from the N-terminus to the C-terminus Constant region (CL).
  • VH heavy chain variable region
  • linker is a peptide linker
  • the first polypeptide has at least about 80% sequence identity to a functionally identical polypeptide of M7824 or SHR-1701.
  • the second polypeptide has at least about 80% sequence identity to a functionally identical polypeptide of M7824 or SHR-1701.
  • the dual PD-L1/TGF ⁇ inhibitor comprises M7824, SHR-1701, or a variant or biosimilar thereof, or a combination thereof.
  • a PI3K inhibitor can be a compound that inhibits one or more PI3K isoforms, eg, alpha, beta, delta, or gamma isoforms.
  • the PI3K inhibitor can be a compound that inhibits the alpha, beta, delta and gamma isoforms of PI3K.
  • the PI3K inhibitor can be a compound that inhibits the beta, delta, and gamma isoforms of PI3K.
  • the PI3K inhibitor may be a compound that inhibits the delta and gamma isoforms of PI3K, ie, a dual PI3K delta/gamma inhibitor.
  • the PI3K inhibitor can comprise a compound of formula (IA-I), (IA-II), (IA-III), or (IA-IV):
  • R can be independently selected from hydrogen, halogen, -OR a , CN, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl, substituted or unsubstituted or unsubstituted C 2-6 alkynyl, substituted or unsubstituted C 3-8 cycloalkyl and substituted or unsubstituted heterocyclic groups;
  • Cy 1 may be selected from substituted or unsubstituted C 3-8 cycloalkyl, substituted or unsubstituted 3- to 15-membered heterocyclic group having at least one heteroatom selected from N, O and S , substituted or unsubstituted C6-20 aryl and substituted or unsubstituted monocyclic groups of 5- to 14-membered heteroaryl groups having one or more heteroatoms selected from N, O and S;
  • Each occurrence of R a may be the same or different and independently selected from hydrogen, halogen, hydroxy, cyano, substituted or unsubstituted C 1-6 alkyl; -NR c R d , where R c , R d independently selected from hydrogen, halogen, hydroxy, cyano, substituted or unsubstituted C1-6 alkyl and C1-6 alkoxy, and -ORc , wherein Rc is substituted or unsubstituted C 1-6 alkyl;
  • n may be an integer from 1-4;
  • q can be 0, 1 or 2;
  • Each occurrence of X can be independently selected from CR 3 or N;
  • R can be independently selected from hydrogen, hydroxy, halogen, carboxyl, cyano, nitro, substituted or unsubstituted C 1-8 alkyl, substituted or unsubstituted C 1- 8 alkoxy, substituted or unsubstituted C 2-10 alkenyl, substituted or unsubstituted C 2-12 alkynyl, substituted or unsubstituted C 6-20 aryl, substituted or unsubstituted Unsubstituted C 6-20 aryl C 1-8 alkyl, substituted or unsubstituted C 3-20 cycloalkyl, substituted or unsubstituted C 3-20 cycloalkyl C 1-8 Alkyl, substituted or unsubstituted C 3-8 cycloalkenyl C 1-8 alkyl, substituted or unsubstituted C 3-8 cycloalkenyl, substituted or unsubstituted with one or more 5- to 14-membered heteroaryl
  • guanidine, -COORx , -C(O)Rx, -C(S) Rx , -C(O ) NRxRy , -C (O ) ONRxRy , - NR y R z , -NR x CONR y R z , -N(R x )SOR y , -N(Rx)SO 2 R y , -( NN(R x )R y ), -NR x C(O ) ORy , -NRxRy , -NRxC ( O) Ry -,- NRxC ( S) Ry , -NRxC ( S) NRyRz , -SONRxRy- , -SO 2 NR x R y -, -OR x , -OR x C(O)NR y R z , -OR x C(O)OR y
  • the PI3K inhibitor can include a compound of formula (IA-V):
  • R can be independently selected from hydrogen, halogen, -OR a , CN, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl, substituted or unsubstituted or unsubstituted C 2-6 alkynyl, substituted or unsubstituted C 3-8 cycloalkyl and substituted or unsubstituted heterocyclic groups;
  • a saturated 3-10 membered ring including carbon atoms bound to R and R, the ring may optionally include one or more heteroatoms that are the same or different and selected from O, NR a and S;
  • Each occurrence of X can be independently selected from CR 3 or N;
  • Each occurrence of R can be independently selected from hydrogen, hydroxy, halogen, carboxyl, cyano, nitro, substituted or unsubstituted C 1-8 alkyl, substituted or unsubstituted C 1-8 Alkoxy, substituted or unsubstituted C 2-10 alkenyl, substituted or unsubstituted C 2-12 alkynyl, substituted or unsubstituted C 6-20 aryl, substituted or unsubstituted Substituted C 6-20 aryl C 1-8 alkyl, substituted or unsubstituted C 3-20 cycloalkyl, substituted or unsubstituted C 3-20 cycloalkyl C 1-8 alkane base, substituted or unsubstituted C 3-8 cycloalkenyl C 1-8 alkyl, substituted or unsubstituted C 3-8 cycloalkenyl, substituted or unsubstituted with one or more 5- to 14-membered heteroaryl
  • R 5 can be hydrogen, C 1-6 alkyl or halogen
  • n can be 0, 1, 2, 3, or 4; and p can be 0, 1, 2, 3, 4, or 5.
  • the PI3K inhibitor can include a compound of formula (IA-VI):
  • R can be independently selected from hydrogen, halogen, -OR a , CN, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted C 2-6 alkenyl, substituted or unsubstituted or unsubstituted C 2-6 alkynyl, substituted or unsubstituted C 3-8 cycloalkyl and substituted or unsubstituted heterocyclic groups;
  • Each occurrence of X can be independently selected from CR 3 or N;
  • Each occurrence of R can be independently selected from hydrogen, hydroxy, halogen, carboxyl, cyano, nitro, substituted or unsubstituted C 1-8 alkyl, substituted or unsubstituted C 1-8 Alkoxy, substituted or unsubstituted C 2-10 alkenyl, substituted or unsubstituted C 2-12 alkynyl, substituted or unsubstituted C 6-20 aryl, substituted or unsubstituted Substituted C 6-20 aryl C 1-8 alkyl, substituted or unsubstituted C 3-20 cycloalkyl, substituted or unsubstituted C 3-20 cycloalkyl C 1-8 alkane base, substituted or unsubstituted C 3-8 cycloalkenyl C 1-8 alkyl, substituted or unsubstituted C 3-8 cycloalkenyl, substituted or unsubstituted with one or more 5- to 14-membered heteroaryl
  • R 5 can be hydrogen, C 1-6 alkyl or halogen
  • n can be 0, 1, 2, 3 or 4;
  • P can be 0, 1, 2, 3, 4 or 5.
  • R can be hydrogen, halogen, substituted or unsubstituted C1-6 alkyl, or OR a , for another example, wherein R a can be alkyl.
  • Cy 1 can be selected from:
  • R 1 and R 2 may each independently represent hydrogen or substituted or unsubstituted C 1-6 alkyl.
  • R3 can be iodo, cyano, substituted or unsubstituted alkyl, substituted or unsubstituted alkynyl, substituted or unsubstituted aryl, substituted or unsubstituted Substituted heteroaryl.
  • R 3 can be hydrogen, halogen, hydroxy, or NH 2 , as another example, wherein said R 3 can be hydrogen.
  • the PI3K inhibitor can include:
  • the PI3K inhibitor may comprise: 2-(1-(9H-purin-6-ylamino)propyl)-3-(3-fluorophenyl)-4H-chromene-4 - Ketones, or their enantiomers, mixtures of enantiomers, mixtures of two or more diastereomers, or isotopic variants; or pharmaceutically acceptable salts, solvates thereof , hydrate or prodrug.
  • the PI3K inhibitor may be (S)-2-(1-(9H-purin-6-ylamino)propyl)-3-(3-fluorophenyl)-4H-chrome En-4-one (Tenalisib, also known as CN401) or a pharmaceutically acceptable salt, solvate, hydrate or prodrug thereof.
  • the pharmaceutical combination may include an anti-PD-L1 antibody or antigen-binding fragment thereof and a PI3K inhibitor.
  • the pharmaceutical combination may include: (i) an anti-PD-L1 antibody or antigen-binding fragment thereof comprising Atezolizumab, Avelumab, BMS-936559, MPDL3280A or durvalumab or an antigen thereof Binding fragments; (ii) dual PI3K ⁇ / ⁇ inhibitors including 2-(1-(9H-purin-6-ylamino)propyl)-3-(3-fluorophenyl) -4H-chromen-4-one, or an enantiomer, mixture of enantiomers, mixture of two or more diastereomers, or isotopic variant thereof; or a pharmaceutically acceptable variant thereof Acceptable salts, solvates, hydrates or prodrugs.
  • an anti-PD-L1 antibody or antigen-binding fragment thereof comprising Atezolizumab, Avelumab, BMS-936559, MPDL3280A or durvalumab or an antigen thereof Binding fragments
  • the pharmaceutical combination can include: (i) an anti-PD-L1 antibody selected from the group consisting of Atezolizumab, Avelumab, BMS-936559, MPDL3280A, durvalumab, or a combination thereof; (ii) (S)-2-(1 -(9H-purin-6-ylamino)propyl)-3-(3-fluorophenyl)-4H-chromen-4-one or a pharmaceutically acceptable salt, solvate, hydrate or prodrug thereof .
  • the pharmaceutical combination may include a TGF ⁇ inhibitor and a PI3K inhibitor.
  • the pharmaceutical combination may include: (i) a TGF ⁇ inhibitor comprising: an anti-TGF ⁇ antibody or antigen-binding fragment thereof, a small molecule TGF ⁇ inhibitor or a combination thereof, the TGF ⁇ antibody or antigen-binding fragment thereof Including Fresolimumab or an antigen-binding fragment thereof, the small molecule TGF ⁇ inhibitor includes SB525334, SD-208, SB431542, LY2109761, LY2157299 (Galunisertib), GW788388, RepSox, SIS3, LDN-193189, EW-7197, LY364947 or a combination thereof; (ii) dual PI3K ⁇ / ⁇ inhibitors comprising 2-(1-(9H-purin-6-ylamino)propyl)-3-(3-fluorophenyl)-4H- A chromen-4-one, or an enantiomer, mixture of enantiomers, mixture of two or more diastereomers, or isotopic variant thereof; or
  • the pharmaceutical combination may include: Fresolimumab or an antigen-binding fragment thereof and (S)-2-(1-(9H-purin-6-ylamino)propyl)-3-(3-fluorophenyl)- 4H-chromen-4-one or a pharmaceutically acceptable salt, solvate, hydrate or prodrug thereof.
  • the pharmaceutical combination may include: Fresolimumab or an antigen-binding fragment thereof, SB431542, and (S)-2-(1-(9H-purin-6-ylamino)propyl)-3-(3-fluorophenyl) )-4H-chromen-4-one or a pharmaceutically acceptable salt, solvate, hydrate or prodrug thereof.
  • the pharmaceutical combination may include the PD-1 inhibitor, the TGF ⁇ inhibitor, and the PI3K inhibitor.
  • the pharmaceutical combination may include: (i) an anti-PD-1 antibody or antigen-binding fragment thereof, the anti-PD-1 antibody or antigen-binding fragment thereof including nivolumab, pembrolizumab, pidilizumab Anti- or AMP-514 or an antigen-binding fragment thereof; (ii) a dual PI3K ⁇ / ⁇ inhibitor comprising 2-(1-(9H-purin-6-ylamino)propyl)-3 -(3-Fluorophenyl)-4H-chromen-4-one, or its enantiomer, mixture of enantiomers, mixture of two or more diastereomers, or Isotopic variants; or pharmaceutically acceptable salts, solvates, hydrates or prodrugs thereof; (iii) anti-TGF ⁇ antibodies or antigen-binding fragments thereof, small molecule TGF ⁇ inhibitors or combinations thereof, said TGF ⁇ antibodies or antigens thereof Binding fragments include Fresolimumab or an anti-
  • the pharmaceutical combination can include: (i) an anti-PD-1 antibody selected from the group consisting of nivolumab, pembrolizumab, pidilizumab, or AMP-514, or a combination thereof; (ii) (S)-2-(1-(9H-purin-6-ylamino)propyl)-3-(3-fluorophenyl)-4H-chromen-4-one or a pharmaceutically acceptable salt thereof, A solvate, hydrate or prodrug; (iii) Fresolimumab or an antigen-binding fragment thereof and/or SB431542.
  • the pharmaceutical combination may include the PD-L1 inhibitor, the TGF ⁇ inhibitor, and the PI3K inhibitor.
  • the pharmaceutical combination may include: (i) an anti-PD-L1 antibody or antigen-binding fragment thereof comprising Atezolizumab, Avelumab, BMS-936559, MPDL3280A or durvalumab or an antigen thereof Binding fragments; (ii) dual PI3K ⁇ / ⁇ inhibitors including 2-(1-(9H-purin-6-ylamino)propyl)-3-(3-fluorophenyl) -4H-chromen-4-one, or an enantiomer, mixture of enantiomers, mixture of two or more diastereomers, or isotopic variant thereof; or a pharmaceutically acceptable variant thereof acceptable salts, solvates, hydrates or prodrugs; (iii) anti-TGF ⁇ antibodies or antigen-binding fragments thereof including Fresolimumab or antigen-binding fragments thereof.
  • the pharmaceutical combination may include: (i) an anti-PD-L1 antibody selected from the group consisting of Atezolizumab, Avelumab, BMS-936559, MPDL3280A, or durvalumab, or a combination thereof; (ii) (S)-2-(1 -(9H-purin-6-ylamino)propyl)-3-(3-fluorophenyl)-4H-chromen-4-one or a pharmaceutically acceptable salt, solvate, hydrate or prodrug thereof ; (iii) Fresolimumab or an antigen-binding fragment thereof.
  • an anti-PD-L1 antibody selected from the group consisting of Atezolizumab, Avelumab, BMS-936559, MPDL3280A, or durvalumab, or a combination thereof.
  • the pharmaceutical combination may include the dual PD-L1/TGF ⁇ inhibitor and a PI3K inhibitor.
  • the PD-L1/TGF ⁇ dual inhibitor may be those described in PCT/CN2019124535, the entire contents of which are incorporated herein by reference.
  • the dual PD-L1/TGF ⁇ inhibitor can be WBP1126 (also known as CN202), or a variant or biosimilar thereof.
  • the WBP1126 can comprise two polypeptides, the polypeptides from the N-terminus to the C-terminus sequentially comprise the heavy chain variable region (VH), CH2, CH3 domains of an anti-PD-L1 antibody, and TGF ⁇ RII or a functionally active fragment thereof, the The C-terminus of the CH3 domain is connected to the N-terminus of the TGF ⁇ RII or its functionally active fragment through a linker; for example, the polypeptide may have the amino acid sequence shown in Table 1:
  • the dual PD-L1/TGF ⁇ inhibitor comprises a first polypeptide and a second polypeptide
  • the first polypeptide comprises at least: (i) the heavy chain of an anti-PD-L1 antibody and (ii) TGF ⁇ RII or a functionally active fragment thereof
  • the second polypeptide comprises at least the light chain variable region of an anti-PD-L1 antibody
  • the heavy chain variable region of the first polypeptide and The light chain variable regions of the second polypeptide are capable of specifically binding PD-L1 when combined.
  • the dual PD-L1/TGF ⁇ inhibitor further comprises a linker connecting the C-terminus of the heavy chain variable region of the anti-PD-L1 antibody or antigen-binding fragment thereof linked to the N-terminus of the TGF ⁇ RII or its functionally active fragment.
  • the dual PD-L1/TGF ⁇ inhibitor comprises a first polypeptide and a second polypeptide, wherein:
  • the first polypeptide sequentially comprises the heavy chain variable region (VH) of the anti-PD-L1 antibody, the CH1 domain, and TGF ⁇ RII or its functionally active fragment from the N-terminus to the C-terminus, and the C-terminus of the CH1 domain is related to the The N-terminus of the TGF ⁇ RII or its functionally active fragment is connected by a linker; and the second polypeptide comprises a light chain variable region (VL) and a light chain constant region (CL) of an anti-PD-L1 antibody from the N-terminus to the C-terminus ).
  • the dual PD-L1/TGF ⁇ inhibitor comprises a first polypeptide and a second polypeptide, wherein:
  • the first polypeptide comprises the heavy chain variable region (VH), CH1, CH2, CH3 domains of the anti-PD-L1 antibody and TGF ⁇ RII or its functionally active fragments in sequence from the N-terminus to the C-terminus, and the CH3 domain is The C-terminus is connected with the N-terminus of the TGF ⁇ RII or its functionally active fragment through a linker; and the second polypeptide comprises the light chain variable region (VL), light chain of an anti-PD-L1 antibody from the N-terminus to the C-terminus Constant region (CL).
  • VH heavy chain variable region
  • CH1, CH2, CH3 domains of the anti-PD-L1 antibody and TGF ⁇ RII or its functionally active fragments in sequence from the N-terminus to the C-terminus
  • the CH3 domain is The C-terminus is connected with the N-terminus of the TGF ⁇ RII or its functionally active fragment through a linker
  • the second polypeptide comprises the light chain variable region (VL), light chain
  • linker is a peptide linker
  • the first polypeptide of the dual PD-L1/TGF ⁇ inhibitor may have at least about 80% sequence identity to a functionally identical polypeptide of M7824 or SHR-1701.
  • the second polypeptide of the dual PD-L1/TGF ⁇ inhibitor may have at least about 80% sequence identity to a functionally identical polypeptide of M7824 or SHR-1701.
  • the PD-L1/TGF ⁇ dual inhibitor may be those described in PCT/EP2015052781, PCT/CN2018086451, all of which are incorporated herein by reference in their entirety.
  • the PD-L1/TGF ⁇ dual inhibitor can be selected from M7824, SHR-1701, or variants or biosimilars thereof, or a combination thereof.
  • the dual PD-L1/TGF ⁇ inhibitor and the PI3K inhibitor may not be mixed with each other in the pharmaceutical combination.
  • the dual PD-L1/TGF ⁇ inhibitor and the PI3K inhibitor can each be independently present in separate containers.
  • the present application provides a pharmaceutical composition
  • a pharmaceutical composition comprising the aforementioned PI3K inhibitor and a second therapeutic agent, and optionally one or more pharmaceutically acceptable carriers or excipients.
  • the second therapeutic agent is the aforementioned dual PD-L1/TGF ⁇ inhibitor.
  • the PI3K inhibitor is the aforementioned dual PI3K delta/gamma inhibitor.
  • the pharmaceutical composition includes a dual PD-L1/TGF ⁇ inhibitor and a PI3K ⁇ / ⁇ dual inhibitor.
  • the pharmaceutical composition may comprise: i) WBP1126 or a variant or biosimilar thereof;
  • the second therapeutic agent is the aforementioned anti-PD-L1 antibody
  • the PI3K inhibitor is the aforementioned dual PI3K ⁇ / ⁇ inhibitor.
  • the pharmaceutical composition may include: i) nivolumab, pembrolizumab, pidilizumab, or a combination thereof; and ii) Tenalisib or a pharmaceutically acceptable salt, solvate, hydrate thereof or prodrugs.
  • the second therapeutic agent is the aforementioned dual PD-L1/TGF ⁇ inhibitor
  • the PI3K inhibitor is a PI3K ⁇ inhibitor
  • the pharmaceutical composition may comprise: i) WBP1126 or a variant or biosimilar thereof;
  • Alplisib or a pharmaceutically acceptable salt, solvate, hydrate or prodrug thereof.
  • the second therapeutic agent and the PI3K inhibitor are present in a single or separate dosage form.
  • per unit dosage form comprises a dose of 1-1000 mg of the first therapeutic agent, such as a PI3K inhibitor active substance (eg CN401 (Tenalisib)), For example 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 40, 50, 60 , 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1000mg or a value between any two of the above; and, each unit dosage form (unit dosage form) contains a second therapeutic agent in a dose of 1-1000 mg, such as a PD-L1 inhibitor, a TGF ⁇ inhibitor, a PD-L1 inhibitor, Any one of PD-L1/TGF ⁇ dual inhibitor (eg CN202 (WBP1126)) or PD-1/TGF ⁇ dual inhibitor or combination thereof, eg 1,
  • a PI3K inhibitor active substance eg CN
  • the pharmaceutical combinations of the present application may be used in combination or in combination with one or more third active substances or therapies useful in the treatment, prevention or amelioration of a proliferative disorder, disease or condition.
  • the drug combinations of the present application are administered in combination with any conventional anti-tumor therapy including, but not limited to, immunotherapy, therapeutic antibodies, targeted therapy, surgery, radiation therapy, or chemotherapy.
  • the active substance may be a chemotherapeutic drug, for example, an alkylating agent, an antimetabolite, an anthracycline, a plant alkaloid, a topoisomerase inhibitor, an antineoplastic antibiotic, a hormonal drug, an antiangiogenic agent, a differentiation inducer , cell growth arrest inducers, apoptosis inducers, cytotoxic drugs and other antitumor drugs.
  • chemotherapeutic drug for example, an alkylating agent, an antimetabolite, an anthracycline, a plant alkaloid, a topoisomerase inhibitor, an antineoplastic antibiotic, a hormonal drug, an antiangiogenic agent, a differentiation inducer , cell growth arrest inducers, apoptosis inducers, cytotoxic drugs and other antitumor drugs.
  • chemotherapeutic drug for example, an alkylating agent, an antimetabolite, an anthracycline, a plant alkaloid, a top
  • chemotherapeutic agents include, but are not limited to, alkylating agents (eg, cisplatin, carboplatin, oxaliplatin, dichloromethyldiethylamine, cyclophosphamide, chlorambucil, dacarbazine, lolimus) pyridoxine, carmustine, procarbazine, chlorambucil, and ifosfamide), antimetabolites (eg, fluorouracil (5-FU), gemcitabine, methotrexate, cytarabine, fludara pyridine and fluorouracil), antimitotics (including taxanes (such as paclitaxel and docetaxel) and vinca alkaloids (such as vinblastine, vincristine, vinblastine, vinorelbine, and vinblastine) Desine), anthracyclines (including doxorubicin, daunorubicin, valrubicin, idarubicin, and epirubicin, and
  • the third active substance can be selected from vincristine, vinblastine, vindesine, etoposide, docetaxel, paclitaxel, irinotecan, vinorelbine, mitoxantrone , any one of vinflunine and topotecan or any combination thereof.
  • third therapies that can be used in conjunction with the methods provided herein include, but are not limited to, surgery, radiation therapy, endocrine therapy, biological response modifiers (eg, interferons, interleukins, and tumor necrosis factor (TNF)) , hyperthermia and cryotherapy, and medications to reduce any adverse effects (eg, antiemetics).
  • biological response modifiers eg, interferons, interleukins, and tumor necrosis factor (TNF)
  • hyperthermia and cryotherapy e.g, hyperthermia and cryotherapy
  • medications to reduce any adverse effects eg, antiemetics.
  • the present application provides a method for treating and/or preventing tumors, comprising administering to a subject in need thereof: an effective amount of the aforementioned pharmaceutical combination, or the aforementioned pharmaceutical composition.
  • the method comprises administering to a subject in need thereof: an effective amount of the aforementioned PI3K inhibitor and the aforementioned inhibitor of immune checkpoints. In certain embodiments, the method comprises administering to a subject in need thereof: an effective amount of the aforementioned PI3K inhibitor and the aforementioned TGF ⁇ inhibitor. In certain embodiments, the method comprises administering to a subject in need thereof: an effective amount of the aforementioned PI3K inhibitor, the aforementioned TGF ⁇ inhibitor, and the aforementioned inhibitor of immune checkpoints.
  • the method can comprise administering to a subject in need thereof: an effective amount of the aforementioned PI3K inhibitor, such as the aforementioned (S)-2-(1-(9H-purin-6-ylamino)propyl) -3-(3-fluorophenyl)-4H-chromen-4-one or a pharmaceutically acceptable salt thereof and the aforementioned anti-PD-L1 antibody, for example, Atezolizumab, Avelumab, BMS-936559, MPDL3280A, durvalumab, or their combination.
  • the aforementioned PI3K inhibitor such as the aforementioned (S)-2-(1-(9H-purin-6-ylamino)propyl) -3-(3-fluorophenyl)-4H-chromen-4-one or a pharmaceutically acceptable salt thereof
  • the aforementioned anti-PD-L1 antibody for example, Atezolizumab, Avelumab, BMS-936559, MPDL3280A, durvalumab
  • the method can comprise administering to a subject in need thereof: an effective amount of the aforementioned PI3K inhibitor, such as the aforementioned (S)-2-(1-(9H-purin-6-ylamino)propyl) -3-(3-Fluorophenyl)-4H-chromen-4-one or a pharmaceutically acceptable salt thereof and the aforementioned TGF ⁇ inhibitor, eg, Fresolimumab, SB525334 or a combination thereof.
  • the aforementioned PI3K inhibitor such as the aforementioned (S)-2-(1-(9H-purin-6-ylamino)propyl) -3-(3-Fluorophenyl)-4H-chromen-4-one or a pharmaceutically acceptable salt thereof
  • the aforementioned TGF ⁇ inhibitor eg, Fresolimumab, SB525334 or a combination thereof.
  • the method can comprise administering to a subject in need thereof: an effective amount of the aforementioned PI3K inhibitor, such as the aforementioned (S)-2-(1-(9H-purin-6-ylamino)propyl) -3-(3-Fluorophenyl)-4H-chromen-4-one or a pharmaceutically acceptable salt thereof; the aforementioned TGF ⁇ inhibitors, eg, Fresolimumab, SB525334 or a combination thereof; the aforementioned anti-PD-L1 Antibodies, eg, Atezolizumab, Avelumab, BMS-936559, MPDL3280A, durvalumab, or a combination thereof.
  • the aforementioned PI3K inhibitor such as the aforementioned (S)-2-(1-(9H-purin-6-ylamino)propyl) -3-(3-Fluorophenyl)-4H-chromen-4-one or a pharmaceutically acceptable salt thereof
  • the method can comprise administering to a subject in need thereof: an effective amount of the aforementioned PI3K inhibitor, such as the aforementioned (S)-2-(1-(9H-purin-6-ylamino)propyl) -3-(3-fluorophenyl)-4H-chromen-4-one or a pharmaceutically acceptable salt thereof; the aforementioned TGF ⁇ inhibitors, eg, Fresolimumab, SB525334 or a combination thereof; the aforementioned anti-PD-1 Antibodies, eg, nivolumab, pembrolizumab, pidilizumab, or a combination thereof.
  • the aforementioned PI3K inhibitor such as the aforementioned (S)-2-(1-(9H-purin-6-ylamino)propyl) -3-(3-fluorophenyl)-4H-chromen-4-one or a pharmaceutically acceptable salt thereof
  • the aforementioned TGF ⁇ inhibitors eg, Fresolim
  • the method comprises administering to a subject in need thereof: an effective amount of the aforementioned PI3K inhibitor and the aforementioned dual PD-L1/TGF ⁇ inhibitor.
  • the method can comprise administering to a subject in need thereof: an effective amount of the aforementioned PI3K inhibitor, such as the aforementioned (S)-2-(1-(9H-purin-6-ylamino)propyl) -3-(3-fluorophenyl)-4H-chromen-4-one; and the aforementioned dual PD-L1/TGF ⁇ inhibitors, such as WBP1126, M7824, SHR-1701, or variants or biosimilars thereof, or a combination thereof.
  • the aforementioned PI3K inhibitor such as the aforementioned (S)-2-(1-(9H-purin-6-ylamino)propyl) -3-(3-fluorophenyl)-4H-chromen-4-one
  • the aforementioned dual PD-L1/TGF ⁇ inhibitors such as WBP1126, M7824, SHR-1701, or variants or biosimilars thereof, or a combination thereof.
  • the PI3K inhibitor can be administered concurrently with the dual PD-L1/TGF ⁇ inhibitor.
  • the PI3K inhibitor can be administered after the dual PD-L1/TGF ⁇ inhibitor.
  • the PI3K inhibitor can be administered before the dual PD-L1/TGF ⁇ inhibitor.
  • it also includes administering to a subject in need thereof an effective amount of the aforementioned small molecule TGF ⁇ inhibitor.
  • the tumor can include solid tumors and non-solid tumors.
  • the tumor may comprise a tumor with abnormal PI3K expression.
  • the tumor may comprise a tumor with abnormal PD-L1 expression.
  • the tumor may comprise a tumor with abnormal expression of TGF[beta].
  • the tumor may comprise a gastrointestinal tumor, melanoma, or lymphoma.
  • the methods of the present application delay resistance compared to the time at which resistance typically develops when treating a subject with either an agent or an inhibitor alone as a monotherapy.
  • resistance is delayed by at least 2 weeks, eg, at least 2 weeks, 4 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 8 months, 10 months, 12 months, 1 year, 2 years, 4 years, 6 years, 8 years or more.
  • remission e.g., complete remission or partial remission
  • remission (eg, complete remission or partial remission) extends for at least 2 weeks, eg, at least 2 weeks, 4 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 8 months, 10 months, 12 months, 1 year, 2 years, 4 years, 6 years, 8 years or more.
  • the addition of a PI3K inhibitor or a second agent to the treatment regimen increases or restores sensitivity to the cancer-resistant agent.
  • adding a second agent to a treatment regimen increases or restores sensitivity to a cancer-resistant PI3K inhibitor.
  • MRD minimal residual disease
  • the methods described herein include selecting a subject for treatment with a combination of a PI3K inhibitor and a second agent.
  • a subject eg, a patient suffering from a cancer, eg, a cancer described herein
  • selection is based on the presence of MRD above a preselected level (eg, 1 malignant cell in 100 normal cells, 1 malignant cell in 1000 normal cells, or 1 in 10,000 normal cells malignant tumor cells).
  • Methods for monitoring minimal residual disease negativity (MRD) are known in the art. See, eg, Zhou, J. et al., Blood, 2007, 110:1607-1611. Such methods include DNA-based tests or RNA-based tests.
  • MRD is monitored using flow cytometry, sequencing, or PCR.
  • the present application provides the use of the aforementioned pharmaceutical combination or the aforementioned pharmaceutical composition in the preparation of a medicament for treating and/or preventing tumors.
  • the use of the aforementioned PI3K inhibitors and the aforementioned inhibitors of immune checkpoints in the preparation of medicaments for treating and/or preventing tumors For example, the use of the aforementioned PI3K inhibitor and the aforementioned TGF ⁇ inhibitor in the preparation of a medicament for treating and/or preventing tumors.
  • the present application provides the aforementioned pharmaceutical combination or the aforementioned pharmaceutical composition for treating and/or preventing tumors.
  • the present application provides a method of inhibiting tumor growth, the method comprising contacting the tumor with the aforementioned pharmaceutical combination or the aforementioned pharmaceutical composition.
  • the method comprises contacting the tumor with the aforementioned PI3K inhibitor and the aforementioned inhibitor of immune checkpoints. In certain embodiments, the method comprises contacting the tumor with the aforementioned PI3K inhibitor and the aforementioned TGF ⁇ inhibitor. In certain embodiments, the method comprises contacting the tumor with the aforementioned PI3K inhibitor, the aforementioned inhibitor of an immune checkpoint, and the aforementioned TGF ⁇ inhibitor.
  • the present application provides a method of inhibiting tumor growth, the method comprising contacting the tumor with the aforementioned PI3K inhibitor and the aforementioned dual PD-L1/TGF ⁇ inhibitor.
  • the present application provides a kit comprising: (1) a first container, and the aforementioned PI3K inhibitor located in the first container; (2) a second container, and located in the first container The aforementioned dual PD-L1/TGF ⁇ inhibitor in two containers.
  • the present application provides a kit, which may include: (1) a first container, and the aforementioned PI3K inhibitor located in the first container; (2) a second container, and located in the first container The aforementioned inhibitor of immune checkpoint or the aforementioned TGF ⁇ inhibitor in the second container.
  • the kit further comprises (3) a third container, and the aforementioned inhibitor of immune checkpoint or the aforementioned TGF ⁇ inhibitor located in the third container, the agent in the third container Different from the medicament in the second container.
  • the dosage form of the drug in the kit can be an oral dosage form or an injectable dosage form.
  • the kit may also contain instructions.
  • mice (16-20 g) aged 6-8 weeks were purchased from Beijing Weitong Lihua Laboratory Animal Technology Co., Ltd., and 500,000 A20 cells were inoculated subcutaneously in each mouse, which were inoculated on the right back of the mouse.
  • the tumor volume grew to about 60mm 3 , the patients were randomly assigned to the control group and the experimental group.
  • the percent median regression for a group on a given day is then obtained by taking the median individual percent regression calculated for each animal in the group on that day.
  • the date of the calculation was determined on the day the ⁇ T/ ⁇ C (ie, the ratio of the median change from baseline in tumor volume between the experimental and experimental groups) was calculated, unless the median percent regression did not represent activity in that group. In that case, the first day when the median percent regression was maximal was determined to be the day.
  • Regression was defined as partial (PR) if the tumor volume dropped to 50% of the tumor volume at the start of treatment. Complete regression (CR) was considered to have been achieved when the tumor volume was below 14 mm3 or could not be recorded.
  • Example 1.1 Study on the combination of CN401 (Tenalisib) and CN202 (WBP1126) (5mg/kg) in A20 mouse B-cell lymphoma model (see Table 2 for experimental design)
  • 6-8-week-old balb/c mice (18-20 g) were purchased from Beijing Weitong Lihua Laboratory Animal Technology Co., Ltd. Each mouse was inoculated with 500,000 A20 cells subcutaneously and inoculated on the right back of the mouse. Treatment was initiated when the tumor had grown to about 62 mm in volume. Among them, CN401 was orally administered at 150 mg/kg twice a day for 3 weeks.
  • CN202 was administered by intraperitoneal injection at a dose of 5 mg/kg 3 times a week for 3 weeks.
  • the control group received vehicle without active product (0.5% MC and PBS). Tumor size and body weight were measured three times a week.
  • This animal experiment was carried out in accordance with the requirements of AAALAC, and was approved by the Bikai Animal Experiment Center IACUC to conduct this animal experiment.
  • Blank control group PBS i.p.tiw ⁇ 3weeks+0.5%MC p.o.bid ⁇ 3weeks
  • Blank control group PBS i.p.tiw ⁇ 3weeks+0.5%MC p.o.bid ⁇ 3weeks
  • CN401 150mg/kg p.o.bid ⁇ 3weeks 3
  • CN202 5mg/kg i.p.tiw ⁇ 3weeks 4
  • Example 1.2 Study on the combination of CN401 and CN202 (15mg/kg) in A20 mouse B-cell lymphoma model (see Table 4 for experimental design)
  • Example 1.1 Refer to the test method of Example 1.1, except that the dosage of CN202 is replaced by 5 mg/kg to 15 mg/kg.
  • Blank control group PBS i.p.tiw ⁇ 3weeks+0.5%MC p.o.bid ⁇ 3weeks
  • Blank control group PBS i.p.tiw ⁇ 3weeks+0.5%MC p.o.bid ⁇ 3weeks
  • CN401 150mg/kg PBS i.p.tiw ⁇ 3weeks+0.5%MC p.o.bid ⁇ 3weeks
  • CN202 15mg/kg i.p.tiw ⁇ 3weeks 4
  • CN401 combined with different doses of CN202 (5mg/kg, 15mg/kg) produced a synergistic effect on tumor inhibition, and had no significant effect on mouse body weight, showing good tolerance .
  • 6-8-week-old balb/c mice (18-20 g) were purchased from Beijing Weitong Lihua Laboratory Animal Technology Co., Ltd. Each mouse was inoculated with 500,000 A20 cells subcutaneously and inoculated on the right back of the mouse. Treatment was initiated when the tumor had grown to approximately 80 mm in volume. Among them, CN202 was administered by intraperitoneal injection, 3 times a week, at a dose of 5 mg/kg for 3 weeks. CN401 was administered orally at 150 mg/kg twice daily for 3 weeks. The control group received vehicle without active product (0.5% MC and PBS). Tumor size and body weight were measured three times a week. This animal experiment was carried out in accordance with the requirements of AAALAC, and was approved by the Bikai Animal Experiment Center IACUC to conduct this animal experiment.
  • Blank control group PBS i.p.tiw ⁇ 3weeks+0.5%MC p.o.bid ⁇ 3weeks
  • Blank control group PBS i.p.tiw ⁇ 3weeks+0.5%MC p.o.bid ⁇ 3weeks
  • Blank control group PBS i.p.tiw ⁇ 3weeks+0.5%MC p.o.bid ⁇ 3weeks
  • CN202 5mg/kg i.p.tiw ⁇ 3weeks 3
  • CN401 150mg/kg p.o.bid ⁇ 3weeks 4
  • Example 2.1 The test method of Example 2.1 was followed, except that the PI3K inhibitor was administered orally at 150 mg/kg twice a day from the CN401 (Tenalisib) administration, and was replaced by Alpelisib at 50 mg/kg orally once a day for 3 weeks , for 3 weeks.
  • 6-8-week-old balb/c mice (18-20 g) were purchased from Beijing Weitong Lihua Laboratory Animal Technology Co., Ltd. Each mouse was inoculated with 500,000 A20 cells subcutaneously and inoculated on the right back of the mouse. Treatment was initiated when the tumor had grown to about 62 mm in volume. CN401 was administered orally at 150 mg/kg twice daily for 3 weeks. Among them, the anti-PD-L1 antibody (Atezolizumab) was administered by intraperitoneal injection twice a week at a dose of 5 mg/kg for 3 weeks. The control group received vehicle without active product (0.5% MC and PBS). Tumor size and body weight were measured three times a week. This animal experiment was carried out in accordance with the requirements of AAALAC, and was approved by the Bikai Animal Experiment Center IACUC to conduct this animal experiment.
  • AAALAC AAALAC
  • Example 4 Study on the combination of CN202 and nab-pac and the combination of CN401, CN202 and nab-pac in the EMT-6 tumor model (see Table 12 for the experimental design)
  • balb/c mice (18-20g) were purchased from Beijing Weitong Lihua Laboratory Animal Technology Co., Ltd., and each mouse was inoculated with 500,000 EMT-6 cells (mouse triple negative breast cancer cells) , inoculated into the 2-3 pairs of mammary fat pads from top to bottom on the left side of the thorax of mice. Group dosing treatment was started when the tumor volume grew to about 87 mm 3 .
  • CN401 was administered orally at 150 mg/kg twice daily for 4 weeks.
  • CN202 and nab-paclitaxel were administered by intraperitoneal injection.
  • CN202 was administered 3 times a week at a dose of 15 mg/kg for 2 weeks; the same dose, CN202 was administered once a week for 2 weeks.
  • Nab-paclitaxel was administered twice weekly at a dose of 10 mg/kg for 2 weeks.
  • the control group received vehicle without active product (0.5% MC and PBS). Tumor size and body weight were measured three times a week. This animal experiment was carried out in accordance with the requirements of AAALAC, and was approved by the Bikai Animal Experiment Center IACUC to conduct this animal experiment.

Abstract

一种药物组合,其包含磷酸肌醇3-激酶(PI3K)抑制剂和第二治疗剂,其中所述第二治疗剂为免疫检查点的抑制剂,TGFβ抑制剂,双功能免疫检查点/TGFβ抑制剂或其组合。还提供了包括施用组合的治疗方法,和药物组合用于治疗肿瘤的用途。

Description

治疗肿瘤的方法和组合 技术领域
本申请涉及生物医药领域,具体的涉及一种治疗肿瘤的方法和组合。
背景技术
磷酸肌醇3-激酶(PI3K)信号转导途径是人类癌症中最高度突变的系统之一。PI3K是细胞内脂质激酶的独特且保守的家族的成员,其使磷脂酰肌醇或磷酸肌醇上的3’-OH基团磷酸化。PI3K家族包括15种激酶,其具有不同的底物特异性、表达模式和调节模式。I类PI3K(p110α、p110β、p110δ和p110γ)通常由酪氨酸激酶或G-蛋白偶联受体活化以产生(3,4,5)-三磷酸磷脂酰肌醇(PIP3),其接合下游效应子,如AKT/PDK1途径中的那些,mTOR、Tec家族激酶和Rho家族GTP酶。II类和III类PI3K通过合成3-二磷酸磷脂酰肌醇(PI(3)P)和(3,4)-二磷酸磷脂酰肌醇(PI(3,4)P2),在细胞内运输中起关键作用。PI3K是控制细胞生长(mTORC1)或监测基因组完整性(ATM、ATR、DNA-PK和hSmg-1)的蛋白激酶。
存在四种I类PI3K的哺乳动物亚型:PI3K-α、β、δ(Ia类PI3K)和PI3K-γ(Ib类PI3K)。这些酶催化PIP3的产生,导致对细胞存活、分化和功能重要的下游效应途径的活化。PI3K-α和PI3K-β广泛表达,是来自细胞表面受体的信号转导的重要介质。PI3K-α是最常见于癌症中的突变的亚型,并且在胰岛素信号转导和葡萄糖稳态中起作用(Knight等人Cell(2006)125(4):733–47;Vanhaesebroeck等人Current Topic Microbiol.Immunol.(2010)347:1–19)。PI3K-β在磷酸酶和张力蛋白同源物(PTEN)缺失的癌症中被活化。两种亚型都是研发中的小分子疗法的癌症靶标。
PI3K-δ和-γ优先在白细胞中表达,并且在白细胞功能中是重要的。这些亚型也有助于血液学恶性肿瘤的发展和维持(Vanhaesebroeck等人Current Topic Microbiol.Immunol.(2010)347:1–19;Clayton等人J Exp Med.(2002)196(6):753–63;Fung-Leung Cell Signal.(2011)23(4):603–8;Okkenhaug等人Science(2002)297(5583):1031–34)。PI3K-δ通过与PI3K调节亚基(p85)的Sarc同源2(SH2)结构域的相互作用或通过与RAS的直接相互作用被细胞受体(例如,受体酪氨酸激酶)活化。
转化生长因子β(TGFβ)是对免疫系统有显著影响的强效细胞因子。TGFβ在免疫系统中的主要功能是维持对外来病原体的耐受和初始免疫应答。在哺乳动物中鉴定到TGFβ的三种同种型,即TGFβ1、TGFβ2和TGFβ3,其中TGFβ1是主要的同种型。TGFβ以潜伏形式分泌,分泌的全部TGFβ中只有一小部分在生理条件下活化。TGFβ的生物学作用大部分通过TGFβ 与受体ALK5和TGFβ受体II(TGFβR2)的结合而实现。具体说,活性TGFβ二聚体能结合四聚体ALK5和TGFβR2复合物,以启动信号转导。TGFβ的初始结合不需要ALK5,但信号转导需要它。
程序性死亡受体1(programmed death 1,PD-1)为CD28超家族成员。PD-1表达于活化的T细胞,B细胞及髓系细胞,其有两个配体,即程序性死亡配体-1(programmed death ligand 1,PD-L1)和PD-L2。PD-L1与T细胞上的受体PD-1相互作用,在免疫应答的负调控方面发挥着重要作用。在许多人类肿瘤组织中均可检测到PD-L1蛋白的表达,肿瘤部位的微环境可诱导肿瘤细胞上的PD-L1的表达,表达的PD-L1有利于肿瘤的发生和生长,诱导抗肿瘤T细胞的凋亡。PD-1/PD-L1通路抑制剂可以阻断PD-1与PD-L1的结合,阻断负向调控信号,使T细胞恢复活性,从而增强免疫应答
转化生长因子-β(transforming growth factor-β,TGF-β)属于调节细胞生长和分化的TGF-β超家族。TGF-β通过异源四聚体受体复合物传递信号,这个受体复合物是由两个I型和两个II型的跨膜丝氨酸/苏氨酸激酶受体组成。
TGFβ能影响许多细胞功能,例如细胞增殖、分化、细胞-细胞和细胞-基质粘附、细胞运动和淋巴细胞的活化。(有关TGFβ在调节免疫应答中的作用的综述可参见Li等(2006)Annu.Rev.Immunol.24:99-146。)而且,相信TGFβ能诱导或介导许多疾病的进展,例如骨质疏松症,高血压,动脉粥样硬化,肝硬化,肾、肝和肺的纤维性疾病,以及肿瘤进展。在某些疾病,例如糖尿病性肾病、血管球性肾炎、环孢菌素介导的肾脏损伤和系统性红斑狼疮(SLE)的动物模型中,TGFβ可提高慢性炎症引起的末端器官损伤,而TGFβ拮抗剂能有效减轻这种损伤(Border等(1990)Nature346:371-374;Border等(1992)Nature 360:361-364;Isaka等(1999)KidneyInt.55:465-475;Sharma等(1996)Diabetes 45:522;Xin等(2004)Transplantation 15:1433;Benigni等(2003)J.Am.Soc.Nephrol.14:1816)。在癌症方面,TGFβ对恶性细胞可能有直接抑制作用,并可能提高一系列肿瘤生长因子和血管新生因子的产量或活性。
在肿瘤治疗中,人们早已认识到化疗所带来的高毒性及可导致耐药性癌细胞产生的负面影响。即使是靶向性针对与肿瘤生存生长相关的过度表达或激活的蛋白的治疗手段,仍会有癌细胞通过变异来减少或逃脱对靶向性治疗所针对通路的依赖,并利用其它的通路继续生存。肿瘤免疫治疗主要通过免疫学原理和方法,提高肿瘤细胞的免疫原性和对效应细胞杀伤的敏感性,激发和增强机体抗肿瘤免疫应答,并应用免疫细胞和效应分子输注宿主体内,协同机体免疫系统杀伤肿瘤、抑制肿瘤生长。降低治疗肿瘤药物毒性、抗性,减少毒副作用仍是肿瘤治疗亟待解决的问题。
发明内容
本申请的目的在于提供了包含PI3K抑制剂与选择的第二治疗剂的组合和方法。在某些实施方式中,已发现PI3K抑制剂与选自以下一种或多种的第二治疗剂的组合在治疗肿瘤中(例如,在减少癌细胞生长或活力或二者中)具有协同作用:免疫检查点的抑制剂,TGFβ抑制剂,或其组合。PI3K抑制剂和选择的第二治疗剂的组合可以允许PI3K抑制剂、第二治疗剂或二者与单一疗法剂量相比在实现相同的治疗效果的情况下,以所需的剂量更低的剂量施用。在一些实施方式中,所述组合可以允许PI3K抑制剂、第二治疗剂或二者比PI3K抑制剂或第二治疗剂作为单一疗法施用时以更低的频率施用。这样的组合可以提供有利的效果,例如在减少、预防、延迟和/或降低以下一种或多种后果的发生:原本与更高剂量的药剂的施用相关的副作用、毒性或抗性。
一方面,本申请提供一种药物组合,包含磷酸肌醇3-激酶(PI3K)抑制剂和第二治疗剂,其中所述第二治疗剂为免疫检查点的抑制剂,TGFβ抑制剂,双功能免疫检查点/TGFβ抑制剂或其组合。
在某些实施方式中,其中所述免疫检查点的抑制剂包括能够阻断程序性死亡1(PD-1)与程序性死亡配体1(PD-L1)相互作用的试剂。
在某些实施方式中,其中所述免疫检查点的抑制剂包括程序性死亡配体1(PD-L1)和/或程序性死亡1(PD-1)的抑制剂。
在某些实施方式中,其中所述双功能免疫检查点/TGFβ抑制剂包括PD-L1/TGFβ双重抑制剂,或PD-1/TGFβ双重抑制剂。
在某些实施方式中,所述药物组合包括:
1)PD-L1抑制剂和PI3K抑制剂的组合;
2)TGFβ抑制剂和PI3K抑制剂的组合;
3)PD-1抑制剂、TGFβ抑制剂和PI3K抑制剂的组合;
4)PD-L1抑制剂、TGFβ抑制剂和PI3K抑制剂的组合;
5)PD-L1/TGFβ双重抑制剂和PI3K抑制剂的组合;或
6)PD-1/TGFβ双重抑制剂和PI3K抑制剂的组合。
在某些实施方式中,其中所述免疫检查点的抑制剂包括核酸(例如,dsRNA、siRNA或shRNA),多肽(例如,可溶性配体,抗体或其抗原结合片段,免疫粘附素,融合蛋白,寡肽和其它分子),或化合物。
在某些实施方式中,其中所述抗体选自:人抗体、人源化抗体、嵌合抗体、多特异性抗 体、单克隆抗体和多克隆抗体。
在某些实施方式中,其中所述抗原结合片段选自:Fab、Fab’、F(ab’) 2、Fv、scFv、双抗体、Fd、dAb、VHH、大抗体和互补决定区(CDR)片段。
在某些实施方式中,其中所述抗体或其抗原结合片段包含含有HCDR1,HCDR2和HCDR3的重链可变区(VH)。
在某些实施方式中,其中所述抗体或其抗原结合片段还包含或不包含CH1结构域。
在某些实施方式中,其中所述抗体或其抗原结合片段还包含或不包含CH2和CH3结构域。
在某些实施方式中,其中所述抗体或其抗原结合片段还包含抗体重链恒定区。
在某些实施方式中,其中所述抗体重链恒定区为人抗体重链恒定区。
在某些实施方式中,其中所述人抗体重链恒定区选自源自下组的恒定区:IgG1、IgG2、IgG3、IgG4和它们的变体。
在某些实施方式中,其中所述抗体或其抗原结合片段还包含含有LCDR1,LCDR2,LCDR3的轻链可变区(VL)。
在某些实施方式中,其中所述抗体或其抗原结合片段还包含轻链恒定区(CL)。
在某些实施方式中,其中所述免疫检查点的抑制剂包含抗PD-L1抗体或其抗原结合片段,或抗PD-1抗体或其抗原结合片段。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包含HCDR1,HCDR2,HCDR3,它们各自分别与以下分子的HCDR1,HCDR2,HCDR3具有至少约80%(例如,约85%,90%或95%)的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A(RG7446)或durvalumab(MEDI4736)。
在某些实施方式中,其中所述重链可变区分别与以下分子的重链可变区具有至少约80%(例如,约85%,90%或95%)的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链,其中所述重链分别与以下分子的重链具有至少约80%(例如,约85%,90%或95%)的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段还包含抗体的轻链可变区,其中所述轻链可变区包含LCDR1,LCDR2,LCDR3,它们各自分别与以下分子的LCDR1,LCDR2,LCDR3具有至少约80%(例如,约85%,90%或95%)的序列同一性: Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab。
例如,所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区和轻链可变区,其中所述重链可变区包含HCDR1,HCDR2,HCDR3,其中所述轻链可变区包含LCDR1,LCDR2,LCDR3,它们各自分别与以下分子的HCDR1,HCDR2,HCDR3,LCDR1,LCDR2,LCDR3具有至少约80%(例如,约85%,90%或95%)的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A(RG7446)或durvalumab(MEDI4736)。
在某些实施方式中,其中所述轻链可变区分别与以下分子的轻链可变区具有至少约80%(例如,约85%,90%或95%)的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab。
例如,所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区和轻链可变区,它们各自分别与以下分子的重链可变区和轻链可变区具有至少约80%(例如,约85%,90%或95%)的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A(RG7446)或durvalumab(MEDI4736)。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段还包含抗体的轻链,其中所述轻链分别与以下分子的轻链具有至少约80%(例如,约85%,90%或95%)的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab。
例如,所述抗PD-L1抗体或其抗原结合片段包含抗体的重链和轻链,它们各自分别与以下分子的重链和轻链具有至少约80%(例如,约85%,90%或95%)的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A(RG7446)或durvalumab(MEDI4736)。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段包括:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab,或其抗原结合片段,或其变体或生物类似物,或其组合。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包括一个或多个选自下组的重链可变区CDRs(HCDRs):(a)具有与SEQ ID NO:1所示氨基酸序列至少70%序列同一性的HCDR1;(b)具有与SEQ ID NO:2至少70%序列同一性的HCDR2;和(c)具有与SEQ ID NO:3至少70%序列同一性的HCDR3。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包括一个或多个选自下组的HCDRs:(a)具有SEQ ID NO:1所示氨基酸序列的HCDR1或者通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:1所示氨基酸序列不超过2个氨基酸差异的HCDR1;(b)具有SEQ ID NO:2所示氨基 酸序列的HCDR2或者通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:2所示氨基酸序列不超过2个氨基酸差异的HCDR2;和(c)具有SEQ ID NO:3所示氨基酸序列的HCDR3或者通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:3所示氨基酸序列不超过2个氨基酸差异的HCDR3。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包含HCDR1,HCDR2,HCDR3,所述HCDR1包含与SEQ ID NO:1所示氨基酸序列至少70%序列同一性的氨基酸序列,所述HCDR2包含与SEQ ID NO:2所示氨基酸序列至少70%序列同一性的氨基酸序列,所述HCDR3包含与SEQ ID NO:3所示氨基酸序列至少70%序列同一性的氨基酸序列。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包含HCDR1,HCDR2,HCDR3,所述HCDR1包含SEQ ID NO:1所示的氨基酸序列或通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:1所示的氨基酸序列不超过2个氨基酸差异的氨基酸序列;所述HCDR2包含SEQ ID NO:2所示的氨基酸序列或通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:2所示的氨基酸序列不超过2个氨基酸差异的氨基酸序列;所述HCDR3包含SEQ ID NO:3所示的氨基酸序列或通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:3所示的氨基酸序列不超过2个氨基酸差异的氨基酸序列。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包含选自下组的氨基酸序列:(a)SEQ ID NO:4所示的氨基酸序列;(b)与SEQ ID NO:4所示的氨基酸序列至少85%,90%,95%或者99%序列同一性的氨基酸序列;和(c)通过加成、消除或取代反应所得到的与SEQ ID NO:4所示的氨基酸序列具有1个或更多差异的氨基酸序列。
在某些实施方式中,其中所述TGFβ抑制剂结合TGFβ1、TGFβ2或TGFβ3。
在某些实施方式中,其中所述TGFβ抑制剂结合TGFβ或转化生长因子β受体(TGFβR)。
在某些实施方式中,其中所述TGFβ抑制剂结合TGFβI型受体(TGFβRI),TGFβII型受体(TGFβRII)或TGFβIII型受体(TGFβRIII)。
在某些实施方式中,其中所述TGFβ抑制剂包括:1)抗TGFβ抗体或其抗原结合片段;2)抗TGFβR抗体或其抗原结合片段;3)小分子TGFβ抑制剂;4)包含转化生长因子β受体II(TGFβRII)或其功能活性片段、或其变体或生物类似物的蛋白质,或它们的组合。
在某些实施方式中,其中所述抗TGFβ抗体或其抗原结合片段包含抗体的重链可变区, 其中所述重链可变区包含HCDR1,HCDR2,HCDR3,它们各自分别与Fresolimumab分子的HCDR1,HCDR2,HCDR3具有至少约80%的序列同一性。
在某些实施方式中,其中所述重链可变区与Fresolimumab分子的重链可变区具有至少约80%的序列同一性。
在某些实施方式中,其中所述重链与Fresolimumab分子的重链具有至少约80%的序列同一性。
在某些实施方式中,其中所述抗TGFβ抗体或其抗原结合片段包含抗体的轻链可变区,其中所述轻链可变区包含LCDR1,LCDR2,LCDR3,它们各自分别与Fresolimumab分子的LCDR1,LCDR2,LCDR3具有至少约80%的序列同一性。
在某些实施方式中,其中所述轻链可变区与Fresolimumab分子的轻链可变区具有至少约80%的序列同一性。
在某些实施方式中,其中所述轻链与Fresolimumab分子的轻链具有至少约80%的序列同一性。
在某些实施方式中,其中所述抗TGFβ抗体或其抗原结合片段可以包括:Fresolimumab,或其抗原结合片段,或其变体或生物类似物。
在某些实施方式中,其中所述小分子TGFβ抑制剂包括以下化合物:SB525334、SD-208、SB431542、LY2109761、LY2157299、GW788388、RepSox、SIS3、LDN-193189、EW-7197、LY364947、或其组合。
在某些实施方式中,其中所述TGFβRII具有野生型人TGFβ受体2型同种型A序列(例如,NCBI参照序列(Ref Seq)登录号NP_001020018的氨基酸序列)的多肽,或具有野生型人TGFβ受体2型同种型B序列(例如,NCBI Ref Seq登录号NP_003233的氨基酸序列)的多肽,或与其氨基酸序列具有基本相同的序列的多肽。
在某些实施方式中,其中所述TGFβRII或其功能活性片段包括:人TGFβRII胞外域(ECD),NCBI Ref Seq登录号NP_001020018或NCBI Ref Seq登录号NP_003233的任何部分,或者与其氨基酸序列基本相同的序列。
在某些实施方式中,其中所述TGFβRII或其功能活性片段包含:
(a)SEQ ID NO:6所示的氨基酸序列;
(b)具有与SEQ ID NO:6所示的氨基酸序列至少约85%序列同一性的氨基酸序列;或
(c)具有与SEQ ID NO:6所示的氨基酸序列相比,添加,缺失和/或取代一个或多个氨基酸的氨基酸序列。
在某些实施方式中,其中所述双功能免疫检查点/TGFβ抑制剂为融合蛋白。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂或PD-1/TGFβ双重抑制剂为融合蛋白。
在某些实施方式中,其中所述PD-L1//TGFβ双重抑制剂包含PD-L1靶向部分和TGFβ受体结构域,所述TGFβ受体结构域包含转化生长因子β受体II(TGFβRII)或其功能活性片段。
在某些实施方式中,其中所述PD-L1//TGFβ双重抑制剂包含多肽,其中所述多肽至少包含:(i)抗PD-L1抗体的重链可变区;和(ii)TGFβRII或其功能活性片段。
在某些实施方式中,其中所述抗PD-L1抗体重链可变区包含HCDR1,HCDR2,HCDR3;其中所述HCDR1包含与SEQ ID NO:1所示氨基酸序列至少约70%序列同一性的氨基酸序列,所述HCDR2包含与SEQ ID NO:2所示氨基酸序列至少约70%序列同一性的氨基酸序列,所述HCDR3包含与SEQ ID NO:3所示氨基酸序列至少约70%序列同一性的氨基酸序列;或
其中所述HCDR1,HCDR2,HCDR3分别与以下分子的HCDR1,HCDR2,HCDR3具有至少约80%,约85%,90%,95%或者99%的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A(RG7446)或durvalumab(MEDI-4736)。
在某些实施方式中,其中所述抗PD-L1抗体重链可变区包含HCDR1,HCDR2,HCDR3,所述HCDR1包含SEQ ID NO:1所示的氨基酸序列或通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:1所示的氨基酸序列不超过2个氨基酸差异的氨基酸序列;所述HCDR2包含SEQ ID NO:2所示的氨基酸序列或通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:2所示的氨基酸序列不超过2个氨基酸差异的氨基酸序列;所述HCDR3包含SEQ ID NO:3所示的氨基酸序列或通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:3所示的氨基酸序列不超过2个氨基酸差异的氨基酸序列。
在某些实施方式中,其中所述抗PD-L1抗体重链可变区包含选自下组的氨基酸序列:(a)SEQ ID NO:4所示的氨基酸序列;(b)与SEQ ID NO:4所示的氨基酸序列至少约85%,90%,95%或者99%序列同一性的氨基酸序列;(c)通过加成、消除或取代反应所得到的与SEQ ID NO:4所示的氨基酸序列具有1个或更多差异的氨基酸序列;和(d)与以下分子的重链可变区具有至少约80%的序列同一性的氨基酸序列:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab。
在某些实施方式中,其中所述TGFβRII或其功能活性片段包含:
(a)SEQ ID NO:6所示的氨基酸序列;
(b)具有与SEQ ID NO:6所示的氨基酸序列至少约85%序列同一性的氨基酸序列;或
(c)具有与SEQ ID NO:6所示的氨基酸序列相比,添加,缺失和/或取代一个或多个氨基酸的氨基酸序列。
在某些实施方式中,其中所述多肽还包含连接子,所述连接子将所述抗PD-L1的抗体或其抗原结合片段的重链可变区的C末端与所述TGFβRII或其功能活性片段的N末端相连接。
在某些实施方式中,所述多肽还包括CH2、CH3结构域,所述多肽自N末端至C末端依次包含抗PD-L1抗体的重链可变区(VH)、CH2、CH3结构域和TGFβRII或其功能活性片段,所述CH3结构域的C末端与所述TGFβRII或其功能活性片段N末端通过连接子相连接。
在某些实施方式中,其中所述CH2、CH3结构域源自IgG。例如,所述CH2、CH3结构域均源自IgG1、IgG2、IgG3或IgG4。
在某些实施方式中,其中所述连接子为肽连接子。
在某些实施方式中,其中所述肽连接子的氨基酸序列为(G 4S) x,其中x为3-6中的任意整数。
在某些实施方式中,其中所述肽连接子包含选自下组氨基酸序列:(a)SEQ ID NO:5所示的氨基酸序列;(b)具有与SEQ ID NO:5所示的氨基酸序列至少约85%,90%,95%或者99%序列同一性的氨基酸序列;和(c)通过加成、消除或取代反应所得到的与SEQ ID NO:5所示的氨基酸序列具有1个或更多差异的氨基酸序列。
在某些实施方式中,其中所述多肽包含选自下组的氨基酸序列:(a)SEQ ID NO:7所示的氨基酸序列;(b)具有与SEQ ID NO:7所示的氨基酸序列至少约85%,90%,95%或者99%序列同一性的氨基酸序列;(c)通过加成、消除或取代反应所得到的与SEQ ID NO:7所示的氨基酸序列具有1个或更多差异的氨基酸序列。
在某些实施方式中,其中所述PD-L1//TGFβ双重抑制剂包含两条前述多肽。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂包含第一多肽和第二多肽,其中所述第一多肽至少包含:(i)抗PD-L1抗体的重链可变区;和(ii)TGFβRII或其功能活性片段;其中所述第二多肽至少包含抗PD-L1抗体的轻链可变区;其中所述第一多肽的重链可变区和所述第二多肽的轻链可变区在组合时能够特异性结合PD-L1。
在某些实施方式中,其中所述第一多肽还包含连接子,所述连接子将所述抗PD-L1的 抗体或其抗原结合片段的重链可变区的C末端与所述TGFβRII或其功能活性片段的N末端相连接。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂包含第一多肽和第二多肽,其中:
所述第一多肽自N末端至C末端依次包含抗PD-L1抗体的重链可变区(VH)、CH1结构域以及TGFβRII或其功能活性片段,所述CH1结构域的C末端与所述TGFβRII或其功能活性片段N末端通过连接子相连接;和所述第二多肽自N末端至C末端包含抗PD-L1抗体的轻链可变区(VL)和轻链恒定区(CL)。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂包含第一多肽和第二多肽,其中:
所述第一多肽自N末端至C末端依次包含抗PD-L1抗体的重链可变区(VH)、CH1、CH2、CH3结构域和TGFβRII或其功能活性片段,所述CH3结构域的C末端与所述TGFβRII或其功能活性片段N末端通过连接子相连接;和所述第二多肽自N末端至C末端包含抗PD-L1抗体的轻链可变区(VL)、轻链恒定区(CL)。
在某些实施方式中,其中所述连接子为肽连接子。
在某些实施方式中,其中所述第一多肽与以下分子的具有相同功能的多肽具有至少约80%序列同一性:M7824或SHR-1701。
在某些实施方式中,其中所述第二多肽与以下分子的具有相同功能的多肽具有至少约80%序列同一性:M7824或SHR-1701。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂包括M7824、SHR-1701,或其变体或生物类似物,或其组合。
在某些实施方式中,其中所述PI3K抑制剂为PI3Kδ/γ双重抑制剂。
在某些实施方式中,其中所述PI3K抑制剂包括式(IA-I)、(IA-II)、(IA-III)、或(IA-IV)所示的化合物:
Figure PCTCN2022075642-appb-000001
或它们的对映异构体、对映异构体的混合物、两种或更多种非对映异构体的混合物、同位素变体、药学上可接受的盐、溶剂化物、水合物或前药,其中
每次出现的R独立地选自氢、卤素、-OR a、CN、经取代或未经取代的C 1-6烷基、经取代或未经取代的C 2-6烯基、经取代或未经取代的C 2-6炔基、经取代或未经取代的C 3-8环烷基和经取代或未经取代的杂环基团;
R 1和R 2可相同或不同并且独立地选自氢、卤素和经取代或未经取代的C 1-6烷基,或与共有原子直接结合的R 1和R 2可连接形成=O基或经取代或未经取代的饱和或不饱和3-10元环,包括与R 1和R 2结合的碳原子,所述环可任选地包括一个或多个相同或不同且选自O、NR a和S的杂原子;
Cy 1为选自经取代或未经取代的C 3-8环烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环基团、经取代或未经取代的C6-20芳基和经取代或未经取代的具有一个或多个选自N、0和S的杂原子的5至14元杂芳基的单环基团;
每次出现的R a可相同或不同且独立地选自氢、卤素、羟基、氰基、经取代或未经取代的C 1-6烷基;-NR cR d,其中R c,R d独立地选自氢、卤素、羟基、氰基、经取代或未经取代的C 1-6烷基和C 1-6烷氧基,和-OR c,其中R c为经取代或未经取代的C 1-6烷基;
n为1-4的整数;并且
q为0、1或2;
每次出现的X独立地选自CR 3或N;
并且每次出现的R 3独立地选自氢、羟基、卤素、羧基、氰基、硝基、经取代或未经取代的C 1-8烷基、经取代或未经取代的C 1-8烷氧基、经取代或未经取代的C 2-10烯基、经取代或未经取代的C 2-12炔基、经取代或未经取代的C 6-20芳基、经取代或未经取代的C 6-20芳基C 1-8烷基、经取代或未经取代的C 3-20环烷基、经取代或未经取代的C 3-20环烷基C 1-8烷基、经取代或未经取代的C 3-8环烯基C 1-8烷基、经取代或未经取代的C 3-8环烯基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基C 1-8烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环基C 1-8烷基环、经取代或未经取代的胍、-C00R x、-C(O)R x、-C(S)R x、-C(O)NR xR y、-C(O)ONR xR y,-NR yR z、-NR xCONR yR z,-N(R x)SOR y、-N(Rx)SO 2R y、-(=N-N(R x)R y)、-NR xC(O)OR y、-NR xR y、-NR xC(O)R y-、-NR xC(S)R y、-NR xC(S)NR yR z、-SONR xR y-、-SO 2NR xR y-、-OR x、-OR xC(O)NR yR z、- OR xC(O)OR y-、-OC(O)R x、-OC(O)NR xR y、-R xNR yC(O)R z、-R xOR y、-R xC(O)OR y、-R xC(O)NR yR z、-R xC(O)R x、-R xOC(O)R x、-SR x、-SOR x、-SO 2R x和-ONO 2,其中以上每个基团中的R x、R y和R z可为氢、经取代或未经取代的C 1-8烷基、经取代或未经取代的C 1-8烷氧基、经取代或未经取代的C 2-10烯基、经取代或未经取代的C 2-12炔基、经取代或未经取代的C 6-20芳基、经取代或未经取代的C 6-20芳基C 1-8烷基、经取代或未经取代的C 3-20环烷基、经取代或未经取代的C 3-20环烷基C 1-8烷基、经取代或未经取代的C 3-8环烯基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基C 1-8烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环、经取代或未经取代的具有至少一个选自N、0和S的杂原子的3至15元杂环基C 1-8烷基环或经取代或未经取代的氨基,或R x、R y和R z的任两个可连接形成经取代或未经取代的饱和或不饱和3-10元环,所述环可任选地包括可能相同或不同且选自O、NR x或S的杂原子,其中R x为氢或经取代或未经取代的烷基。
在某些实施方式中,其中所述PI3K抑制剂包括式(IA-V)所示的化合物:
Figure PCTCN2022075642-appb-000002
或其对映异构体、对映异构体的混合物、两种或更多种非对映异构体的混合物、或同位素变体;或其药学上可接受的盐、溶剂化物、水合物或前药,其中
每次出现的R独立地选自氢、卤素、-OR a、CN、经取代或未经取代的C 1-6烷基、经取代或未经取代的C 2-6烯基、经取代或未经取代的C 2-6炔基、经取代或未经取代的C 3-8环烷基和经取代或未经取代的杂环基团;R 1和R 2可相同或不同并且独立地选自氢、卤素和经取代或未经取代的C 1-6烷基,或与共有原子直接结合的R 1和R 2可连接形成=O基或经取代或未经取代的饱和或不饱和3-10元环,包括与R 1和R 2结合的碳原子,所述环可任选地包括一个或多个相同或不同且选自O、NR a和S的杂原子;
每次出现的X独立地选自CR 3或N;并且
每次出现的R 3独立地选自氢、羟基、卤素、羧基、氰基、硝基、经取代或未经取代的 C 1-8烷基、经取代或未经取代的C 1-8烷氧基、经取代或未经取代的C 2-10烯基、经取代或未经取代的C 2-12炔基、经取代或未经取代的C 6-20芳基、经取代或未经取代的C 6-20芳基C 1-8烷基、经取代或未经取代的C 3-20环烷基、经取代或未经取代的C 3-20环烷基C 1-8烷基、经取代或未经取代的C 3-8环烯基C 1-8烷基、经取代或未经取代的C 3-8环烯基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基C 1-8烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环基C 1-8烷基环、经取代或未经取代的胍、-C00R x、-C(O)R x、-C(S)R x、-C(O)NR xR y、-C(O)ONR xR y,-NR yR z、-NR xCONR yR z,-N(R x)SOR y、-N(Rx)SO 2R y、-(=N-N(R x)R y)、-NR xC(O)OR y、-NR xR y、-NR xC(O)R y-、-NR xC(S)R y、-NR xC(S)NR yR z、-SONR xR y-、-SO 2NR xR y-、-OR x、-OR xC(O)NR yR z、-OR xC(O)OR y-、-OC(O)R x、-OC(O)NR xR y、-R xNR yC(O)R z、-R xOR y、-R xC(O)OR y、-R xC(O)NR yR z、-R xC(O)R x、-R xOC(O)R x、-SR x、-SOR x、-SO 2R x和-ONO 2,其中以上每个基团中的R x、R y和R z可为氢、经取代或未经取代的C 1-8烷基、经取代或未经取代的C 1-8烷氧基、经取代或未经取代的C 2-10烯基、经取代或未经取代的C 2-12炔基、经取代或未经取代的C 6-20芳基、经取代或未经取代的C 6-20芳基C 1-8烷基、经取代或未经取代的C 3-20环烷基、经取代或未经取代的C 3-20环烷基C 1-8烷基、经取代或未经取代的C 3-8环烯基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基C 1-8烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环、经取代或未经取代的具有至少一个选自N、0和S的杂原子的3至15元杂环基C 1-8烷基环或经取代或未经取代的氨基,或R x、R y和R z的任两个可连接形成经取代或未经取代的饱和或不饱和3-10元环,所述环可任选地包括可能相同或不同且选自O、NR x或S的杂原子,其中R x为氢或经取代或未经取代的烷基;
每次出现的R 5为氢、C 1-6烷基或卤素;
n为0、1、2、3或4;并且P为0、1、2、3、4或5。
在某些实施方式中,其中所述PI3K抑制剂包括式(IA-VI)所示的化合物:
Figure PCTCN2022075642-appb-000003
或其对映异构体、对映异构体的混合物、两种或更多种非对映异构体的混合物、或同位素变体;或其药学上可接受的盐、溶剂化物、水合物或前药,其中
每次出现的R独立地选自氢、卤素、-OR a、CN、经取代或未经取代的C 1-6烷基、经取代或未经取代的C 2-6烯基、经取代或未经取代的C 2-6炔基、经取代或未经取代的C 3-8环烷基和经取代或未经取代的杂环基团;
R 1和R 2可相同或不同并且独立地选自氢、卤素和经取代或未经取代的C 1-6烷基,或与共有原子直接结合的R 1和R 2可连接形成=O基或经取代或未经取代的饱和或不饱和3-10元环,包括与R 1和R 2结合的碳原子,所述环可任选地包括一个或多个相同或不同且选自O、NR a和S的杂原子;
每次出现的X独立地选自CR 3或N;并且
每次出现的R 3独立地选自氢、羟基、卤素、羧基、氰基、硝基、经取代或未经取代的C 1-8烷基、经取代或未经取代的C 1-8烷氧基、经取代或未经取代的C 2-10烯基、经取代或未经取代的C 2-12炔基、经取代或未经取代的C 6-20芳基、经取代或未经取代的C 6-20芳基C 1-8烷基、经取代或未经取代的C 3-20环烷基、经取代或未经取代的C 3-20环烷基C 1-8烷基、经取代或未经取代的C 3-8环烯基C 1-8烷基、经取代或未经取代的C 3-8环烯基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基C 1-8烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环基C 1-8烷基环、经取代或未经取代的胍、-C00R x、-C(O)R x、-C(S)R x、-C(O)NR xR y、-C(O)ONR xR y,-NR yR z、-NR xCONR yR z,-N(R x)SOR y、-N(Rx)SO 2R y、-(=N-N(R x)R y)、-NR xC(O)OR y、-NR xR y、-NR xC(O)R y-、-NR xC(S)R y、-NR xC(S)NR yR z、-SONR xR y-、-SO 2NR xR y-、-OR x、-OR xC(O)NR yR z、-OR xC(O)OR y-、-OC(O)R x、-OC(O)NR xR y、-R xNR yC(O)R z、-R xOR y、-R xC(O)OR y、-R xC(O)NR yR z、-R xC(O)R x、-R xOC(O)R x、-SR x、-SOR x、-SO 2R x和-ONO 2,其中以上每个基团中的R x、R y和 R z可为氢、经取代或未经取代的C 1-8烷基、经取代或未经取代的C 1-8烷氧基、经取代或未经取代的C 2-10烯基、经取代或未经取代的C 2-12炔基、经取代或未经取代的C 6-20芳基、经取代或未经取代的C 6-20芳基C 1-8烷基、经取代或未经取代的C 3-20环烷基、经取代或未经取代的C 3-20环烷基C 1-8烷基、经取代或未经取代的C 3-8环烯基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基C 1-8烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环、经取代或未经取代的具有至少一个选自N、0和S的杂原子的3至15元杂环基C 1-8烷基环或经取代或未经取代的氨基,或R x、R y和R z的任两个可连接形成经取代或未经取代的饱和或不饱和3-10元环,所述环可任选地包括可能相同或不同且选自O、NR x或S的杂原子,其中R x为氢或经取代或未经取代的烷基;
每次出现的R 5为氢、C 1-6烷基或卤素;
n为0、1、2、3或4;并且
P为0、1、2、3、4或5。
在某些实施方式中,其中R为氢、卤素、经取代或未经取代的C 1-6烷基或OR a
在某些实施方式中,其中所述R a为烷基。
在某些实施方式中,其中所述Cy 1选自:
Figure PCTCN2022075642-appb-000004
在某些实施方式中,其中所述R 1和R 2各自独立地表示氢或经取代或未经取代的C 1-6烷基。
在某些实施方式中,其中所述R 3为碘代、氰基、经取代或未经取代的烷基、经取代或未经取代的炔基、经取代或未经取代的芳基、经取代或未经取代的杂芳基。
在某些实施方式中,其中X为CR 3并且每次出现的R 3独立为氢、卤素、羟基或NH 2
在某些实施方式中,其中所述R 3为氢。
在某些实施方式中,其中所述PI3K抑制剂包括:
2-[(6-氨基-9H-嘌呤-9-基)甲基]-6-溴-3-苯基-4H-色烯-4-酮;
2-[(6-氨基-9H-嘌呤-9-基)甲基]-3-苯基-4H-色烯-4-酮;
2-((9H-嘌呤-6-基硫代)甲基)-3-苯基-4H-色烯-4-酮;
2-[(9H-嘌呤-6-基硫代)甲基]-6-溴-3-苯基-4H-色烯-4-酮;
2-((4-氨基-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-溴-3-苯基-4!1-色烯-4-酮;
2-[(6-氨基-9H-嘌呤-9-基)甲基]-6-溴-3-(4-氟苯基)-4H-色烯-4-酮;
2-[(6-氨基-9H-嘌呤-9-基)甲基]-3-(4-氟苯基)-4H-色烯-4-酮;
2-[(6-氨基-9H-嘌呤-9-基)甲基]-6-溴-3-邻甲苯基-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-6-溴-3-苯基-4H-色烯-4-酮;
2-(1-(9H-嘌呤-6-基硫代)乙基)-6-溴-3-苯基-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-3-苯基-4H-色烯-4-酮;
(S)-2-(1-(9H-嘌呤-6-基氨基)乙基)-6-溴-3-苯基-4H-色烯-4-酮;
2-((9H-嘌呤-6-基氨基)甲基)-6-溴-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-溴-3-苯基-4H-色烯-4-酮;
2-((6-氨基-9H-嘌呤-9-基)甲基)-6-甲氧基-3-苯基-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-6-溴-3-(2-氟苯基)-4H-色烯-4-酮;
2-((6-氨基-9H-嘌呤-9-基)甲基)-6-溴-3-(2-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)丙基)-3-苯基-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((6-氨基-9H-嘌呤-9-基)甲基)-3-(2-氟苯基)-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-3-(2-氟苯基)-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)丙基)-3-(2-氟苯基)-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)丙基)-3-(4-氟苯基)-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)丙基)-6-氟-3-苯基-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-3-(4-氟苯基)-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-6-氟-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
2-((9H-嘌呤-6-基氨基)甲基)-3-苯基-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-3-邻甲苯基-4H-色烯-4-酮;
2-((9H-嘌呤-6-基氨基)甲基)-3-(2-氟苯基)-4H-色烯-4-酮;
2-((9H-嘌呤-6-基氨基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
(S)-2-(1-(9H-嘌呤-6-基氨基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-6-氟-3-(2-氟苯基)-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-3-(3,5-二氟苯基)-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
(R)-2-(1-(9H-嘌呤-6-基氨基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
(S)-2-(1-(9H-嘌呤-6-基氨基)乙基)-6-氟-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-氟-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-氟-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(吡啶-3-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(3-羟基丙-1-炔基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(3-(羟基甲基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(1H-吲唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(3-氟苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(3-羟基丙基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
N-(3-(4-氨基-1-((4-氧代-3-苯基-4H-色烯-2-基)甲基)-1H-吡唑并[3,4-d]嘧啶-3-基)苯基)乙酰胺;
2-((4-氨基-3-(3-氟-5-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(3-氟-5-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(3-氟-5-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3-氟-5-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-羟基-3-甲基丁-1-炔基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
2-(1_(4-氨基-3-(1H-吡唑-4-基)-1H-吡唑并[3,4-(1]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
(S)-2-(1-(9H-嘌呤-6-基氨基)乙基)-3-苯基-4H-色烯-4-酮;
(S)-2-(1-(9H-嘌呤-6-基氨基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-氟-5-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-氟-5-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(1H-吲唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3,5-二甲基-1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2-(羟基甲基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-氟-3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-氟-3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-羟基丙-1-炔基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-氟-4-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-氟-4-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-氯-5-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-氯-5-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-(三氟甲氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((6-氨基-9H-嘌呤-9-基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-氟-2-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-氟-2-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3-氨基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2-氨基嘧啶-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(1H-吲哚-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-氯-3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-氯-3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2-氯-5-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2-氯-5-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3,4-二甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3,4-二羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(1H-吲哚-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲基-1H-吲哚-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
叔丁基-(5-(4-氨基-1-(1-(3-(3-氟苯基)-4-氧代-4H-色烯-2-基)乙基)-1H-吡唑并[3,4-d]嘧啶-3-基)噻吩-2-基)氨基甲酸甲酯;
2-(1-(4-氨基-3-(5-(氨基甲基)噻吩-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-氟-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-氟-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲基-1H-吲唑-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
N-(4-(4-氨基-1-(1-(3-(3-氟苯基)-4-氧代-4H-色烯-2-基)乙基)-1H-吡唑并[3,4-d]嘧啶-3-基)苯基)乙酰胺;
2-(1-(4-氨基-3-(4-氨基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2,3-二氢苯并呋喃-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-乙基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲基-1H-吲哚-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2-甲氧基嘧啶-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
4-(4-氨基-1-(1-(3-(3-氟苯基)-4-氧代-4H-色烯-2-基)乙基)-1H-吡唑并[3,4-d]嘧啶-3-基)噻吩-2-甲醛;
2-(1-(4-氨基-3-(5-(羟基甲基)噻吩-3-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2-甲基-1H-苯并[d]咪唑-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲基-1H-吲哚-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((6-氨基-9H-嘌呤-9-基)甲基)-6-氟-3-苯基-4H-色烯-4-酮;2-((6-氨基-9H-嘌呤-9-基)甲基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3-氟-5-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3-氟-5-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-((9H-嘌呤-6-基氨基)甲基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-((9H-嘌呤-6-基氨基)甲基)-6-氟-3-苯基-4H-色烯-4-酮;(R)-2-(1-(9H-嘌呤-6-基氨基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-氟-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3,5-二氟-4-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3,5-二氟-4-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3,5-二氟-4-甲氧基苯基)-1H-吡唑并[3,4-(1]嘧啶-1-基)甲基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3,5-二氟-4-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
(+)-2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
(-)-2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3,5-二甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-甲氧基-3,5-二甲基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2-氟-5-异丙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H- 色烯-4-酮;
2-(1-(4-氨基-3-(2,3-二氢苯并[b][1,4]二噁英-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(1-苄基-1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2-甲基吡啶-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3,4-二氢-2H-苯并[b][1,4]二氧杂卓-7-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(6-吗啉基吡啶-3-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(二苯并[b,d]呋喃-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-(苄氧基)-3-氯苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-氯-4-异丙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-(二甲基氨基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-乙氧基-3-氟苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-异丙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-(三氟甲氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(3-(4-乙酰基苯基)-4-氨基-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-(苄氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯- 4-酮;
2-(1-(4-氨基-3-(4-(二甲基氨基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-(甲磺酰基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-乙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(苯并[b]噻吩-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(5-氯噻吩-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3,5-二甲基异噁唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-丙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2~(1-(4-氨基-3-(呋喃-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-乙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-氯-4-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-氟-4-异丙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(6-氟吡啶-3-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(嘧啶-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-(甲氧基甲基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(6-羟基萘-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯- 4-酮;
2-(1-(4-氨基-3-(3-异丙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(1-甲基-1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(1,3-二甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2,3-二甲基-2H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(6-甲氧基萘-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(苯并[b]噻吩-3-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2,4-二甲氧基嘧啶-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(6-乙氧基萘-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
3-(4-氨基-1-(1-(3-(3-氟苯基)-4-氧代-4H-色烯-2-基)乙基)-1H-吡唑并[3,4-d]嘧啶-3-基)-N-环丙基苯甲酰胺;
2-(1-(4-氨基-3-(3-(吗啉-4-羰基)苯基)-1H-吡唑并[3,4-(1]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-(二氟甲氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
5-(4-氨基-1-(1-(3-(3-氟苯基)-4-氧代-4H-色烯-2-基)乙基)-1H-吡唑并[3,4-d]嘧啶-3-基)呋喃-2-甲醛;
和/或它们的对映异构体、对映异构体的混合物、两种或更多种非对映异构体的混合物、同位素变体、药学上可接受的盐、溶剂化物、水合物或前药。
在某些实施方式中,其中所述PI3K抑制剂包括:2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮,或其对映异构体、对映异构体的混合物、两种或更多种非对映异构体的混合物、或同位素变体;或其药学上可接受的盐、溶剂化物、水合物或前药。
在某些实施方式中,其中所述PI3K抑制剂为(S)-2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮或其药学上可接受的盐、溶剂化物、水合物或前药。
在某些实施方式中,所述药物组合包括:包括PD-L1/TGFβ双重抑制剂和PI3K抑制剂。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂包含抗PD-L1抗体或其抗原结合片段和TGFβRII或其功能活性片段。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂包含两条多肽,其中所述多肽包含选自下组的氨基酸序列:(a)SEQ ID NO:7所示的氨基酸序列;(b)具有与SEQ ID NO:7所示的氨基酸序列至少85%,90%,95%或者99%序列同一性的氨基酸序列;(c)通过加成、消除或取代反应所得到的与SEQ ID NO:7所示的氨基酸序列具有1个或更多差异的氨基酸序列。
在某些实施方式中,其中所述PI3K抑制剂为PI3Kδ/γ双重抑制剂。
在某些实施方式中,其中所述PI3K抑制剂为(S)-2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮或其药学上可接受的盐、溶剂化物、水合物或前药。
在某些实施方式中,所述PD-L1/TGFβ双重抑制剂与所述PI3K抑制剂在所述药物组合中不互相混合。
在某些实施方式中,所述PD-L1/TGFβ双重抑制剂与所述PI3K抑制剂各自独立地存在于单独的容器中。
在某些实施方式中,所述的药物组合还可以进一步地包括第三活性物质。
在某些实施方式中,所述的第三活性物质可以选自长春新碱、长春碱、长春地辛、依托泊苷、多西他赛、紫杉醇、伊立替康、长春瑞滨、米托蒽醌、长春氟宁、拓扑替康中的任意一种或其任意地组合。
另一方面,本申请提供一种药物组合物,所述药物组合包括前述的PI3K抑制剂和第二治疗剂,以及任选的一种或多种药学上可接受的载体或赋形剂。
在某些实施方式中,其中所述第二治疗剂为前述的PD-L1/TGFβ双重抑制剂。
在某些实施方式中,其中所述PI3K抑制剂为前述的PI3Kδ/γ双重抑制剂。
在某些实施方式中,其中所述第二治疗剂与所述PI3K抑制剂存在于单一或分开的剂型中。
另一方面,本申请提供一种治疗和/或预防肿瘤的方法,其包括向有需要的受试者施用:有效量的前述的药物组合或前述的药物组合物。
在某些实施方式中,所述方法包括向有需要的受试者施用:有效量的前述的PI3K抑制剂和前述的免疫检查点的抑制剂。在某些实施方式中,所述方法包括向有需要的受试者施用:有效量的前述的PI3K抑制剂和前述的TGFβ抑制剂。在某些实施方式中,所述方法包括向有需要的受试者施用:有效量的前述的PI3K抑制剂、前述的TGFβ抑制剂和前述的免疫检查点的抑制剂。
在某些实施方式中,所述方法包括向有需要的受试者施用:有效量的前述的PI3K抑制剂和前述的PD-L1/TGFβ双重抑制剂。
在某些实施方式中,其中所述PI3K抑制剂与所述PD-L1/TGFβ双重抑制剂同时施用。
在某些实施方式中,其中所述PI3K抑制剂在所述PD-L1/TGFβ双重抑制剂之后施用。
在某些实施方式中,其中所述PI3K抑制剂在所述PD-L1/TGFβ双重抑制剂之前施用。
在某些实施方式中,还包括向有需要的受试者施用有效量的前述的小分子TGFβ抑制剂。
在某些实施方式中,其中所述肿瘤包括实体瘤和非实体肿瘤。
在某些实施方式中,其中所述肿瘤包括PI3K表达异常的肿瘤。
在某些实施方式中,其中所述肿瘤包括PD-L1表达异常的肿瘤。
在某些实施方式中,其中所述肿瘤包括TGFβ表达异常的肿瘤。
在某些实施方式中,所述肿瘤包括消化道肿瘤、黑素瘤或淋巴瘤。
在某些实施方式中,其中所述肿瘤对所述PI3K抑制剂的抗性延迟。
在某些实施方式中,其中所述受试者中的肿瘤的缓解延长。
在某些实施方式中,其中所述受试者经历肿瘤的完全缓解。
在某些实施方式中,其中最小残留疾病(MRD)的水平降低。
另一方面,本申请提供了前述的药物组合或前述的药物组合物在制备用于治疗和/或预防肿瘤的药物中的用途。
例如,前述的PI3K抑制剂和前述的免疫检查点的抑制剂在制备用于治疗和/或预防肿瘤的药物中的用途。例如,前述的PI3K抑制剂和前述的TGFβ抑制剂在制备用于治疗和/或预防肿瘤的药物中的用途。例如,前述的PI3K抑制剂、前述的免疫检查点的抑制剂和前述的TGFβ抑制剂在制备用于治疗和/或预防肿瘤的药物中的用途。
另一方面,本申请提供了前述的药物组合或前述的药物组合物用于治疗和/或预防肿瘤。
另一方面,本申请提供了一种抑制肿瘤生长的方法,所述方法包括使肿瘤接触前述的药物组合或前述的药物组合物。
在某些实施方式中,所述接触处于体外或离体环境中。
在某些实施方式中,所述方法包括使肿瘤接触前述的PI3K抑制剂和前述的免疫检查点的抑制剂。在某些实施方式中,所述方法包括使肿瘤接触前述的PI3K抑制剂和前述的TGFβ抑制剂。在某些实施方式中,所述方法包括使肿瘤接触前述的PI3K抑制剂、前述的免疫检查点的抑制剂和前述的TGFβ抑制剂。
另一方面,本申请提供了一种抑制肿瘤生长的方法,所述方法包括使肿瘤接触前述的PI3K抑制剂和前述的PD-L1/TGFβ双重抑制剂。
另一方面,本申请提供了一种药盒,其包括:(1)第一容器,以及位于所述第一容器中的前述的PI3K抑制剂;(2)第二容器,以及位于所述第二容器中的前述的PD-L1/TGFβ双重抑制剂。
另一方面,本申请提供了一种药盒,其可以包括:(1)第一容器,以及位于所述第一容器中的前述的PI3K抑制剂;(2)第二容器,以及位于所述第二容器中的前述的免疫检查点的抑制剂或前述的TGFβ抑制剂。
在某些实施方式中,所述药盒还包括(3)第三容器,以及位于第三容器中的前述的免疫检查点的抑制剂或前述的TGFβ抑制剂,所述第三容器中的药剂与第二容器中的药剂不同。
在某些实施方式中,所述药盒中药物的剂型为口服剂型或注射剂型。
在某些实施方式中,所述的药盒还含有说明书。
本领域技术人员能够从下文的详细描述中容易地洞察到本申请的其它方面和优势。下文的详细描述中仅显示和描述了本申请的示例性实施方式。如本领域技术人员将认识到的,本申请的内容使得本领域技术人员能够对所公开的具体实施方式进行改动而不脱离本申请所涉及发明的精神和范围。相应地,本申请的附图和说明书中的描述仅仅是示例性的,而非为限制性的。
附图说明
本申请所涉及的发明的具体特征如所附权利要求书所显示。通过参考下文中详细描述的示例性实施方式和附图能够更好地理解本申请所涉及发明的特点和优势。对附图简要说明如下:
图1A-1F显示了CN401(Tenalisib)(150mg/kg)与CN202(WBP1126)(5mg/kg)在A20小鼠B细胞淋巴瘤动物模型中联合用药结果。
图2A-2F显示了CN401(Tenalisib)(150mg/kg)与CN202(WBP1126)(15mg/kg)在A20小鼠B细胞淋巴瘤动物模型中联合用药结果。
图3A-B显示了CN401(Tenalisib)(150mg/kg)与CN202(WBP1126)(5mg/kg)/(15mg/kg)在A20小鼠B细胞淋巴瘤动物模型中联合用药结果。
图4A-4F显示了CN202(WBP1126)(5mg/kg)与CN401(Tenalisib)(150mg/kg)在A20小鼠B细胞淋巴瘤动物模型中联合用药结果。
图5A-5F显示了CN202(WBP1126)(5mg/kg)与Alpelisib(50mg/kg)在A20小鼠B细胞淋巴瘤动物模型中联合用药结果。
图6A-6F显示了CN401(Tenalisib)(150mg/kg)与抗PD-L1抗体(Atezolizumab)(5mg/kg)在A20小鼠B细胞淋巴瘤动物模型中联合用药结果。
图7A-7B显示了CN401(Tenalisib)(150mg/kg)、CN202(WBP1126)(15mg/kg)与白蛋白紫杉醇(nab-pac)(10mg/kg)在小鼠EMT-60(小鼠乳腺癌细胞)的肿瘤生长曲线和体重生长曲线。
图8A-8G显示了不同给药组中小鼠EMT-60(小鼠乳腺癌细胞)的肿瘤生长曲线。
具体实施方式
以下由特定的具体实施例说明本申请发明的实施方式,熟悉此技术的人士可由本说明书所公开的内容容易地了解本申请发明的其他优点及效果。
术语定义
在本申请中,“磷酸肌醇3-激酶(PI3K)抑制剂”或“PI3K抑制剂”通常是指任何PI3K的抑制剂。PI3K是细胞内脂质激酶的独特且保守的家族的成员,其使磷脂酰肌醇或磷酸肌醇上的3’-OH基团磷酸化。PI3K家族包括具有不同底物特异性、表达模式和调节模式的激酶(参见,例如,Katso等人,2001,Annu.Rev.Cell Dev.Biol.17,615-675;Foster,F.M.等人,2003,J Cell Sci 116,3037-3040)。I类PI3K(例如,p110α、p110β、p110γ和p110δ通常由酪氨酸激酶或G-蛋白偶联受体活化以产生PIP3,PIP3结合下游介质,如Akt/PDK1途径中的那些、mTOR、Tec家族激酶和Rho家族GTP酶。II类PI3K(例如,PI3K-C2α、PI3K-C2β、PI3K-C2γ)和III类PI3K(例如,Vps34)通过合成PI(3)P和PI(3,4)P2在细胞内运输中起关键作用。本申请公开了具体的示例性PI3K抑制剂。例如,PI3K抑制剂抑制PI3K-α、PI3K-β、PI3K-γ和PI3K-δ亚型或其组合。
例如,PI3K抑制剂可以是PI3Kδ/γ双重抑制剂Tenalisib,2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮,其结构式如下:
Figure PCTCN2022075642-appb-000005
例如,PI3K抑制剂可以是PI3K抑制剂Alpelisib,(2S)-1-N-[4-甲基-5-[2-(1,1,1-三氟-2-甲基丙烷-2-基)吡啶-4-基]-1,3-噻唑-2-基]吡咯烷-1,2-二甲酰胺,其结构式如下:
Figure PCTCN2022075642-appb-000006
在一些实施方式中,PI3K抑制剂通常是指抑制一种或多种PI3K亚型的化合物,其IC50小于约1000nM、小于约900nM、小于约800nM、小于约700nM、小于约600nM、小于约500nM、小于约400nM、小于约300nM、小于约200nM、小于约100nM、小于约75nM、小于约50nM、小于约25nM、小于约20nM、小于约15nM、小于约10nM、小于约10nM、小于约5nM或小于约1nM。
在本申请中,术语“烷基”通常是指仅由碳原子和氢原子组成,不含不饱和度,具有1-8个碳原子并且与分子剩余部分经单键连接的直链或支链烃链基,例如甲基、乙基、正丙基、1-甲基乙基(异丙基)、正丁基、正戊基和1,1-二甲基乙基(叔丁基)。术语“(C 1-6)烷基”指以上定义的具有多达6个碳原子的烷基基团。
在本申请中,术语“烯基”通常是指含有碳-碳双键并且可能为具有约2至约10个碳原子的直链或支链的脂肪烃基团,例如乙烯基、1-丙烯基、2-丙烯基(烯丙基)、异丙烯基、2-甲基-1-丙烯基、1-丁烯基和2-丁烯基。术语“(C 2- 6)烯基”指如以上定义具有多达6个碳原子的烯基基团。
在本申请中,术语“炔基”通常是指具有至少一个碳-碳三键并且具有2至12个碳原子的直链或支链烃基(当前优选的是具有2至10个碳原子的基团),例如乙炔基、丙炔基和丁炔基。术语“(C 2-6)炔基”指如以上定义具有多达6个碳原子的炔基基团。
在本申请中,术语“烷氧基”通常是指如以上定义通过氧键与分子的其余部分连接的烷基、环烷基、环烷基烷基基团。术语“经取代的烷氧基”指其中烷基组成经取代(即,-O-(经取代的烷基))的烷氧基基团,其中术语“经取代的烷基”与以上对于“烷基”定义的相同。例如,“烷 氧基”指基团-O-烷基,包括为直链、支链、环状构型的1-8个碳原子及其通过氧与母结构连接的组合。实例包括甲氧基、乙氧基、丙氧基、异丙氧基、环丙氧基和环己氧基。
在本申请中,术语“环烷基”通常是指约3-12个碳原子的非芳香族单环或多环环系,例如环丙基、环丁基、环戊基和环己基。多环环烷基的实例包括全氢萘基、金刚烷基和降莰烷基基团、桥环基团和螺环基团,例如螺(4,4)壬烷-2-基。术语“(C 3-8)环烷基”指如以上定义具有多达8个碳原子的环烷基基团。
在本申请中,术语“环烷基烷基”通常是指含有约3至8个与烷基基团直接连接的碳原子,烷基基团再在来自烷基基团的导致稳定结构形成的任何碳原子处与主要结构连接的含环基团,例如环丙基甲基、环丁基乙基和环戊基乙基。
在本申请中,术语“环烯基”通常是指含有约3至8个具有至少一个碳-碳双键的碳原子的含环基团,例如环丙烯基、环丁烯基和环戊烯基。术语“环烯基烷基”指直接与烷基基团连接,烷基基团再在来自烷基基团的导致稳定结构形成的任何碳原子处与主要结构连接的含环基团。
在本申请中,术语“芳基”通常是指具有6-20个碳原子的芳香族基团,例如苯基、萘基、四氢萘基、茚满基和联苯基。
在本申请中,术语“芳基烷基”通常是指如以上定义直接与以上定义的烷基基团连接的芳基基团,例如-CH 2C 6H 5和-C 2H 5C 6H 5
在本申请中,术语“杂环”通常是指由碳原子和至少一个选自氮、磷、氧和硫的杂原子组成的非芳香族3-15元环基。为了本申请的目的,杂环基可为单、双、三或四环环系,其可包括稠环、桥环或螺环环系,并且杂环基中的氮、磷、碳、氧或硫原子可被任选氧化为各种氧化态。另外,氮原子可被任选地季铵化。杂环基可在导致稳定结构形成的任何杂原子或碳原子处与主要结构连接。术语“杂环基”通常是指如以上定义的杂环型环基。杂环型环基可在导致稳定结构形成的任何杂原子或碳原子处与主要结构连接。
在本申请中,术语“杂环基烷基”通常是指如以上定义与烷基基团直接结合杂环型环基。杂环基烷基可在烷基基团中导致稳定结构形成的碳原子处与主要结构连接。这种杂环烷基的实例包括但不限于二氧戊环基、噻吩基[l,3]二噻烷基、十氢异喹啉基、咪唑啉基、咪唑烷基、异噻唑烷基、异噁唑烷基、吗啉基、八氢吲哚基、八氢异吲哚基、2-氧代哌嗪基、2-氧代哌啶基、2-氧代吡咯烷基、噁唑烷基、哌啶基、哌嗪基、4-哌啶酮基、吡咯烷基、吡唑烷基、奎宁环基、噻唑烷基、四氢呋喃基、三噻烷基、四氢吡喃基、硫代吗啉基、硫杂吗啉基、1-氧代-硫代吗啉基和1,1-二氧代-硫代吗啉基。
在本申请中,术语“杂芳基”通常是指具有一个或多个选自N、O和S的杂原子作为环原子的经任选取代的5-14元芳香环。杂芳基可为单、双或三环环系。这种“杂环”或“杂芳基”基团的实例包括但不限于噁唑基、噻唑基、咪唑基、吡咯基、呋喃基、吡啶基、嘧啶基、吡嗪基、苯并呋喃基、吲哚基、苯并噻唑基、苯并噁唑基、咔唑基、喹啉基、异喹啉基、吖丁啶基、吖啶基、苯并间二氧杂环戊烯基、苯并二噁烷基、苯并呋喃基、咔唑基、噌啉基、二氧戊环基、吲嗪基、萘啶基、全氢氮杂卓基、吩嗪基、吩噻嗪基、吩噁嗪基、酞嗪基、蝶啶基、嘌呤基、喹唑啉基、喹喔啉基、四唑基、四氢异喹啉基、哌啶基、哌嗪基、2-氧代哌嗪基、2-氧代哌啶基、2-氧代吡咯烷基、2-氧代氮杂卓基、氮杂卓基、4-哌啶酮基、吡咯烷基、哒嗪基、噁唑基、噁唑烷基、三唑基、茚满基、异噁唑基、异噁唑烷基、吗啉基、噻唑啉基、噻唑烷基、异噻唑基、奎宁环基、异噻唑烷基、异吲哚基、吲哚啉基、异吲哚啉基、八氢吲哚基、八氢异吲哚基、十氢异喹啉基、苯并咪唑基、噻二唑基、苯并吡喃基、四氢呋喃基、四氢吡喃基、噻吩基、苯并噻吩基、硫杂吗啉基、硫杂吗啉基亚砜、硫杂吗啉基砜、二氧磷杂环戊烷基、噁二唑基、色满基和异色满基。杂芳基环基可在导致稳定结构形成的任何杂原子或碳原子处与主要结构连接。术语“经取代的杂芳基”还包括经一个或多个氧化物(-O-)取代基(例如吡啶基N-氧化物)取代的环系。术语“杂芳基烷基”通常是指如以上定义与烷基基团直接结合的杂芳基环基。杂芳基烷基可在来自烷基基团的导致稳定结构形成的任何碳原子处与主要结构连接。
在本申请中,术语“环”通常是指含有3-10个碳原子的环。
在本申请中,术语“免疫检查点”通常是指在CD4细胞和CD8T细胞的细胞表面上的一组分子。这些分子可以有效地充当下调或抑制抗肿瘤免疫反应的“刹车”。免疫检查点分子包括但不限于程序性细胞死亡1(PD-1)、程序性细胞死亡配体1(PD-L1)、程序性细胞死亡配体2(PD-L2)、淋巴细胞活化基因-3(LAG-3;也称为CD223)、半乳糖凝集素-3、B和T淋巴细胞衰减子(BTLA)、T-细胞膜蛋白3(TIM3)、半乳糖凝集素-9(GAL9)、B7-H1、B7-H3、B7-H4、具有Ig和ITIM结构域的T-细胞免疫受体(TIGIT/Vstm3/WUCAM/VSIG9)、T-细胞活化的V-结构域Ig抑制物(VISTA)、糖皮质激素诱导的肿瘤坏死因子受体相关(GITR)蛋白、疱疹病毒进入介质(HVEM)、OX40、CD27、CD28、CD80、CD86、CD137、CGEN-15001T、CGEN-15022、CGEN-15027、CGEN-15049、CGEN-15052和CGEN-15092。
在本申请中,术语“免疫检查点的抑制剂”通常是指抑制、降低或干扰抑制性检查点分子的活性的分子。不受特定理论的束缚,抑制性检查点分子通过其被配体或反受体结合后向T细胞递送负信号来下调免疫应答(例如,T-细胞活化)。在某些实施方式中,与本申请提供 的方法和组合物一起使用的检查点抑制剂可以直接抑制抑制性检查点分子的活性或降低抑制性检查点分子的表达或干扰抑制性检查点分子和结合伴侣(例如,配体)的相互作用。
在本申请中,“免疫检查点的抑制剂”包括但不限于,蛋白、多肽、肽、反义寡核苷酸、抗体、抗体片段或RNA分子(例如,靶向抑制性检查点分子的表达的抑制性RNA分子)。其抑制作用可以在DNA、RNA或蛋白水平进行。例如,抑制性核酸(例如,dsRNA、siRNA或shRNA)可以用于抑制抑制性分子的表达。在其他实施方式中,抑制信号的抑制剂是与免疫检查点结合的多肽,例如,可溶性配体(例如,PD-1-Ig),抗体或其抗原结合片段;例如,与PD-1、PD-L1、PD-L2、CTLA4、TIM3、LAG3、VISTA、BTLA、TIGIT、LAIR1、CD160、2B4和/或TGFRβ或其组合结合的抗体或其片段。
在本申请中,术语“TGFβRII”或“TGFβ受体II”通常是指具有野生型人2型TGFβ受体同种型A序列(如NCBI参考序列(Ref Seq)登记号NP_001020018的氨基酸序列)的多肽,或具有野生型人2型TGFβ受体同种型B序列(如NCBI Ref Seq登记号NP_003233的氨基酸序列)或具有与它们的氨基酸序列基本相同序列的多肽。TGFβRII可保留野生型序列TGFβ结合活性的至少0.1%、0.5%、1%、5%、10%、25%、35%、50%、75%、90%、95%或99%。
在本申请中,术语“能够结合TGFβ的TGFβRII片段”通常是指NCBI Ref Seq登记NP_001020018或NCBI Ref Seq登记号NP_003233或与它们基本相同序列的任意部分,片段长度至少20(例如至少30、40、50、60、70、80、90、100、110、120、130、140、150、160、175或200)个氨基酸且保留野生型受体或其野生型片段的至少部分TGFβ结合活性(例如至少0.1%、0.5%、1%、5%、10%、25%、35%、50%、75%、90%、95%或99%)。通常,这些片段是可溶性片段。示例性片段之一是具有SEQ ID NO:6序列的TGFβRII胞外域。
在本申请中,“基本相同”通常是指多肽与参照氨基酸序列显示至少50%,优选60%、70%、75%或80%,更优选85%、90%或95%,且最优选99%的氨基酸序列相同性(同一性)。比较序列的长度一般至少10个氨基酸,优选至少15个连续氨基酸,至少20、25、50、75、90、100、150、200、250、300或350个连续氨基酸更好,最好是全长氨基酸序列。例如,适合测定序列相同性百分数和序列相似性百分数的算法可以是BLAST和BLAST 2.0算法,分别可参见Altschul等(1977)NucleicAcids Res.25:3389和Altschul等(1990)J.Mol.Biol.215:403。
在本申请中,术语“TGFβ抑制剂”通常是指抑制TGFβ信号分子产生和信号转导的物 质,所述抑制剂可以是小分子化合物,也可以是大分子化合物(如抗体)。示例性的小分子TGFβ抑制剂包括SB525334、SD-208、SB431542、LY2109761、LY2157299(Galunisertib)、GW788388、RepSox、SIS3、LDN-193189、EW-7197和LY364947等。
Figure PCTCN2022075642-appb-000007
在本申请中,术语“抗体”通常是指对指定蛋白质或肽或其片段有反应性的免疫球蛋白。抗体可以是来自任何类的抗体,包括但不限于IgG、IgA、IgM、IgD和IgE,及来自任何亚类(例如IgG1、IgG2、IgG3、和IgG4)的抗体。抗体可具有选自例如IgG1、IgG2、IgG3、或IgG4的重链恒定区。抗体还可具有选自例如kappa(κ)或lambda(λ)的轻链。本申请的抗体可衍生自任何物种。
在本申请中,术语“抗原结合片段”通常是指抗体分子的一部分,其包含负责抗体与抗原之间的特异性结合的氨基酸。抗原中由抗体特异性地识别和结合的部分是称作如上文所述的“表位”。如上文所述,抗原结合结构域可典型地包含抗体轻链可变区(VL)和抗体重链可变区(VH);然而,其并非必须包含两者。Fd片段例如具有两个VH区并且通常保留完整抗原结合结构域的一些抗原结合功能。抗体的抗原结合片段的实例包括(1)Fab片段,具有VL、VH、恒定轻链(CL)和CH1结构域的单价片段;(2)F(ab′)2片段,具有由铰链区的二硫桥连接 的两个Fab片段的二价片段;(3)具有两个VH和CH1结构域的Fd片段;(4)具有抗体单臂的VL和VH结构域的Fv片段,(5)dAb片段(Ward等人,“Binding Activities of a Repertoire of Single Immunoglobulin Variable DomainsSecreted From Escherichia coli,”Nature 341:544-546(1989),其以引用的方式整体并入本申请),其具有VH结构域;(6)分离的互补决定区(CDR);(7)单链Fv(scFv),例如源于scFV-文库。尽管Fv片段的两个结构域VL和VH是由独立基因编码,但其可通过合成连接子使用重组方法接合,合成连接子使得其被制备为其中VL和VH区配对以形成单价分子的单一蛋白链(称为单链Fv(scFv))(可参见例如Huston等人,“Protein Engineering of AntibodyBinding Sites:Recovery of Specific Activity in an Anti-Digoxin Single-ChainFv Analogue Produced in Escherichia coli,”Proc.Natl.Acad.Sci.USA 85:5879-5883(1988));(8)“VHH”涉及来自骆驼科(骆驼、单峰骆驼、美洲驼、羊驼等)重链抗体的可变抗原结合结构域(参见Nguyen V.K.等人,2000,The EMBO Journal,19,921-930;Muyldermans S.,2001,J Biotechnol.,74,277-302以及综述Vanlandschoot P.等人,2011,Antiviral Research 92,389-407)。VHH也可称为纳米抗体(Nanobody)(Nb)。
在本申请中,术语“可变区”或“可变结构域”通常是指参与抗体与抗原的结合的抗体重链或轻链的结构域。在本申请中,术语“可变”通常是指,抗体的可变结构域的序列的某些部分变化强烈,形成各种特定抗体对其特定抗原的结合和特异性。变异性并非均匀地分布在抗体的整个可变区中。它集中在轻链可变区和重链可变区中的三个区段,被称为互补决定区(CDR)或高变区(HVR),分别为LCDR1、LCDR2、LCDR3、HCDR1、HCDR2和HCDR3。可变域中更高度保守的部分被称为框架区(FR)。天然重链和轻链的可变结构域各自包含四个FR区(H-FR1,H-FR2,H-FR3,H-FR4,L-FR1,L-FR2,L-FR3,L-FR4),大部分采用β-折叠构型,通过三个CDR结构环区连接。每条链中的CDR通过FR区紧密靠近在一起,并与来自另一条链的CDR一起形成抗体的抗原结合位点。
在本领域中,可以通过多种方法来编码抗体的可变区或划分抗体的CDR,例如基于序列可变性的Kabat编号方案和定义规则(参见,Kabat等人,免疫学的蛋白质序列,第五版,美国国立卫生研究院,贝塞斯达,马里兰州(1991)),基于结构环区域位置的Chothia编号方案和定义规则(参见,A1-Lazikani等人,JMol Biol 273:927-48,1997),efranc等人的基于种系V基因的氨基酸序列比对的IMGT编号方案和定义规则,还有Honneger’s编号方案(AHo’s),Martin编号方案,Gelfand编号方案等,可参见Mathieu Dondelinger等人,Understanding the Significance and Implications of Antibody Numbering and Antigen-Binding Surface/Residue Definition,Front.Immunol.,16 October 2018.
在本申请中,“肽连接子”通常是指这样的氨基酸序列:通过所述氨基酸序列,将本申请PD-L1/TGFβ双重抑制剂中的不同结构域的氨基酸序列彼此相连。这种肽连接子的必要技术特征在于所述肽连接子不包含任何的聚合活性。对肽连接子优选的氨基酸残基包括Gly、Ser和Thr,特征在于长度为5至25个氨基酸残基。合适的肽连接子包括在美国专利4,751,180和4,935,233或WO88/09344中描述的那些。肽连接子的优选实施方案的特征在于氨基酸序列Gly-Gly-Gly-Gly-Ser,即Gly 4Ser,或其聚合物,即(Gly 4Ser)x,其中x是整数1或更大。所述肽连接子的特征,包括不促进二级结构,是本领域中已知的并且例如描述于Dall'Acqua等人(Biochem.(1998)37,9266-9273)、Cheadle等人(MolImmunol(1992)29,21-30)以及Raag和Whitlow(FASEB(1995)9(1),73-80)。还不促进任何二级结构的肽连接子是优选的。如实施例中所述,例如通过基因工程提供所述结构域与彼此的连接。用于制备融合和可操作地连接双特异性单链构建体和在哺乳动物细胞或细菌中表达它们的方法在本领域中是公知的(例如WO99/54440或Sambrook等人,Molecular Cloning:ALaboratoryManual,ColdSpring Harbor Laboratory Press,Cold Spring Harbor,NewYork,2001)。
在本申请中,术语“拮抗剂”和“抑制剂”可互换使用,通常是指能够降低或抑制靶蛋白或多肽的生物功能,如降低或抑制靶蛋白或多肽的活性或表达的化合物或药剂。抑制剂不需要完全消除靶蛋白或多肽的生物学功能,并且在一些实施方式中,使活性降低至少50%、60%、70%、80%、90%、95%或99%。虽然本申请的一些拮抗剂特异性地与目标相互作用(例如,结合于目标),但通过与包括靶蛋白或多肽的信号转导通路中的其他成员相互作用而抑制靶蛋白或多肽的生物活性的化合物也具体地包括在该定义中。被拮抗剂抑制的生物活性的非限定性实例包括与肿瘤的发展、生长或扩散或在自身免疫疾病中表现的不希望的免疫应答相关的那些活性。
在本申请中,术语“有效量”或“治疗有效量”通常是指足以实现下文说明的预期应用的本申请所述的化合物或药物组合物的量,所述预期应用包括但不限于疾病治疗。治疗有效量可根据以下变化:预期应用(体内或体外);或所治疗的对象及疾病状况,例如,对象的体重和年龄、疾病状况的严重程度;给药方式等,其可以由本领域普通技术人员容易地确定。术语还适用于将在靶细胞中诱导特定应答的剂量,所述应答为例如血小板粘附和/或细胞迁移。具体剂量将根据例如以下变化:选择的具体化合物、遵循的给药方案、是否与其他药剂组合施用、施用时间、施用的组织,和运送其的实体递送系统。
在本申请中,术语“体内”通常是指发生在对象体内的事件。
在本申请中,术语“体外”通常是指在对象体外发生的事件。例如,体外分析包括任何 在对象外部进行的分析。体外分析包括基于细胞的分析,其中采用活细胞或死细胞。体外分析还包括无细胞分析,其中不采用完整的细胞。
在本申请中,“组合疗法”或“组合”通常是指使用多于一种化合物或药剂来治疗特定的障碍或病症。例如,PI3K抑制剂可以与至少一种另外的治疗剂组合施用。“组合”不是旨在暗示其他疗法和PI3K抑制剂必须同时施用和/或配制用于一起递送,但这些递送方法在本申请的范围内。PI3K抑制剂可以与一种或多种另外的药剂同时施用,在其之前(例如,5分钟、15分钟、30分钟、45分钟、1小时、2小时、4小时、6小时、12小时、24小时、48小时、72小时、96小时、1周、2周、3周、4周、5周、6周、8周、12周或16周前)施用或在其之后(例如,5分钟、15分钟、30分钟、45分钟、1小时、2小时、4小时、6小时、12小时、24小时、48小时、72小时、96小时、1周、2周、3周、4周、5周、6周、8周、12周或16周后)施用。一般地,每种治疗剂将以针对该具体药剂确定的剂量和/或按针对该具体药剂确定的时间表施用。其他治疗剂可以与本申请提供的PI3K抑制剂在单一组合物或分别在不同组合物中施用。本申请也考虑更高的组合,例如PI3K抑制剂+PD-L1抑制剂+TGFβ抑制剂的三联疗法。
组合疗法术语“施用”通常是指通过任意引入或递送途径将所述药物组合引入受试者的身体中。可以采用本领域技术人员已知的用于使细胞、器官或组织与所述药物组合接触的任何方法。包括而不限于动脉内、鼻内、腹内、静脉内、肌内、皮下透皮或口服。每日剂量可以划分成一个、两个或更多个合适形式的剂量以在某个时间段期间的一个、两个或更多个时间施用。
在某些实施方式中,术语“施用”包括向受试者施用两种或多种药剂以使药剂和/或其代谢物同时或基本同时存在于对象中。在某些实施方式中,PI3K抑制剂与另外的抗癌剂的共同施用(两种组分在下文中称为“两种活性剂”)是指两种活性剂的任何施用,分开或一起施用,其中两种活性剂作为旨在获得组合治疗的益处的适宜剂量方案的部分施用。因此,两种活性剂可以作为相同药物组合物的部分施用或在分开的药物组合物中施用。另外的药剂可以在施用PI3K抑制剂之前、同时或之后施用,或者以其某些组合施用。当PI3K抑制剂例如在标准疗程期间以重复的间隔向患者施用时,另外的药剂可以在每次施用PI3K抑制剂之前、同时或之后施用,或者以其某些组合施用,或以相对于PI3K抑制剂治疗不同的间隔施用,或在使用PI3K抑制剂的疗程之前、在使用PI3K抑制剂的疗程期间的任何时间或在使用PI3K抑制剂的疗程之后以单剂量施用。在某些实施方式中,第一药剂可以在施用第二治疗剂之前(例如,5分钟、15分钟、30分钟、45分钟、1小时、2小时、4小时、6小时、12 小时、24小时、48小时、72小时、96小时、1周、2周、3周、4周、5周、6周、8周或12周之前),基本上同时或之后(例如,5分钟、15分钟、30分钟、45分钟、1小时、2小时、4小时、6小时、12小时、24小时、48小时、72小时、96小时、1周、2周、3周、4周、5周、6周、8周或12周之后)施用。
在本申请中,“单一疗法”是指单独(本申请也称为单独地(alone))使用药剂(例如,作为单一化合物或药剂),例如在没有第二活性成分的情况下治疗相同的适应症,例如癌症。例如,在上下文中,术语单一疗法包括单独使用PI3K抑制剂或第二药剂治疗癌症。
在本申请中,术语“协同作用”或“协同的”包括与两种或多种药剂的单独作用相比更高的两种或多种药剂的组合的累加作用。在某些实施方式中,协同或协同作用是指,例如在药物组合物中或在治疗方法中组合使用两种或更多种药剂的有利作用。在某些实施方式中,通过使用与本申请所述的第二治疗剂(例如,一种或多种第二治疗剂)组合的PI3K抑制剂实现一种或多种有利作用。
在一些实施方式中,协同作用是指实现作用需要更低剂量的一种或两种药剂。例如,当至少一种药剂在比当该药剂作为单一疗法施用时实现相同的治疗效果所需的药剂的剂量更低的剂量下施用时,组合可以提供选择的效果,例如治疗效果。在某些实施方式中,PI3K抑制剂(例如,PI3K抑制剂)和第二药剂(如本申请所述)的组合允许PI3K抑制剂在比当PI3K抑制剂作为单一疗法施用时实现相同的治疗效果所需的更低的剂量下施用。
在一些实施方式中,协同作用减少、预防、延迟或降低原本与施用药剂中的至少一种相关的一种或多种副作用、毒性、抗性的发生或发生的可能性。
在一些实施方式中,协同作用减少对药剂中的至少一种的抗性(例如,抗性的测量值降低或产生抗性的可能性降低)或延迟对药剂中的至少一种的抗性的产生。
在一些实施方式中,协同作用是最小残留疾病(MRD)的降低。在某些实施方式中,PI3K抑制剂(例如,本申请所述的PI3K抑制剂)和第二药剂(例如,本申请所述的第二药剂)的组合有效降低对象中的MRD,例如,低于先前在对象中测量的水平(例如,在施用组合之前测量的水平)。在某些实施方式中,PI3K抑制剂和第二药剂的组合有效使对象中的MRD降低到在施用单一疗法治疗期间或之后观察到的水平以下,所述单一疗法例如包含PI3K抑制剂或第二药剂中任一种的单一疗法。在某些实施方式中,MRD降低到在使用包含PI3K抑制剂的单一疗法治疗期间观察到的水平以下。在某些实施方式中,MRD降低到在使用包含第二药剂的单一疗法治疗期间观察到的水平以下。在某些实施方式中,组合有效使MRD的水平降低到预选的截止值(例如,100个正常细胞中有1个恶性肿瘤细胞,1000个正常细胞中有1 个恶性肿瘤细胞,或10,000个正常细胞中有1个恶性肿瘤细胞,或100,000个正常细胞中有1个恶性肿瘤细胞)以下。在某些实施方式中,预选的截止值是1000个正常细胞中有1个恶性肿瘤细胞。在某些实施方式中,预选的截止值是100,000个正常细胞中有1个恶性肿瘤细胞。
在一些实施方式中,协同作用是指PI3K抑制剂(例如,PI3K抑制剂或其药学上可接受的形式)和第二治疗剂(例如,一种或多种另外的治疗剂或其药学上可接受的形式)的组合产生比PI3K抑制剂和第二药剂的累加作用更高的治疗作用。
在本申请中,术语“药学上可接受的”通常是指不干扰活性成分生物学活性的有效性的一种或多种无毒物质。这类制剂通常可含有盐、缓冲剂、防腐剂、相容性载体和任选的其它治疗剂。
在本申请中,化合物的“药学上可接受的形式”包括但不限于,公开的化合物的药学上可接受的盐、水合物、溶剂化物、异构体、前药及同位素标记的衍生物。在某些实施方式中,“药学上可接受的形式”包括但不限于,公开的化合物的药学上可接受的盐、异构体、前药及同位素标记的衍生物。
在本申请中,术语“药学上可接受的盐”通常是指在合理医学判断的范围内适合用于与对象的组织接触而无过度的毒性、刺激性、过敏反应等,且与合理的效益/风险比相符的那些盐。药学上可接受的盐在本领域是熟知的。例如,Berge等人在J.Pharmaceutical Sciences(1977)66:1–19中详细描述了药学上可接受的盐。本申请提供的化合物的药学上可接受的盐包括衍生自适合的无机和有机酸和碱的盐。药学上可接受的无毒的酸加成盐的实例是与无机酸(如盐酸、氢溴酸、磷酸、硫酸和高氯酸)或有机酸(如乙酸、草酸、顺丁烯二酸、酒石酸、柠檬酸、丁二酸或丙二酸)形成的氨基的盐,或通过使用在本领域使用的其他方法如离子交换形成的氨基的盐。其他药学上可接受的盐包括己二酸盐、海藻酸盐、抗坏血酸盐、天冬氨酸盐、苯磺酸盐、苯磺酸盐(besylate)、苯甲酸盐、硫酸氢盐、硼酸盐、丁酸盐、樟脑酸盐、樟脑磺酸盐、柠檬酸盐、环戊烷丙酸盐、二葡糖酸盐、十二烷基硫酸盐、乙磺酸盐、甲酸盐、反丁烯二酸盐、葡庚糖酸盐、甘油磷酸盐、葡糖酸盐、半硫酸盐、庚酸盐、己酸盐、氢碘酸盐、2-羟基-乙磺酸盐、乳糖醛酸盐、乳酸盐、月桂酸盐、月桂基硫酸盐、苹果酸盐、顺丁烯二酸盐、丙二酸盐、甲磺酸盐、2-萘磺酸盐、烟酸盐、硝酸盐、油酸盐、草酸盐、棕榈酸盐、双羟萘酸盐、果胶酸盐、过硫酸盐、3-苯基丙酸盐、磷酸盐、苦味酸盐、三甲基乙酸盐、丙酸盐、硬脂酸盐、丁二酸盐、硫酸盐、酒石酸盐、硫氰酸盐、对甲苯磺酸盐、十一烷酸盐、戊酸盐等。在一些实施方式中,可以衍生出盐的有机酸包括例如,乙酸、 丙酸、乙醇酸、丙酮酸、草酸、顺丁烯二酸、丙二酸、丁二酸、反丁烯二酸、酒石酸、柠檬酸、苯甲酸、肉桂酸、扁桃酸、甲磺酸、乙磺酸、对甲苯磺酸、水杨酸等。
衍生自适合的碱的药学上可接受的盐包括碱金属盐、碱土金属盐、铵盐和N +(C 1-4烷基) 4盐。代表性的碱金属盐或碱土金属盐包括钠盐、锂盐、钾盐、钙盐、镁盐、铁盐、锌盐、铜盐、锰盐、铝盐等。适宜时,另外的药学上可接受的盐包括,无毒铵、季铵和使用抗衡离子形成的胺阳离子,所述抗衡离子如卤离子、氢氧根、羧酸根、硫酸根、磷酸根、硝酸根、低级烷基磺酸根和芳基磺酸根。可以衍生出盐的有机碱包括例如一级胺、二级胺和三级胺、被取代的胺(包括天然存在的被取代的胺)、环胺、碱性离子交换树脂等,如异丙胺、三甲胺、二乙胺、三乙胺、三丙胺和乙醇胺。在一些实施方式中,药学上可接受的碱加成盐选自铵盐、钾盐、钠盐、钙盐和镁盐。
在本申请中,术语“溶剂化物”通常是指还包括化学计量的或非化学计量的量的通过非共价分子间力结合的溶剂的化合物。溶剂化物可以是所公开的化合物或其药学上可接受的盐。当溶剂为水时,溶剂化物为“水合物”。药学上可接受的溶剂和水合物是例如可以包括1个至约100个、或1个至约10个、或1个至约2个、约3个或约4个溶剂或水分子的复合物。应理解的是,在本申请中的术语“化合物”包括化合物和化合物的溶剂化物,以及其混合物。
在本申请中,术语“前药”通常是指在体内转化得到所公开的化合物或化合物的药学上可接受的形式的化合物。前药在向对象施用时可以是非活性的,但是在体内例如通过水解(例如,在血液中水解)转化为活性化合物。在某些情况下,前药具有比母体化合物改善的物理和/或递送性质。前药通常被设计成提高与母体化合物相关的基于药学和/或药物动力学的性质。前药化合物通常提供溶解度、组织相容性或在哺乳动物有机体内延迟释放的优点(参见,例如,Bundgard,H.,Design ofProdrugs(1985),pp.7-9,21-24(Elsevier,Amsterdam))。前药的讨论提供于Higuchi,T.等人,“Pro-drugs as Novel Delivery Systems,”A.C.S.Symposium Series,Vol.14,Chp 1,pp 1-12和Bioreversible Carriers in Drug Design,ed.Edward B.Roche,American Pharmaceutical Association and Pergamon Press,1987中,将两者的全部内容以引用方式并入本申请。前药的示例性的优点可以包括但不限于其物理性质,如对于在生理pH下的肠胃外施用比母体化合物水溶性提高,或其增强来自消化道的吸收,或其可以增强长期储存的药物稳定性。
在本申请中,术语“前药”还旨在包括任何共价键合的载体,当向对象施用该前药时,所述载体在体内释放活性化合物。如本申请所述的活性化合物的前药可以通过以如下的方式修饰存在于活性化合物中的基团来制备,即使该修饰在常规操作中或在体内裂解成母体活性 化合物。前药包括其中的羟基、氨基或巯基键合于任意基团的化合物,当向对象施用活性化合物的前药时,所述任意基团裂解以分别形成游离的羟基、游离的氨基或游离的巯基。前药的实例包括但不限于,活性化合物中的醇的乙酸酯、甲酸酯和苯甲酸酯衍生物或胺官能团的乙酰胺、甲酰胺和苯甲酰胺衍生物等。前药的其他实例包括包含-NO、-NO2、-ONO或-ONO2部分的化合物。前药通常可以使用熟知方法制备,如在Burger’s Medicinal Chemistry and Drug Discovery,172-178,949-982(Manfred E.Wolff ed.,5th ed.,1995)和Design of Prodrugs(H.Bundgaard ed.,Elsevier,New York,1985)中描述的那些。
例如,如果所公开的化合物或化合物的药学上可接受的形式含有羧酸官能团,则前药可以包含通过用以下基团代替酸基的氢原子而形成的药学上可接受的酯,所述基团如(C1-C8)烷基、(C2-C12)酰氧基甲基、具有4至9个碳原子的1-(酰氧基)乙基、具有5至10个碳原子的1-甲基-1-(酰氧基)-乙基、具有3至6个碳原子的烷氧基羰基氧基甲基、具有4至7个碳原子的1-(烷氧基羰基氧基)乙基、具有5至8个碳原子的1-甲基-1-(烷氧基羰基氧基)乙基、具有3至9个碳原子的N-(烷氧基羰基)氨基甲基、具有4至10个碳原子的1-(N-(烷氧基羰基)氨基)乙基、3-酞基、4-巴豆酰内酯基、γ-丁内酯-4-基、二-N,N-(C1-C2)烷基氨基(C2-C3)烷基(如β-二甲基氨基乙基)、氨甲酰基-(C1-C2)烷基、N,N-二(C1-C2)烷基氨甲酰基-(C1-C2)烷基,和哌啶基、吡咯烷基或吗啉基(C2-C3)烷基。
类似地,如果所公开的化合物或该化合物的药学上可接受的形式含有醇官能团,则前药可以通过用以下基团代替醇基的氢原子而形成,所述基团如(C1-C6)酰氧基甲基、1-((C1-C6)酰氧基)乙基、1-甲基-1-((C1-C6)酰氧基)乙基、(C1-C6)烷氧基羰基氧基甲基、N-(C1-C6)烷氧基羰基氨基甲基、丁二酰基、(C1-C6)酰基、α-氨基(C1-C4)酰基、芳基酰基和α-氨基酰基或α-氨基酰基-α-氨基酰基,其中每个α-氨基酰基独立地选自天然存在的L-氨基酸、P(O)(OH)2、-P(O)(O(C1-C6)烷基)2和糖基(通过将糖的半缩醛形式的羟基去除而产生的基团)。
如果所公开的化合物或该化合物的药学上可接受的形式包括胺官能团,则前药可以通过用以下基团代替胺基团中的氢原子而形成,所述基团如R-羰基、RO-羰基、NRR′-羰基,其中R和R′各自独立地为(C1-C10)烷基、(C3-C7)环烷基、苄基、天然α-氨基酰基或天然α-氨基酰基-天然α-氨基酰基;-C(OH)C(O)OY1,其中Y1为H、(C1-C6)烷基或苄基;-C(OY2)Y3,其中Y2为(C1-C4)烷基和Y3为(C1-C6)烷基、羧基(C1-C6)烷基、氨基(C1-C4)烷基或单-N-或二-N,N-(C1-C6)烷基氨基烷基;-C(Y4)Y5,其中Y4为H或甲基和Y5为单-N-或二-N,N-(C1-C6)烷基氨基、吗啉基、哌啶-1-基或吡咯烷-1-基。
在本申请中,“异构体”通常是指具有相同分子式的不同的化合物。“立体异构体”通常是指仅原子的空间排列方式不同的异构体。在本申请中,术语“异构体”包括任何以及所有几何异构体和立体异构体。例如,“异构体”包括几何双键顺式和反式异构体,也称为E–和Z–异构体;R–和S–对映异构体;非对映异构体、(d)–异构体和(l)–异构体、其外消旋混合物;及其落入本公开范围内的其他混合物。
在本申请中,“对映异构体”通常是指一对彼此的不可重叠的镜像的立体异构体。一对对映异构体的1:1混合物为“外消旋”混合物。适宜时,术语“(±)”用于表示外消旋混合物。“非对映异构体”是具有至少两个不对称原子,但不为彼此的镜像的立体异构体。绝对立体化学是根据Cahn-Ingold-Prelog R-S系统规定的。当化合物为纯对映异构体时,每个手性碳处的立体化学可以由R或S来指定。绝对构型未知的拆分的化合物可以根据其在钠D线的波长下使平面偏振光旋转的方向(右旋或左旋)来指定为(+)或(-)。本申请描述的某些化合物含有一个或多个不对称中心,因此可产生对映异构体、非对映异构体和其他可以根据绝对立体化学定义的立体异构形式,如(R)-或(S)-。本申请的化学实体、药物组合物和方法旨在包括所有这样的可能的异构体,包括外消旋混合物、光学纯形式和中间混合物。光学活性的(R)-和(S)-异构体可以使用手性合成子或手性试剂制备,或使用常规技术拆分。当本申请所述的化合物包含烯属双键或其他几何不对称中心时,并且除非另有说明,否则所述化合物旨在包括E和Z几何异构体二者。
在本申请中,术语“对映异构体纯度”通常是指特定对映异构体相对于另一种对映异构体的存在的相对量,以百分比表示。例如,如果可能具有(R)-或(S)-异构构型的化合物作为外消旋混合物存在,则就(R)-或(S)-异构体中的任一种而言,对映异构体纯度为约50%。如果该化合物的一种构形式优于另一种,例如,80%(S)-和20%(R)-,则该化合物就(S)-异构形式而言的对映体纯度为80%。化合物的对映异构体纯度可以通过本领域已知的多种方式测定,包括但不限于使用手性载体的色谱法,偏振光旋转的偏振测量,使用手性位移试剂(包括但不限于含有镧系元素的手性配合物或Pirkle醇)的核磁共振光谱法,或使用手性化合物如Mosher酸对化合物进行衍生化,然后进行色谱或核磁共振光谱。
在本申请中,术语“互变异构体”通常是指一种类型的异构体,其包括由氢原子的至少一种形式迁移及化合价的至少一种变化(例如,单键到双键、三键到双键或三键到单键,反之亦然)产生的两种或多种可相互转化的化合物。“互变异构”包括质子移变或质子移动互变异构,其视为酸碱化学的一个子集。“质子移变互变异构”或“质子移动互变异构”涉及质子的迁移,伴随键级的变化。互变异构体的确切比例取决于多种因素,包括温度、溶剂和pH。 当互变异构化可能存在(例如,在溶液中)时,可以达到互变异构体的化学平衡。互变异构(即,提供互变异构体对的反应)可以由酸或碱催化,或可以在没有外部试剂的作用或存在下发生。示例性的互变异构包括但不限于,酮-烯醇;酰胺-酰亚胺;内酰胺-内酰亚胺;烯胺-亚胺;和烯胺-(不同的)烯胺互变异构。酮-烯醇互变异构的具体的实例是戊烷-2,4-二酮与4-羟基戊-3-烯-2-酮互变异构体的相互转化。互变异构的另一个实例是酚-酮互变异构。酚-酮互变异构的具体实例为吡啶-4-醇和吡啶-4(1H)-酮互变异构体的相互转化。
除非另有规定,本申请描绘的结构还旨在包括区别仅在于存在一个或多个同位素富集的原子的化合物。例如,除了氢在分子中的一个或多个原子处被氘或氚代替或富集氘或氚,或碳在分子中的一个或多个原子处被 13C或 14C代替或富集 13C或 14C以外,具有本申请的结构的化合物在本公开的范围内。
在本申请中,本申请的化合物还包括同位素标记的化合物,其与本申请所述的那些相同,但一个或多个原子被原子质量或质量数与通常在自然界中发现的原子质量或质量数不同的原子代替。可以加入所公开的化合物中的同位素的实例包括氢、碳、氮、氧、磷、硫、氟和氯的同位素,分别例如 2H、 3H、 13C、 14C、 15N、 18O、 17O、 31P、 32P、 35S、 18F和 36Cl。某些同位素标记的所公开的化合物(例如,被 3H和/或 14C标记的那些)在化合物和/或基质组织分布分析中是有用的。氚(即, 3H)和碳-14(即, 14C)同位素可以使得制备容易且可检测。此外,使用较重的同位素如氘(即, 2H)取代可以由于更高的代谢稳定性而提供某些治疗优点(例如,体内半衰期延长或剂量需求降低)。同位素标记的所公开的化合物一般可以通过用同位素标记的试剂取代非同位素标记的试剂来制备。在一些实施方式中,本申请提供了在构成该化合物的一个或多个原子处还含有非天然比例的原子同位素的化合物。本申请公开的所有同位素变化,无论是否具有放射性,均包括在本公开的范围内。
在本申请中,“药学上可接受的载体”或“药学上可接受的赋形剂”包括任何及所有溶剂、分散介质、包衣、抗细菌剂和抗真菌剂、等渗剂和吸收延迟剂等。针对药物活性物质使用这样的介质和试剂在本领域中是熟知的。除非任何常规的介质或试剂与活性成分不相容,考虑其在本申请公开的实验组合物中的使用。补充的活性成分也可以加入到药物组合物中。
在本申请中,术语“肿瘤”通常是指恶性或良性的任何赘生性细胞生长和增殖,以及任何癌前和癌细胞和组织。在本申请中,术语“赘生的”是指恶性或良性的任何形式的异常调节或未经调节的细胞生长,导致异常的组织生长。因此,“赘生性细胞”包括具有异常调节或未经调节的细胞生长的恶性和良性细胞。术语“癌症”,“癌性”,“细胞增殖性病症”,“增殖性病症”和“肿瘤”在本申请中提到时并不互相排斥。在一些实施方案中,肿瘤可以指含有多数癌细胞, 例如显示本申请中描述的任何癌症的特征细胞的肉体块。在本申请中,肿瘤可以是实体瘤或非实体瘤(例如,血液瘤、淋巴瘤)。术语“实体瘤”通常指代异常的组织生长或团块,其通常不含有囊肿或液性区。实体瘤可以是良性(非癌性)或恶性(癌性)的。在本申请中,所述肿瘤可以为细胞和组织的PD-1或PD-L1表达异常的肿瘤。在本申请中,所述肿瘤可以为细胞和组织的TGFβ或TGFβR表达异常或活性异常的肿瘤。在本申请中,所述肿瘤可以为细胞和组织的PI3K表达异常或活性异常的肿瘤。
在本申请中,术语“抗性”通常是指当癌症对治疗具有降低的响应,例如,直至癌症不响应治疗的程度。癌症在治疗开始时可以是抗性的或其在治疗期间产生抗性。癌症对象可以具有导致其对治疗产生抗性的一种或多种突变,或对象可以在治疗期间产生这样的突变。术语“难治性”可以指已经证明治疗(例如化疗药物、生物制剂和/或放射疗法)无效的癌症。难治性癌症肿瘤可以缩小,但尚未达到确定治疗有效的程度。然而,通常,肿瘤保持与治疗前相同的尺寸(稳定的疾病),或者生长(进行性疾病)。
在本申请中,术语“治疗(treatment)”和“治疗(treating)”通常是指获得有益或希望的结果的方法,所述有益或希望的结果包括但不限于治疗益处。治疗益处包括但不限于根除、抑制、减少或改善所治疗的潜在障碍。另外,治疗益处是通过根除抑制、减少或改善与潜在的障碍相关的一种或多种生理症状实现的,从而在患者中观察到改善,但是患者仍然可能患有潜在障碍。
在本申请中,术语“预防(prevention)”和“预防(preventing)”通常是指获得有益或希望的结果的方法,所述有益或希望的结果包括但不限于预防益处。为了预防益处,可以向处于患上特定疾病的风险的患者或向报告具有疾病的一种或多种生理症状的患者施用药物组合物,即使尚未诊断出该疾病。
在本申请中,术语“受试者”或“患者”通常是指人类(即,任何年龄组的男性或女性,例如,小儿对象(例如,婴儿、儿童、青少年)或成人对象(例如,年轻人、中年人或老年人))和/或其他灵长类动物(例如,食蟹猴、恒河猴);哺乳动物,包括商业上相关的哺乳动物,如牛、猪、马、绵羊、山羊、猫和/或犬;和/或鸟类,包括商业上相关的鸟类,如鸡、鸭、鹅、鹌鹑和/或火鸡。
在本申请中,术语“约”或“大约”通常是指本领域普通技术人员测定的具体值的可接受的误差,其部分取决于测量或测定值的方式。在某些实施方式中,术语“约”或“大约”通常是指1、2、3或4个标准偏差。在某些实施方式中,术语“约”或“大约”通常是指在给定值或范围50%、20%、15%、10%、9%、8%、7%、6%、5%、4%、3%、2%、1%、 0.5%或0.05%内。
在本申请中,术语“包含”或者“包括”通常是指开放式形式,应当理解为还可以含有没有述及的其他物质。例如,一种药物组合,包含磷酸肌醇3-激酶(PI3K)抑制剂和第二治疗剂,其中所述第二治疗剂为免疫检查点的抑制剂,TGFβ抑制剂,双功能免疫检查点/TGFβ抑制剂或它们的组合。其中“包含”应当理解为除含有磷酸肌醇3-激酶(PI3K)抑制剂和第二治疗剂之外,还可以含有其他物质。
发明详述
药物组合
一方面,本申请提供一种药物组合,包含磷酸肌醇3-激酶(PI3K)抑制剂和第二治疗剂,其中所述第二治疗剂为免疫检查点的抑制剂,TGFβ抑制剂,双功能免疫检查点/TGFβ抑制剂或其组合。
在某些实施方式中,其中所述免疫检查点的抑制剂可以包括能够阻断程序性死亡1(PD-1)与程序性死亡配体1(PD-L1)相互作用的试剂。
在某些实施方式中,其中所述免疫检查点的抑制剂可以包括程序性死亡配体1(PD-L1)和/或程序性死亡1(PD-1)的抑制剂。例如,人PD-1的抑制剂,例如人PD-L1的抑制剂。又例如,免疫检查点的抑制剂可以包含抗PD-L1抗体或其抗原结合片段,或抗PD-1抗体或其抗原结合片段。在某些实施方式中,PD-1或PD-L1的抑制剂可以是针对PD-1或PD-L1的抗体分子。PD-1或PD-L1抑制剂可以单独或与其他免疫检查点调节剂组合施用,例如,与LAG-3、TIM-3或CTLA-4的抑制剂组合。
在某些实施方式中,所述药物组合可以包括:
1)PD-L1抑制剂和PI3K抑制剂的组合;
2)TGFβ抑制剂和PI3K抑制剂的组合;
3)PD-1抑制剂、TGFβ抑制剂和PI3K抑制剂的组合;
4)PD-L1抑制剂、TGFβ抑制剂和PI3K抑制剂的组合;
5)PD-L1/TGFβ双重抑制剂和PI3K抑制剂的组合;或
6)PD-1/TGFβ双重抑制剂和PI3K抑制剂的组合。
在某些实施方式中,其中所述免疫检查点的抑制剂可以包括核酸(例如,dsRNA、siRNA或shRNA),多肽(例如,可溶性配体,抗体或其抗原结合片段,免疫粘附素,融合蛋白,寡肽和其它分子),或化合物。例如,抗体可以选自:人抗体、人源化抗体、嵌合抗体、多 特异性抗体、单克隆抗体和多克隆抗体。例如,抗原结合片段可以选自:Fab、Fab’、F(ab’) 2、Fv、scFv、双抗体、Fd、dAb、VHH、大抗体和互补决定区(CDR)片段。
在某些实施方式中,其中所述抗体或其抗原结合片段可以包含含有HCDR1,HCDR2和HCDR3的重链可变区(VH)。
在某些实施方式中,其中所述抗体或其抗原结合片段还可以包含或不包含CH1结构域。
在某些实施方式中,其中所述抗体或其抗原结合片段还可以包含或不包含CH2和CH3结构域。例如,所述抗体或其抗原结合片段包含VH-CH2-CH3的结构域。
在某些实施方式中,其中所述抗体或其抗原结合片段还可以包含抗体重链恒定区。
在某些实施方式中,其中所述抗体重链恒定区可以为人抗体重链恒定区。例如,所述人抗体重链恒定区可以选自源自下组的恒定区:IgG1、IgG2、IgG3、IgG4和它们的变体。
在某些实施方式中,其中所述抗体或其抗原结合片段还可以包含含有LCDR1,LCDR2,LCDR3的轻链可变区。
例如,所述抗体或其抗原结合片段可以包含可以含有HCDR1,HCDR2和HCDR3的重链可变区(VH),和含有LCDR1,LCDR2,LCDR3的轻链可变区。
在某些实施方式中,其中所述抗体或其抗原结合片段还可以包含轻链恒定区(CL)。例如,所述抗体或其抗原结合片段可以包含含有VH和CH1的重链,和含有VL和CL的抗体轻链。又例如,所述抗体或其抗原结合片段可以包含含有VH、CH1、CH2和CH3的重链,和含有VL和CL的抗体轻链。
PD-1/PD-L1抑制剂
在某些实施方式中,免疫检查点的抑制剂可以是PD-1/PD-L1抑制剂。PD-l/PD-L1抑制剂的实例包括但不限于US 7,488,802、US 7,943,743、US 8,008,449、US 8,168,757、US 8,217,149、US 8,609,089、US 2010/028330、US 2012/0114649、WO 2003/042402、WO 2008/156712、WO 2010/089411、WO 2010/036959、WO 2011/066342、WO 2011/159877、WO 2011/082400和WO 2011/161699中描述的那些,将其所有的全部内容通过引用并入本申请。
在某些实施方式中,检查点调节剂可以是PD-1抑制剂。例如,检查点调节剂可以是抗PD-1抗体。
例如,抗PD-1抗体可以是纳武单抗。纳武单抗的供选择的名称包括MDX-1106、MDX-1106-04、ONO-4538或BMS-936558,并且CAS登记号为:946414-94-4。纳武单抗是特异性阻断PD-1的全长人IgG4单克隆抗体。纳武单抗(克隆5C4)和特异性与PD-1结合的其他人单 克隆抗体公开于US 8,008,449和WO 2006/121168中。
例如,抗PD-1抗体可以是帕姆单抗。帕姆单抗(商品名KEYTRUDA,原名兰利珠单抗(Lambrolizumab),也称为Merck 3745、MK-3475或SCH-900475)是与PD-1结合的人源化IgG4单克隆抗体。帕姆单抗公开于例如,Hamid,O.等人(2013)New England Journalof Medicine369(2):134–44,WO 2009/114335,和US 8,354,509中。
例如,抗PD-1抗体可以是皮地利珠单抗。皮地利珠单抗(CT-011;CureTech)是与PD-1结合的人源化IgG1k单克隆抗体。皮地利珠单抗和其他人源化抗PD-1单克隆抗体公开于WO 2009/101611中。其他抗PD1抗体公开于US 8,609,089、US 2010/028330和/或US 2012/0114649中。
例如,抗PD-1抗体可以是AMP-514(Amplimmune)。
例如,PD-1抑制剂可以是免疫粘附素(例如,包含与恒定区(例如,免疫球蛋白序列的Fc区)融合的PD-Ll或PD-L2的细胞外或PD-1结合部分的免疫粘附素)。
在某些实施方式中,检查点调节剂是PD-L1抑制剂。例如,检查点调节剂可以是抗PD-L1抗体或其抗原结合片段。“PD-L1抗体或其抗原结合片段”可包括本领域中所述的任何抗PD-L1抗体或其抗原结合片段。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包含HCDR1,HCDR2,HCDR3,它们各自分别与以下分子的HCDR1,HCDR2,HCDR3具有至少约80%(例如,约85%,90%或95%)的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A(RG7446)或durvalumab(MEDI4736)。
在某些实施方式中,其中所述重链可变区分别与以下分子的重链可变区具有至少约80%(例如,约85%,90%或95%)的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链,其中所述重链分别与以下分子的重链具有至少约80%(例如,约85%,90%或95%)的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段还包含抗体的轻链可变区,其中所述轻链可变区包含LCDR1,LCDR2,LCDR3,它们各自分别与以下分子的LCDR1,LCDR2,LCDR3具有至少约80%(例如,约85%,90%或95%)的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab。
例如,所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区和轻链可变区,其 中所述重链可变区包含HCDR1,HCDR2,HCDR3,其中所述轻链可变区包含LCDR1,LCDR2,LCDR3,它们各自分别与以下分子的HCDR1,HCDR2,HCDR3,LCDR1,LCDR2,LCDR3具有至少约80%(例如,约85%,90%或95%)的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A(RG7446)或durvalumab(MEDI4736)。
在某些实施方式中,其中所述轻链可变区分别与以下分子的轻链可变区具有至少约80%(例如,约85%,90%或95%)的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab。
例如,所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区和轻链可变区,它们各自分别与以下分子的重链可变区和轻链可变区具有至少约80%(例如,约85%,90%或95%)的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A(RG7446)或durvalumab(MEDI4736)。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段还包含抗体的轻链,其中所述轻链分别与以下分子的轻链具有至少约80%(例如,约85%,90%或95%)的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab。
例如,所述抗PD-L1抗体或其抗原结合片段包含抗体的重链和轻链,它们各自分别与以下分子的重链和轻链具有至少约80%(例如,约85%,90%或95%)的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A(RG7446)或durvalumab(MEDI4736)。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段包括:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab,或其抗原结合片段,或其变体或生物类似物,或其组合。
在某些实施方式中,其中所述抗PD-L1抗体还可以是市售可得的或已通过文献公开的PD-L1抗体。包括但不限于,如PD-L1抗体BMS-936559,MPDL3280A,MEDI4736,MSB0010718C(参见US2014341917、US20130034559、US8779108)等。
例如,抗PD-L1抗体是MDX-1105。MDX-1105,也称为BMS-936559,是抗PD-Ll抗体,如WO 2007/005874中所述。例如,抗PD-L1抗体是YW243.55.S70。YW243.55.S70抗体是抗PD-L1抗体,如WO 2010/077634中所述。YW243.55.S70的重链和轻链可变区序列也描述于WO2010/077634中。例如,抗PD-L1抗体是MDPL3280A(Genentech/Roche)。MDPL3280A是与PD-L1结合的人Fc优化的IgG1单克隆抗体。MDPL3280A和其他PD-L1的人单克隆抗体描述于US 7,943,743和US 2012/0039906中。例如,抗PD-L1抗体是Avelumab(MSB0010718C)。MSB0010718C(也称为A09-246-2;Merck Serono)是与PD-L1结合的单克隆 抗体。其他人源化抗-PD-L1抗体公开于WO 2013/079174中。例如,抗PD-L1抗体是durvalumab(CAS Number:1428935-60-7)。例如,抗PD-L1抗体是Atezolizumab(CAS Number:1380723-44-3)。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包括一个或多个选自下组的重链可变区CDRs(HCDRs):(a)具有与SEQ ID NO:1所示氨基酸序列至少约70%序列同一性的HCDR1;(b)具有与SEQ ID NO:2至少约70%序列同一性的HCDR2;和(c)具有与SEQ ID NO:3至少约70%序列同一性的HCDR3。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包括一个或多个选自下组的HCDRs:(a)具有SEQ ID NO:1所示氨基酸序列的HCDR1或者通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:1所示氨基酸序列不超过2个氨基酸差异的HCDR1;(b)具有SEQ ID NO:2所示氨基酸序列的HCDR2或者通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:2所示氨基酸序列不超过2个氨基酸差异的HCDR2;和(c)具有SEQ ID NO:3所示氨基酸序列的HCDR3或者通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:3所示氨基酸序列不超过2个氨基酸差异的HCDR3。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包含HCDR1,HCDR2,HCDR3,所述HCDR1包含与SEQ ID NO:1所示氨基酸序列至少约70%序列同一性的氨基酸序列,所述HCDR2包含与SEQ ID NO:2所示氨基酸序列至少约70%序列同一性的氨基酸序列,所述HCDR3包含与SEQ ID NO:3所示氨基酸序列至少约70%序列同一性的氨基酸序列。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包含HCDR1,HCDR2,HCDR3,所述HCDR1包含SEQ ID NO:1所示的氨基酸序列或通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:1所示的氨基酸序列不超过2个氨基酸差异的氨基酸序列;所述HCDR2包含SEQ ID NO:2所示的氨基酸序列或通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:2所示的氨基酸序列不超过2个氨基酸差异的氨基酸序列;所述HCDR3包含SEQ ID NO:3所示的氨基酸序列或通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:3所示的氨基酸序列不超过2个氨基酸差异的氨基酸序列。
在某些实施方式中,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区, 其中所述重链可变区包含选自下组的氨基酸序列:(a)SEQ ID NO:4所示的氨基酸序列;(b)与SEQ ID NO:4所示的氨基酸序列至少约85%,90%,95%或者99%序列同一性的氨基酸序列;和(c)通过加成、消除或取代反应所得到的与SEQ ID NO:4所示的氨基酸序列具有1个或更多差异的氨基酸序列。
TGFβ抑制剂
在某些实施方式中,其中所述TGFβ抑制剂结合TGFβ1、TGFβ2或TGFβ3。
在某些实施方式中,其中所述TGFβ抑制剂结合TGFβ或转化生长因子β受体(TGFβR)。例如,所述TGFβ抑制剂可以结合TGFβI型受体(TGFβRI),TGFβII型受体(TGFβRII)或TGFβIII型受体(TGFβRIII)。
在某些实施方式中,其中所述TGFβ抑制剂包括:1)抗TGFβ抗体或其抗原结合片段;2)抗TGFβR抗体或其抗原结合片段;3)小分子TGFβ抑制剂;4)包含转化生长因子β受体II(TGFβRII)或其功能活性片段、或其变体或生物类似物的蛋白质,或它们的组合。
在某些实施方式中,其中所述抗TGFβ抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包含HCDR1,HCDR2,HCDR3,它们各自分别与Fresolimumab分子(CAS Number:948564-73-6)的HCDR1,HCDR2,HCDR3具有至少约80%(例如,约85%,90%或95%)的序列同一性。
在某些实施方式中,其中所述重链可变区可以与Fresolimumab分子的重链可变区具有至少约80%(例如,约85%,90%或95%)的序列同一性。
在某些实施方式中,其中所述重链可以与Fresolimumab分子的重链具有至少约80%(例如,约85%,90%或95%)的序列同一性。
在某些实施方式中,其中所述抗TGFβ抗体或其抗原结合片段包含抗体的轻链可变区,其中所述轻链可变区包含LCDR1,LCDR2,LCDR3,它们各自分别与Fresolimumab分子的LCDR1,LCDR2,LCDR3具有至少约80%(例如,85%,90%或95%)的序列同一性。
例如,所述抗TGFβ抗体或其抗原结合片段包含抗体的重链可变区和轻链可变区,其中所述重链可变区包含HCDR1,HCDR2,HCDR3,其中所述轻链可变区包含LCDR1,LCDR2,LCDR3,它们各自分别可以与Fresolimumab分子的HCDR1,HCDR2,HCDR3,LCDR1,LCDR2,LCDR3具有至少约80%(例如,85%,90%或95%)的序列同一性。
在某些实施方式中,其中所述轻链可变区可以与Fresolimumab分子的轻链可变区具有至少约80%的序列同一性。
例如,所述抗TGFβ抗体或其抗原结合片段包含抗体的重链可变区和轻链可变区,它们 各自分别可以与Fresolimumab分子的重链可变区和轻链可变区具有至少约80%(例如,约85%,90%或95%)的序列同一性。
在某些实施方式中,其中所述轻链与Fresolimumab分子的轻链具有至少约80%的序列同一性。
例如,所述抗TGFβ抗体或其抗原结合片段包含抗体的重链和轻链,它们各自分别可以与Fresolimumab分子的重链和轻链具有至少约80%(例如,85%,90%或95%)的序列同一性。
在某些实施方式中,其中所述抗TGFβ抗体或其抗原结合片段可以包括:Fresolimumab,或其抗原结合片段,或其变体或生物类似物。
在某些实施方式中,其中所述小分子TGFβ抑制剂可以包括以下化合物:SB525334、SD-208、SB431542、LY2109761、LY2157299、GW788388、RepSox、SIS3、LDN-193189、EW-7197、LY364947、或其组合。
在某些实施方式中,其中所述TGFβRII可以具有野生型人TGFβ受体2型同种型A序列(例如,NCBI参照序列(Ref Seq)登录号NP_001020018的氨基酸序列)的多肽,或具有野生型人TGFβ受体2型同种型B序列(例如,NCBI Ref Seq登录号NP_003233的氨基酸序列)的多肽,或与其氨基酸序列具有基本相同(例如,约80%,85%,90%或95%相同性)的多肽。
在某些实施方式中,其中所述功能活性片段可以包括:人TGFβRII胞外域(ECD),NCBI Ref Seq登录号NP_001020018或NCBI Ref Seq登录号NP_003233的任何部分,或者与其氨基酸序列具有基本相同(例如,约80%,85%,90%或95%相同性)的序列。
在某些实施方式中,其中所述TGFβRII或其功能活性片段可以包含:
(a)SEQ ID NO:6所示的氨基酸序列;
(b)具有与SEQ ID NO:6所示的氨基酸序列至少约85%序列同一性的氨基酸序列;或
(c)具有与SEQ ID NO:6所示的氨基酸序列相比,添加,缺失和/或取代一个或多个氨基酸的氨基酸序列。
PD-L1/TGFβ双重抑制剂
在某些实施方式中,其中所述双功能免疫检查点/TGFβ抑制剂为融合蛋白。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂或PD-1/TGFβ双重抑制剂为融合蛋白。
在某些实施方式中,其中所述PD-L1//TGFβ双重抑制剂包含PD-L1靶向部分和TGFβ 受体结构域,所述TGFβ受体结构域包含转化生长因子β受体II(TGFβRII)或其功能活性片段。
在某些实施方式中,其中所述PD-L1//TGFβ双重抑制剂包含多肽,其中所述多肽至少包含:(i)抗PD-L1抗体的重链可变区;和(ii)TGFβRII或其功能活性片段。
在某些实施方式中,其中所述抗PD-L1抗体重链可变区包含HCDR1,HCDR2,HCDR3,所述HCDR1包含与SEQ ID NO:1所示氨基酸序列至少70%序列同一性的氨基酸序列,所述HCDR2包含与SEQ ID NO:2所示氨基酸序列至少70%序列同一性的氨基酸序列,所述HCDR3包含与SEQ ID NO:3所示氨基酸序列至少70%序列同一性的氨基酸序列。
在某些实施方式中,其中所述抗PD-L1抗体重链可变区包含HCDR1,HCDR2,HCDR3,所述HCDR1包含SEQ ID NO:1所示的氨基酸序列或通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:1所示的氨基酸序列不超过2个氨基酸差异的氨基酸序列;所述HCDR2包含SEQ ID NO:2所示的氨基酸序列或通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:2所示的氨基酸序列不超过2个氨基酸差异的氨基酸序列;所述HCDR3包含SEQ ID NO:3所示的氨基酸序列或通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:3所示的氨基酸序列不超过2个氨基酸差异的氨基酸序列。
在某些实施方式中,其中所述抗PD-L1抗体重链可变区包含选自下组的氨基酸序列:(a)SEQ ID NO:4所示的氨基酸序列;(b)与SEQ ID NO:4所示的氨基酸序列至少85%,90%,95%或者99%序列同一性的氨基酸序列;和(c)通过加成、消除或取代反应所得到的与SEQ ID NO:4所示的氨基酸序列具有1个或更多差异的氨基酸序列。
在某些实施方式中,其中所述TGFβRII或其功能活性片段包含:
(a)SEQ ID NO:6所示的氨基酸序列;
(b)具有与SEQ ID NO:6所示的氨基酸序列至少约85%序列同一性的氨基酸序列;或
(c)具有与SEQ ID NO:6所示的氨基酸序列相比,添加,缺失和/或取代一个或多个氨基酸的氨基酸序列。
在某些实施方式中,其中所述多肽还包含连接子,所述连接子将所述抗PD-L1的抗体或其抗原结合片段的重链可变区的C末端与所述TGFβRII或其功能活性片段的N末端相连接。
在某些实施方式中,所述多肽还包括CH2、CH3结构域,所述多肽自N末端至C末端依次包含抗PD-L1抗体的重链可变区(VH)、CH2、CH3结构域和TGFβRII或其功能活性片 段,所述CH3结构域的C末端与所述TGFβRII或其功能活性片段N末端通过连接子相连接。
在某些实施方式中,其中所述CH2、CH3结构域源自IgG。
在某些实施方式中,其中所述连接子为肽连接子。
在某些实施方式中,其中所述肽连接子的氨基酸序列为G(G 4S) x,其中x为3-6。
在某些实施方式中,其中所述肽连接子包含选自下组氨基酸序列:(a)SEQ ID NO:5所示的氨基酸序列;(b)具有与SEQ ID NO:5所示的氨基酸序列至少85%,90%,95%或者99%序列同一性的氨基酸序列;和(c)通过加成、消除或取代反应所得到的与SEQ ID NO:5所示的氨基酸序列具有1个或更多差异的氨基酸序列。
在某些实施方式中,其中所述多肽包含选自下组的氨基酸序列:(a)SEQ ID NO:7所示的氨基酸序列;(b)具有与SEQ ID NO:7所示的氨基酸序列至少85%,90%,95%或者99%序列同一性的氨基酸序列;(c)通过加成、消除或取代反应所得到的与SEQ ID NO:7所示的氨基酸序列具有1个或更多差异的氨基酸序列。
在某些实施方式中,其中所述PD-L1//TGFβ双重抑制剂包含两条前述多肽。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂包含第一多肽和第二多肽,其中所述第一多肽至少包含:(i)抗PD-L1抗体的重链可变区;和(ii)TGFβRII或其功能活性片段;其中所述第二多肽至少包含抗PD-L1抗体的轻链可变区;其中所述第一多肽的重链可变区和所述第二多肽的轻链可变区在组合时能够特异性结合PD-L1。
在某些实施方式中,其中所述第一多肽还包含连接子,所述连接子将所述抗PD-L1的抗体或其抗原结合片段的重链可变区的C末端与所述TGFβRII或其功能活性片段的N末端相连接。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂包含第一多肽和第二多肽,其中:
所述第一多肽自N末端至C末端依次包含抗PD-L1抗体的重链可变区(VH)、CH1结构域以及TGFβRII或其功能活性片段,所述CH1结构域的C末端与所述TGFβRII或其功能活性片段N末端通过连接子相连接;和所述第二多肽自N末端至C末端包含抗PD-L1抗体的轻链可变区(VL)和轻链恒定区(CL)。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂包含第一多肽和第二多肽,其中:
所述第一多肽自N末端至C末端依次包含抗PD-L1抗体的重链可变区(VH)、CH1、 CH2、CH3结构域以及TGFβRII或其功能活性片段,所述CH3结构域的C末端与所述TGFβRII或其功能活性片段N末端通过连接子相连接;和所述第二多肽自N末端至C末端包含抗PD-L1抗体的轻链可变区(VL)、轻链恒定区(CL)。
在某些实施方式中,其中所述连接子为肽连接子。
在某些实施方式中,其中所述第一多肽与以下分子的具有相同功能的多肽具有至少约80%序列同一性:M7824或SHR-1701。
在某些实施方式中,其中所述第二多肽与以下分子的具有相同功能的多肽具有至少约80%序列同一性:M7824或SHR-1701。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂包括M7824、SHR-1701,或其变体或生物类似物,或其组合。
PI3K抑制剂
在某些实施方式中,PI3K抑制剂可以是抑制一种或多种PI3K亚型,例如α、β、δ或γ亚型的化合物。在一个实施方式中,PI3K抑制剂可以是抑制PI3K的α、β、δ和γ亚型的化合物。在另一个实施方式中,PI3K抑制剂可以是抑制PI3K的β、δ和γ亚型的化合物。在另一个实施方式中,PI3K抑制剂可以是抑制PI3K的δ和γ亚型的化合物,即PI3Kδ/γ双重抑制剂。
在某些实施方式中,其中所述PI3K抑制剂可以包括式(IA-I)、(IA-II)、(IA-III)、或(IA-IV)所示的化合物:
Figure PCTCN2022075642-appb-000008
或它们的对映异构体、对映异构体的混合物、两种或更多种非对映异构体的混合物、同位素变体、药学上可接受的盐、溶剂化物、水合物或前药,其中
每次出现的R可以独立地选自氢、卤素、-OR a、CN、经取代或未经取代的C 1-6烷基、经取代或未经取代的C 2-6烯基、经取代或未经取代的C 2-6炔基、经取代或未经取代的C 3-8环烷基和经取代或未经取代的杂环基团;
R 1和R 2可相同或不同并且独立地选自氢、卤素和经取代或未经取代的C 1-6烷基,或与 共有原子直接结合的R 1和R 2可连接形成=O基或经取代或未经取代的饱和或不饱和3-10元环,包括与R 1和R 2结合的碳原子,所述环可任选地包括一个或多个相同或不同且选自O、NR a和S的杂原子;
Cy 1可以为选自经取代或未经取代的C 3-8环烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环基团、经取代或未经取代的C6-20芳基和经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基的单环基团;
每次出现的R a可相同或不同且独立地选自氢、卤素、羟基、氰基、经取代或未经取代的C 1-6烷基;-NR cR d,其中R c,R d独立地选自氢、卤素、羟基、氰基、经取代或未经取代的C 1-6烷基和C 1-6烷氧基,和-OR c,其中R c为经取代或未经取代的C 1-6烷基;
n可以为1-4的整数;并且
q可以为0、1或2;
每次出现的X可以独立地选自CR 3或N;
并且每次出现的R 3可以独立地选自氢、羟基、卤素、羧基、氰基、硝基、经取代或未经取代的C 1-8烷基、经取代或未经取代的C 1-8烷氧基、经取代或未经取代的C 2-10烯基、经取代或未经取代的C 2-12炔基、经取代或未经取代的C 6-20芳基、经取代或未经取代的C 6-20芳基C 1-8烷基、经取代或未经取代的C 3-20环烷基、经取代或未经取代的C 3-20环烷基C 1-8烷基、经取代或未经取代的C 3-8环烯基C 1-8烷基、经取代或未经取代的C 3-8环烯基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基C 1-8烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环基C 1-8烷基环、经取代或未经取代的胍、-C00R x、-C(O)R x、-C(S)R x、-C(O)NR xR y、-C(O)ONR xR y,-NR yR z、-NR xCONR yR z,-N(R x)SOR y、-N(Rx)SO 2R y、-(=N-N(R x)R y)、-NR xC(O)OR y、-NR xR y、-NR xC(O)R y-、-NR xC(S)R y、-NR xC(S)NR yR z、-SONR xR y-、-SO 2NR xR y-、-OR x、-OR xC(O)NR yR z、-OR xC(O)OR y-、-OC(O)R x、-OC(O)NR xR y、-R xNR yC(O)R z、-R xOR y、-R xC(O)OR y、-R xC(O)NR yR z、-R xC(O)R x、-R xOC(O)R x、-SR x、-SOR x、-SO 2R x和-ONO 2,其中以上每个基团中的R x、R y和R z可为氢、经取代或未经取代的C 1-8烷基、经取代或未经取代的C 1-8烷氧基、经取代或未经取代的C 2-10烯基、经取代或未经取代的C 2-12炔基、经取代或未经取代的C 6-20芳基、经取代或未经取代的C 6-20芳基C 1-8烷基、经取代或未经取代的C 3-20环烷基、经取代或未经取代的C 3-20环烷基C 1-8烷基、经取代或未经取代的C 3-8环烯基、经取代或未经 取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基C 1-8烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环、经取代或未经取代的具有至少一个选自N、0和S的杂原子的3至15元杂环基C 1-8烷基环或经取代或未经取代的氨基,或R x、R y和R z的任两个可连接形成经取代或未经取代的饱和或不饱和3-10元环,所述环可任选地包括可能相同或不同且选自O、NR x或S的杂原子,其中R x为氢或经取代或未经取代的烷基。
在本申请中,术语“经取代”通常是指用以下可相同或不同且独立地选自以下的取代基的任一个或任何组合取代:氢、羟基、卤素、羧基、氰基、硝基、=O基、=S基、C 1-C 8烷基、C 1-C 8烷氧基、C 2-C 10烯基、C 2-C 12炔基、C 6-C 20芳基、C 6-C 20芳基(C 1-C 8烷基)、C 3-C 12环烷基、C 3-C 12环烷基(C 1-C 8烷基)、C 3-C 8环烯基、C 3-C 8环烯基(C 1-C 8烷基);5-14元杂芳基,其中所述杂原子选自N、O和S;5-14元杂芳基(C 1-C 8烷基),其中所述杂原子选自N、O和S;3-15元杂环,其中所述杂原子选自N、O和S;以及3-15元杂环基(C 1-C 8烷基),其中所述杂原子选自N、O和S。
在某些实施方式中,胍、-C00R x、-C(O)R x、-C(S)R x、-C(O)NR xR y、-C(O)ONR xR y,-NR yR z、-NR xCONR yR z,-N(R x)SOR y、-N(Rx)SO 2R y、-(=N-N(R x)R y)、-NR xC(O)OR y、-NR xR y、-NR xC(O)R y-、-NR xC(S)R y、-NR xC(S)NR yR z、-SONR xR y-、-SO 2NR xR y-、-OR x、-OR xC(O)NR yR z、-OR xC(O)OR y-、-OC(O)R x、-OC(O)NR xR y、-R xNR yC(O)R z、-R xOR y、-R xC(O)OR y、-R xC(O)NR yR z、-R xC(O)R x、-R xOC(O)R x、-SR x、-SOR x、-SO 2R x和-ONO 2,其中以上每个基团中的R x、R y和R z可为氢、C 1-C 8烷基、C 1-C 8烷氧基、C 2-C 10烯基、C 2-C 12炔基、C 6-C 20芳基、C 6-C 20芳基(C 1-C 8烷基)、C 3-C 12环烷基、C 3-C 12环烷基(C 1-C 8烷基)、C 3-C 8环烯基、氨基;5-14元杂芳基,其中所述杂原子选自N、O和S;5-14元杂芳基(C 1-C 8烷基),其中所述杂原子选自N、O和S;3-15元杂环,其中所述杂原子选自N、O和S;3-15元杂环基(C 1-C 8烷基),或者R x、R y和R z的任两个可连接形成饱和或不饱和3-10元环,所述环可任选地包括可相同或不同且选自O、NR x或S的杂原子,其中R x为氢或C 1-6烷基。
在某些实施方式中,其中所述PI3K抑制剂可以包括式(IA-V)所示的化合物:
Figure PCTCN2022075642-appb-000009
或其对映异构体、对映异构体的混合物、两种或更多种非对映异构体的混合物、或同位素变体;或其药学上可接受的盐、溶剂化物、水合物或前药,其中
每次出现的R可以独立地选自氢、卤素、-OR a、CN、经取代或未经取代的C 1-6烷基、经取代或未经取代的C 2-6烯基、经取代或未经取代的C 2-6炔基、经取代或未经取代的C 3-8环烷基和经取代或未经取代的杂环基团;R 1和R 2可相同或不同并且独立地选自氢、卤素和经取代或未经取代的C 1-6烷基,或与共有原子直接结合的R 1和R 2可连接形成=O基或经取代或未经取代的饱和或不饱和3-10元环,包括与R 1和R 2结合的碳原子,所述环可任选地包括一个或多个相同或不同且选自O、NR a和S的杂原子;
每次出现的X可以独立地选自CR 3或N;并且
每次出现的R 3可以独立地选自氢、羟基、卤素、羧基、氰基、硝基、经取代或未经取代的C 1-8烷基、经取代或未经取代的C 1-8烷氧基、经取代或未经取代的C 2-10烯基、经取代或未经取代的C 2-12炔基、经取代或未经取代的C 6-20芳基、经取代或未经取代的C 6-20芳基C 1-8烷基、经取代或未经取代的C 3-20环烷基、经取代或未经取代的C 3-20环烷基C 1-8烷基、经取代或未经取代的C 3-8环烯基C 1-8烷基、经取代或未经取代的C 3-8环烯基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基C 1-8烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环基C 1-8烷基环、经取代或未经取代的胍、-C00R x、-C(O)R x、-C(S)R x、-C(O)NR xR y、-C(O)ONR xR y,-NR yR z、-NR xCONR yR z,-N(R x)SOR y、-N(Rx)SO 2R y、-(=N-N(R x)R y)、-NR xC(O)OR y、-NR xR y、-NR xC(O)R y-、-NR xC(S)R y、-NR xC(S)NR yR z、-SONR xR y-、-SO 2NR xR y-、-OR x、-OR xC(O)NR yR z、-OR xC(O)OR y-、-OC(O)R x、-OC(O)NR xR y、-R xNR yC(O)R z、-R xOR y、-R xC(O)OR y、-R xC(O)NR yR z、-R xC(O)R x、-R xOC(O)R x、-SR x、-SOR x、-SO 2R x和-ONO 2,其中以上每个基团中的R x、R y和R z可为氢、经取代或未经取代的C 1-8烷基、经取代或未经取代的C 1-8烷氧 基、经取代或未经取代的C 2-10烯基、经取代或未经取代的C 2-12炔基、经取代或未经取代的C 6-20芳基、经取代或未经取代的C 6-20芳基C 1-8烷基、经取代或未经取代的C 3-20环烷基、经取代或未经取代的C 3-20环烷基C 1-8烷基、经取代或未经取代的C 3-8环烯基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基C 1-8烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环、经取代或未经取代的具有至少一个选自N、0和S的杂原子的3至15元杂环基C 1-8烷基环或经取代或未经取代的氨基,或R x、R y和R z的任两个可连接形成经取代或未经取代的饱和或不饱和3-10元环,所述环可任选地包括可能相同或不同且选自O、NR x或S的杂原子,其中R x为氢或经取代或未经取代的烷基;
每次出现的R 5可以为氢、C 1-6烷基或卤素;
n可以为0、1、2、3或4;并且P可以为0、1、2、3、4或5。
在某些实施方式中,其中所述PI3K抑制剂可以包括式(IA-VI)所示的化合物:
Figure PCTCN2022075642-appb-000010
或其对映异构体、对映异构体的混合物、两种或更多种非对映异构体的混合物、或同位素变体;或其药学上可接受的盐、溶剂化物、水合物或前药,其中
每次出现的R可以独立地选自氢、卤素、-OR a、CN、经取代或未经取代的C 1-6烷基、经取代或未经取代的C 2-6烯基、经取代或未经取代的C 2-6炔基、经取代或未经取代的C 3-8环烷基和经取代或未经取代的杂环基团;
R 1和R 2可相同或不同并且独立地选自氢、卤素和经取代或未经取代的C 1-6烷基,或与共有原子直接结合的R 1和R 2可连接形成=O基或经取代或未经取代的饱和或不饱和3-10元环,包括与R 1和R 2结合的碳原子,所述环可任选地包括一个或多个相同或不同且选自O、NR a和S的杂原子;
每次出现的X可以独立地选自CR 3或N;并且
每次出现的R 3可以独立地选自氢、羟基、卤素、羧基、氰基、硝基、经取代或未经取 代的C 1-8烷基、经取代或未经取代的C 1-8烷氧基、经取代或未经取代的C 2-10烯基、经取代或未经取代的C 2-12炔基、经取代或未经取代的C 6-20芳基、经取代或未经取代的C 6-20芳基C 1-8烷基、经取代或未经取代的C 3-20环烷基、经取代或未经取代的C 3-20环烷基C 1-8烷基、经取代或未经取代的C 3-8环烯基C 1-8烷基、经取代或未经取代的C 3-8环烯基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基C 1-8烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环基C 1-8烷基环、经取代或未经取代的胍、-C00R x、-C(O)R x、-C(S)R x、-C(O)NR xR y、-C(O)ONR xR y,-NR yR z、-NR xCONR yR z,-N(R x)SOR y、-N(Rx)SO 2R y、-(=N-N(R x)R y)、-NR xC(O)OR y、-NR xR y、-NR xC(O)R y-、-NR xC(S)R y、-NR xC(S)NR yR z、-SONR xR y-、-SO 2NR xR y-、-OR x、-OR xC(O)NR yR z、-OR xC(O)OR y-、-OC(O)R x、-OC(O)NR xR y、-R xNR yC(O)R z、-R xOR y、-R xC(O)OR y、-R xC(O)NR yR z、-R xC(O)R x、-R xOC(O)R x、-SR x、-SOR x、-SO 2R x和-ONO 2,其中以上每个基团中的R x、R y和R z可为氢、经取代或未经取代的C 1-8烷基、经取代或未经取代的C 1-8烷氧基、经取代或未经取代的C 2-10烯基、经取代或未经取代的C 2-12炔基、经取代或未经取代的C 6-20芳基、经取代或未经取代的C 6-20芳基C 1-8烷基、经取代或未经取代的C 3-20环烷基、经取代或未经取代的C 3-20环烷基C 1-8烷基、经取代或未经取代的C 3-8环烯基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基C 1-8烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环、经取代或未经取代的具有至少一个选自N、0和S的杂原子的3至15元杂环基C 1-8烷基环或经取代或未经取代的氨基,或R x、R y和R z的任两个可连接形成经取代或未经取代的饱和或不饱和3-10元环,所述环可任选地包括可能相同或不同且选自O、NR x或S的杂原子,其中R x为氢或经取代或未经取代的烷基;
每次出现的R 5可以为氢、C 1-6烷基或卤素;
n可以为0、1、2、3或4;并且
P可以为0、1、2、3、4或5。
例如,其中R可以为氢、卤素、经取代或未经取代的C 1-6烷基或OR a,又例如,其中所述R a可以为烷基。
例如,其中所述Cy 1可以选自:
Figure PCTCN2022075642-appb-000011
例如,其中所述R 1和R 2可以各自独立地表示氢或经取代或未经取代的C 1-6烷基。
例如,其中所述R 3可以为碘代、氰基、经取代或未经取代的烷基、经取代或未经取代的炔基、经取代或未经取代的芳基、经取代或未经取代的杂芳基。
例如,其中X可以为CR 3并且每次出现的R 3独立为氢、卤素、羟基或NH 2,又例如,其中所述R 3可以为氢。
又例如,其中所述PI3K抑制剂可以包括:
2-[(6-氨基-9H-嘌呤-9-基)甲基]-6-溴-3-苯基-4H-色烯-4-酮;
2-[(6-氨基-9H-嘌呤-9-基)甲基]-3-苯基-4H-色烯-4-酮;
2-((9H-嘌呤-6-基硫代)甲基)-3-苯基-4H-色烯-4-酮;
2-[(9H-嘌呤-6-基硫代)甲基]-6-溴-3-苯基-4H-色烯-4-酮;
2-((4-氨基-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-溴-3-苯基-4!1-色烯-4-酮;
2-[(6-氨基-9H-嘌呤-9-基)甲基]-6-溴-3-(4-氟苯基)-4H-色烯-4-酮;
2-[(6-氨基-9H-嘌呤-9-基)甲基]-3-(4-氟苯基)-4H-色烯-4-酮;
2-[(6-氨基-9H-嘌呤-9-基)甲基]-6-溴-3-邻甲苯基-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-6-溴-3-苯基-4H-色烯-4-酮;
2-(1-(9H-嘌呤-6-基硫代)乙基)-6-溴-3-苯基-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-3-苯基-4H-色烯-4-酮;
(S)-2-(1-(9H-嘌呤-6-基氨基)乙基)-6-溴-3-苯基-4H-色烯-4-酮;
2-((9H-嘌呤-6-基氨基)甲基)-6-溴-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-溴-3-苯基-4H-色烯-4-酮;
2-((6-氨基-9H-嘌呤-9-基)甲基)-6-甲氧基-3-苯基-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-6-溴-3-(2-氟苯基)-4H-色烯-4-酮;
2-((6-氨基-9H-嘌呤-9-基)甲基)-6-溴-3-(2-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)丙基)-3-苯基-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((6-氨基-9H-嘌呤-9-基)甲基)-3-(2-氟苯基)-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-3-(2-氟苯基)-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)丙基)-3-(2-氟苯基)-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)丙基)-3-(4-氟苯基)-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)丙基)-6-氟-3-苯基-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-3-(4-氟苯基)-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-6-氟-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
2-((9H-嘌呤-6-基氨基)甲基)-3-苯基-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-3-邻甲苯基-4H-色烯-4-酮;
2-((9H-嘌呤-6-基氨基)甲基)-3-(2-氟苯基)-4H-色烯-4-酮;
2-((9H-嘌呤-6-基氨基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
(S)-2-(1-(9H-嘌呤-6-基氨基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-6-氟-3-(2-氟苯基)-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-3-(3,5-二氟苯基)-4H-色烯-4-酮;
2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
(R)-2-(1-(9H-嘌呤-6-基氨基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
(S)-2-(1-(9H-嘌呤-6-基氨基)乙基)-6-氟-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-氟-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-氟-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(吡啶-3-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(3-羟基丙-1-炔基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(3-(羟基甲基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(1H-吲唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(3-氟苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(3-羟基丙基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
N-(3-(4-氨基-1-((4-氧代-3-苯基-4H-色烯-2-基)甲基)-1H-吡唑并[3,4-d]嘧啶-3-基)苯基)乙酰胺;
2-((4-氨基-3-(3-氟-5-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(3-氟-5-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(3-氟-5-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3-氟-5-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-羟基-3-甲基丁-1-炔基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
2-(1_(4-氨基-3-(1H-吡唑-4-基)-1H-吡唑并[3,4-(1]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
(S)-2-(1-(9H-嘌呤-6-基氨基)乙基)-3-苯基-4H-色烯-4-酮;
(S)-2-(1-(9H-嘌呤-6-基氨基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-氟-5-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-氟-5-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(1H-吲唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3,5-二甲基-1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2-(羟基甲基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-氟-3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-氟-3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-羟基丙-1-炔基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-氟-4-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-氟-4-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-氯-5-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-氯-5-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-(三氟甲氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H- 色烯-4-酮;
2-(1-(4-氨基-3-(4-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((6-氨基-9H-嘌呤-9-基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-氟-2-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-氟-2-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3-氨基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-((4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2-氨基嘧啶-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(1H-吲哚-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-氯-3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-氯-3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2-氯-5-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2-氯-5-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3,4-二甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3,4-二羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-(3-氟苯基)-4H-色 烯-4-酮;
2-(1-(4-氨基-3-(1H-吲哚-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲基-1H-吲哚-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
叔丁基-(5-(4-氨基-1-(1-(3-(3-氟苯基)-4-氧代-4H-色烯-2-基)乙基)-1H-吡唑并[3,4-d]嘧啶-3-基)噻吩-2-基)氨基甲酸甲酯;
2-(1-(4-氨基-3-(5-(氨基甲基)噻吩-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-氟-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-氟-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲基-1H-吲唑-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
N-(4-(4-氨基-1-(1-(3-(3-氟苯基)-4-氧代-4H-色烯-2-基)乙基)-1H-吡唑并[3,4-d]嘧啶-3-基)苯基)乙酰胺;
2-(1-(4-氨基-3-(4-氨基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2,3-二氢苯并呋喃-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-乙基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲基-1H-吲哚-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2-甲氧基嘧啶-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色 烯-4-酮;
4-(4-氨基-1-(1-(3-(3-氟苯基)-4-氧代-4H-色烯-2-基)乙基)-1H-吡唑并[3,4-d]嘧啶-3-基)噻吩-2-甲醛;
2-(1-(4-氨基-3-(5-(羟基甲基)噻吩-3-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2-甲基-1H-苯并[d]咪唑-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲基-1H-吲哚-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((6-氨基-9H-嘌呤-9-基)甲基)-6-氟-3-苯基-4H-色烯-4-酮;2-((6-氨基-9H-嘌呤-9-基)甲基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3-氟-5-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3-氟-5-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-((9H-嘌呤-6-基氨基)甲基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-((9H-嘌呤-6-基氨基)甲基)-6-氟-3-苯基-4H-色烯-4-酮;(R)-2-(1-(9H-嘌呤-6-基氨基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-氟-3-苯基-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3,5-二氟-4-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3,5-二氟-4-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3,5-二氟-4-甲氧基苯基)-1H-吡唑并[3,4-(1]嘧啶-1-基)甲基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
2-((4-氨基-3-(3,5-二氟-4-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
(+)-2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
(-)-2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3,5-二甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-甲氧基-3,5-二甲基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2-氟-5-异丙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2,3-二氢苯并[b][1,4]二噁英-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(1-苄基-1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2-甲基吡啶-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3,4-二氢-2H-苯并[b][1,4]二氧杂卓-7-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(6-吗啉基吡啶-3-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(二苯并[b,d]呋喃-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-(苄氧基)-3-氯苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-氯-4-异丙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-(二甲基氨基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-乙氧基-3-氟苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-异丙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-(三氟甲氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(3-(4-乙酰基苯基)-4-氨基-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-(苄氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-(二甲基氨基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-(甲磺酰基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-乙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(苯并[b]噻吩-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(5-氯噻吩-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3,5-二甲基异噁唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-丙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2~(1-(4-氨基-3-(呋喃-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(4-乙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-氯-4-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-氟-4-异丙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(6-氟吡啶-3-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(嘧啶-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-(甲氧基甲基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(6-羟基萘-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-异丙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(1-甲基-1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(1,3-二甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2,3-二甲基-2H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(6-甲氧基萘-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(苯并[b]噻吩-3-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(2,4-二甲氧基嘧啶-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(6-乙氧基萘-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
3-(4-氨基-1-(1-(3-(3-氟苯基)-4-氧代-4H-色烯-2-基)乙基)-1H-吡唑并[3,4-d]嘧啶-3-基)-N-环丙基苯甲酰胺;
2-(1-(4-氨基-3-(3-(吗啉-4-羰基)苯基)-1H-吡唑并[3,4-(1]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
2-(1-(4-氨基-3-(3-(二氟甲氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
5-(4-氨基-1-(1-(3-(3-氟苯基)-4-氧代-4H-色烯-2-基)乙基)-1H-吡唑并[3,4-d]嘧啶-3-基)呋喃-2-甲醛;
和/或它们的对映异构体、对映异构体的混合物、两种或更多种非对映异构体的混合物、同位素变体、药学上可接受的盐、溶剂化物、水合物或前药。
在一个实施方式中,其中所述PI3K抑制剂可以包括:2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮,或其对映异构体、对映异构体的混合物、两种或更多种非对映异构体的混合物、或同位素变体;或其药学上可接受的盐、溶剂化物、水合物或前药。
在一个实施方式中,其中所述PI3K抑制剂可以为(S)-2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮(Tenalisib,又称为CN401)或其药学上可接受的盐、溶剂化物、水合物或前药。
在一个实施方式中,所述药物组合可以包括抗PD-L1抗体或其抗原结合片段和PI3K抑制剂。
例如,所述药物组合可以包括:(i)抗PD-L1抗体或其抗原结合片段,所述抗PD-L1抗体或其抗原结合片段包括Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab或其抗原结合片段;(ii)PI3Kδ/γ双重抑制剂,所述PI3Kδ/γ双重抑制剂包括2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮,或其对映异构体、对映异构体的混合物、两种或更多种非对映异构体的混合物、或同位素变体;或其药学上可接受的盐、溶剂化物、水合物或前药。
又例如,所述药物组合可以包括:(i)选自下组的抗PD-L1抗体:Atezolizumab、Avelumab、BMS-936559、MPDL3280A,durvalumab或其组合;(ii)(S)-2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮或其药学上可接受的盐、溶剂化物、水合物或前药。
在一个实施方式中,所述药物组合可以包括TGFβ抑制剂和PI3K抑制剂。
例如,所述药物组合可以包括:(i)TGFβ抑制剂,所述TGFβ抑制剂包括:抗TGFβ抗体或其抗原结合片段,小分子TGFβ抑制剂或其组合,所述TGFβ抗体或其抗原结合片段包 括Fresolimumab或其抗原结合片段,所述小分子TGFβ抑制剂包括SB525334、SD-208、SB431542、LY2109761、LY2157299(Galunisertib)、GW788388、RepSox、SIS3、LDN-193189、EW-7197,LY364947或其组合;(ii)PI3Kδ/γ双重抑制剂,所述PI3Kδ/γ双重抑制剂包括2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮,或其对映异构体、对映异构体的混合物、两种或更多种非对映异构体的混合物、或同位素变体;或其药学上可接受的盐、溶剂化物、水合物或前药。
又例如,所述药物组合可以包括:Fresolimumab或其抗原结合片段和(S)-2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮或其药学上可接受的盐、溶剂化物、水合物或前药。
又例如,所述药物组合可以包括:Fresolimumab或其抗原结合片段、SB431542和(S)-2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮或其药学上可接受的盐、溶剂化物、水合物或前药。
在一个实施方式中,所述药物组合可以包括所述PD-1抑制剂、TGFβ抑制剂和PI3K抑制剂。
例如,所述药物组合可以包括:(i)抗PD-1抗体或其抗原结合片段,所述抗PD-1抗体或其抗原结合片段包括纳武单抗、帕姆单抗、皮地利珠单抗或AMP-514或其抗原结合片段;(ii)PI3Kδ/γ双重抑制剂,所述PI3Kδ/γ双重抑制剂包括2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮,或其对映异构体、对映异构体的混合物、两种或更多种非对映异构体的混合物、或同位素变体;或其药学上可接受的盐、溶剂化物、水合物或前药;(iii)抗TGFβ抗体或其抗原结合片段,小分子TGFβ抑制剂或其组合,所述TGFβ抗体或其抗原结合片段包括Fresolimumab或其抗原结合片段,所述小分子TGFβ抑制剂包括SB525334、SD-208、SB431542、LY2109761、LY2157299(Galunisertib)、GW788388、RepSox、SIS3、LDN-193189、EW-7197,LY364947或其组合。
又例如,所述药物组合可以包括:(i)选自下组的抗PD-1抗体:纳武单抗、帕姆单抗、皮地利珠单抗或AMP-514或其组合;(ii)(S)-2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮或其药学上可接受的盐、溶剂化物、水合物或前药;(iii)Fresolimumab或其抗原结合片段和/或SB431542。
在一个实施方式中,所述药物组合可以包括所述PD-L1抑制剂、TGFβ抑制剂和PI3K抑制剂。
例如,所述药物组合可以包括:(i)抗PD-L1抗体或其抗原结合片段,所述抗PD-L1抗 体或其抗原结合片段包括Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab或其抗原结合片段;(ii)PI3Kδ/γ双重抑制剂,所述PI3Kδ/γ双重抑制剂包括2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮,或其对映异构体、对映异构体的混合物、两种或更多种非对映异构体的混合物、或同位素变体;或其药学上可接受的盐、溶剂化物、水合物或前药;(iii)抗TGFβ抗体或其抗原结合片段,所述TGFβ抗体或其抗原结合片段包括Fresolimumab或其抗原结合片段。
又例如,所述药物组合可以包括:(i)选自下组的抗PD-L1抗体:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab或其组合;(ii)(S)-2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮或其药学上可接受的盐、溶剂化物、水合物或前药;(iii)Fresolimumab或其抗原结合片段。
在一个实施方式中,所述药物组合可以包括所述PD-L1/TGFβ双重抑制剂和PI3K抑制剂。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂可以是PCT/CN2019124535描述的那些,将其所有的全部内容通过引用并入本申请。例如,所述PD-L1/TGFβ双重抑制剂可以是WBP1126(又称为CN202),或其变体或生物类似物。所述WBP1126可以包含两条多肽,所述多肽自N末端至C末端依次包含抗PD-L1抗体的重链可变区(VH)、CH2、CH3结构域以及TGFβRII或其功能活性片段,所述CH3结构域的C末端与所述TGFβRII或其功能活性片段N末端通过连接子相连接;例如,所述多肽可以具有表1所示的氨基酸序列:
表1
Figure PCTCN2022075642-appb-000012
Figure PCTCN2022075642-appb-000013
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂包含第一多肽和第二多肽,其中所述第一多肽至少包含:(i)抗PD-L1抗体的重链可变区;和(ii)TGFβRII或其功能活性片段;其中所述第二多肽至少包含抗PD-L1抗体的轻链可变区;其中所述第一多肽的重链可变区和所述第二多肽的轻链可变区在组合时能够特异性结合PD-L1。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂还包含连接子,所述连接子将所述抗PD-L1的抗体或其抗原结合片段的重链可变区的C末端与所述TGFβRII或其功能活性片段的N末端相连接。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂包含第一多肽和第二多肽,其中:
所述第一多肽自N末端至C末端依次包含抗PD-L1抗体的重链可变区(VH)、CH1 结构域以及TGFβRII或其功能活性片段,所述CH1结构域的C末端与所述TGFβRII或其功能活性片段N末端通过连接子相连接;和所述第二多肽自N末端至C末端包含抗PD-L1抗体的轻链可变区(VL)和轻链恒定区(CL)。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂包含第一多肽和第二多肽,其中:
所述第一多肽自N末端至C末端依次包含抗PD-L1抗体的重链可变区(VH)、CH1、CH2、CH3结构域以及TGFβRII或其功能活性片段,所述CH3结构域的C末端与所述TGFβRII或其功能活性片段N末端通过连接子相连接;和所述第二多肽自N末端至C末端包含抗PD-L1抗体的轻链可变区(VL)、轻链恒定区(CL)。
在某些实施方式中,其中所述连接子为肽连接子。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂的第一多肽可以与以下分子的具有相同功能的多肽具有至少约80%序列同一性:M7824或SHR-1701。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂的第二多肽可以与以下分子的具有相同功能的多肽具有至少约80%序列同一性:M7824或SHR-1701。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂可以是PCT/EP2015052781,PCT/CN2018086451描述的那些,将其所有的全部内容通过引用并入本申请。
在某些实施方式中,其中所述PD-L1/TGFβ双重抑制剂可以选自M7824、SHR-1701,或其变体或生物类似物,或其组合。
在某些实施方式中,所述PD-L1/TGFβ双重抑制剂与所述PI3K抑制剂可以在所述药物组合中不互相混合。
在某些实施方式中,所述PD-L1/TGFβ双重抑制剂与所述PI3K抑制剂可以各自独立地存在于单独的容器中。
药物组合物及用途
另一方面,本申请提供一种药物组合物,所述药物组合包括前述的PI3K抑制剂和第二治疗剂,以及任选的一种或多种药学上可接受的载体或赋形剂。
在某些实施方式中,其中所述第二治疗剂为前述的PD-L1/TGFβ双重抑制剂。
在某些实施方式中,其中所述PI3K抑制剂为前述的PI3Kδ/γ双重抑制剂。
在某些实施方式中所述药物组合物包括PD-L1/TGFβ双重抑制剂和PI3Kδ/γ双重抑制剂。
例如,所述药物组合物可以包括:i)WBP1126或其变体或生物类似物;和
ii)Tenalisib或其药学上可接受的盐、溶剂化物、水合物或前药。
在某些实施方式中,其中所述第二治疗剂为前述的抗PD-L1抗体,所述PI3K抑制剂为前述的PI3Kδ/γ双重抑制剂。
例如,所述药物组合物可以包括:i)纳武单抗、帕姆单抗、皮地利珠单抗或它们的组合;和ii)Tenalisib或其药学上可接受的盐、溶剂化物、水合物或前药。
在某些实施方式中,其中所述第二治疗剂为前述的PD-L1/TGFβ双重抑制剂,所述PI3K抑制剂为PI3Kα抑制剂。
例如,所述药物组合物可以包括:i)WBP1126或其变体或生物类似物;和
ii)Alplisib或其药学上可接受的盐、溶剂化物、水合物或前药。
在某些实施方式中,其中所述第二治疗剂与所述PI3K抑制剂存在于单一或分开的剂型中。
在某些实施方式中,其中所述的药物组合物包括,每单位剂型(unit dosage form)中包含1-1000mg剂量的第一治疗剂,例如PI3K抑制剂活性物质(例如CN401(Tenalisib)),例如1、2、3、4、5、6、7、8、9、10、11、12、13、14、15、16、17、18、19、20、25、30、40、50、60、70、80、90、100、110、120、130、140、150、160、170、180、190、200、250、300、350、400、450、500、600、700、800、900、1000mg或以上任意两个数值之间的值;以及,每单位剂型(unit dosage form)中包含1-1000mg剂量的第二治疗剂,例如PD-L1抑制剂、TGFβ抑制剂、PD-L1抑制剂、PD-L1/TGFβ双重抑制剂(例如CN202(WBP1126))或PD-1/TGFβ双重抑制剂中的任意一种或其组合,例如1、2、3、4、5、6、7、8、9、10、11、12、13、14、15、16、17、18、19、20、25、30、40、50、60、70、80、90、100、110、120、130、140、150、160、170、180、190、200、250、300、350、400、450、500、600、700、800、900、1000mg或以上任意两个数值之间的值。
另外的联合疗法
在某些实施方式中,本申请的药物组合可以与可用于治疗、预防或改善增生性病症、疾病或病况的一种或多种第三活性物质或疗法联合或组合使用。如果需要,将本申请的药物组合与任何常规的抗肿瘤疗法组合施用,这些常规的抗肿瘤疗法包括但不限于免疫疗法、治疗性抗体、靶向治疗、手术、放射治疗,或化疗。
活性物质可以是化学治疗药物,例如,烷化剂、抗代谢物、蒽环霉素、植物生物碱、拓扑异构酶抑制剂、抗瘤抗生素、激素类药、抗血管生成剂、分化诱导剂、细胞生长停滞诱导剂、细胞凋亡诱导剂、细胞毒性类药物和其他抗肿瘤药物。这些药物会以某种方式影响细胞 分裂或DNA合成和功能。代表性的化疗药包括但不限于烷化剂(如顺铂、卡铂、奥沙利铂、二氯甲基二乙胺、环磷酰胺、苯丁酸氮芥、达卡巴嗪、洛莫司汀、卡莫司汀、丙卡巴肼、苯丁酸氮芥和异环磷酰胺),抗代谢类(如氟尿嘧啶(5-FU)、吉西他滨、甲氨蝶呤、阿糖胞苷、氟达拉滨和氟尿嘧啶),抗有丝分裂类(包括紫杉烷类(如紫杉醇和多西他赛(decetaxel))和长春花生物碱类(如长春碱、长春新碱、长春花碱、长春瑞滨和长春地辛),蒽环类(包括多柔比星、柔红霉素、戊柔比星(valrubicin)、伊达比星和表柔比星,以及放线菌素D等放线菌素类),细胞毒性抗生素(包括丝裂霉素、普鲁卡霉素和博来霉素)和拓扑异构酶抑制剂(包括喜树碱类(如伊立替康和拓扑替康)和表鬼臼毒素衍生物(如安吖啶、依托泊苷、磷酸依托泊苷和替尼泊苷)。
在一些实施方式中,所述的第三活性物质可以选自长春新碱、长春碱、长春地辛、依托泊苷、多西他赛、紫杉醇、伊立替康、长春瑞滨、米托蒽醌、长春氟宁、拓扑替康中的任意一种或其任意地组合。
在某些实施方案中,可以与本文提供的方法联合使用的第三疗法包括但不限于手术、放射疗法、内分泌疗法、生物反应调节剂(例如,干扰素、白介素和肿瘤坏死因子(TNF))、过热和冷冻疗法,和减弱任何不良反应的药物(例如,止吐药)。
治疗方法和用途
另一方面,本申请提供一种治疗和/或预防肿瘤的方法,其包括向有需要的受试者施用:有效量的前述的药物组合,或前述的药物组合物。
在某些实施方式中,所述方法包括向有需要的受试者施用:有效量的前述的PI3K抑制剂和前述的免疫检查点的抑制剂。在某些实施方式中,所述方法包括向有需要的受试者施用:有效量的前述的PI3K抑制剂和前述的TGFβ抑制剂。在某些实施方式中,所述方法包括向有需要的受试者施用:有效量的前述的PI3K抑制剂、前述的TGFβ抑制剂和前述的免疫检查点的抑制剂。
例如,所述方法可以包括向有需要的受试者施用:有效量的前述的PI3K抑制剂,例如前述的(S)-2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮或其药学上可接受的盐和前述的抗PD-L1抗体,例如,Atezolizumab、Avelumab、BMS-936559、MPDL3280A、durvalumab,或它们的组合。
例如,所述方法可以包括向有需要的受试者施用:有效量的前述的PI3K抑制剂,例如前述的(S)-2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮或其药学上可接受的盐和前述的TGFβ抑制剂,例如,Fresolimumab,SB525334或它们的组合。
例如,所述方法可以包括向有需要的受试者施用:有效量的前述的PI3K抑制剂,例如 前述的(S)-2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮或其药学上可接受的盐;前述的TGFβ抑制剂,例如,Fresolimumab,SB525334或它们的组合;前述的抗PD-L1抗体,例如,Atezolizumab、Avelumab、BMS-936559、MPDL3280A、durvalumab,或它们的组合。
例如,所述方法可以包括向有需要的受试者施用:有效量的前述的PI3K抑制剂,例如前述的(S)-2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮或其药学上可接受的盐;前述的TGFβ抑制剂,例如,Fresolimumab,SB525334或它们的组合;前述的抗PD-1抗体,例如,纳武单抗、帕姆单抗、皮地利珠单抗,或它们的组合。
在某些实施方式中,所述方法包括向有需要的受试者施用:有效量的前述的PI3K抑制剂和前述的PD-L1/TGFβ双重抑制剂。
例如,所述方法可以包括向有需要的受试者施用:有效量的前述的PI3K抑制剂,例如前述的(S)-2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮;以及前述的PD-L1/TGFβ双重抑制剂,例如WBP1126,M7824、SHR-1701,或其变体或生物类似物,或其组合。
在某些实施方式中,其中所述PI3K抑制剂可以与所述PD-L1/TGFβ双重抑制剂同时施用。
在某些实施方式中,其中所述PI3K抑制剂可以在所述PD-L1/TGFβ双重抑制剂之后施用。
在某些实施方式中,其中所述PI3K抑制剂可以在所述PD-L1/TGFβ双重抑制剂之前施用。
在某些实施方式中,还包括向有需要的受试者施用有效量的前述的小分子TGFβ抑制剂。
在某些实施方式中,其中所述肿瘤可以包括实体瘤和非实体肿瘤。
在某些实施方式中,其中所述肿瘤可以包括PI3K表达异常的肿瘤。
在某些实施方式中,其中所述肿瘤可以包括PD-L1表达异常的肿瘤。
在某些实施方式中,其中所述肿瘤可以包括TGFβ表达异常的肿瘤。
在某些实施方式中,所述肿瘤可以包括消化道肿瘤、黑素瘤或淋巴瘤。
在一些实施方案中,本申请的方法与单独使用药剂或抑制剂中的任一种作为单一疗法治疗对象时通常产生抗性的时间相比,抗性延迟。在一些实施方案中,抗性延迟至少2周,例如,至少2周、4周、1个月、2个月、3个月、4个月、5个月、6个月、8个月、10个月、12个月、1年、2年、4年、6年、8年或更长。在一些实施方案中,根据该方法,与单 独使用药剂或抑制剂中的任一种作为单一疗法治疗对象时缓解通常持续的时间相比,缓解(例如,完全缓解或部分缓解)延长。在一些实施方案中,缓解(例如,完全缓解或部分缓解)延长至少2周、例如,至少2周、4周、1个月、2个月、3个月、4个月、5个月、6个月、8个月、10个月、12个月、1年、2年、4年、6年、8年或更长。
在一些实施方案中,向治疗方案中添加PI3K抑制剂或第二药剂增加或恢复对癌症抗性的药剂的敏感性。例如,在一些实施方案中,向治疗方案中添加第二药剂增加或恢复对癌症抗性的PI3K抑制剂的敏感性。
在某些实施方式中,其中所述受试者中的癌症的缓解延长。
在某些实施方式中,其中所述受试者经历癌症的完全缓解。
在某些实施方式中,其中最小残留疾病(MRD)的水平降低。
在一些实施方案中,本申请所述的方法包括选择用PI3K抑制剂和第二药剂的组合治疗的对象。在某些实施方案中,基于对象中的MRD选择用组合治疗的对象(例如,患有癌症,例如本申请所述的癌症的患者)。在某些实施方案中,选择基于存在的MRD在预选水平以上(例如,100个正常细胞中有1个恶性肿瘤细胞,1000个正常细胞中有1个恶性肿瘤细胞或10,000个正常细胞中有1个恶性肿瘤细胞)。用于监测最小残留疾病阴性(MRD)的方法是本领域已知的。See,例如,Zhou,J.等人,Blood,2007,110:1607-1611。这样的方法包括基于DNA的测试或基于RNA的测试。在某些实施方案中,使用流式细胞术、测序或PCR监测MRD。
另一方面,本申请提供了前述的药物组合或前述的药物组合物在制备用于治疗和/或预防肿瘤的药物中的用途。
例如,前述的PI3K抑制剂和前述的免疫检查点的抑制剂在制备用于治疗和/或预防肿瘤的药物中的用途。例如,前述的PI3K抑制剂和前述的TGFβ抑制剂在制备用于治疗和/或预防肿瘤的药物中的用途。例如,前述的PI3K抑制剂、前述的免疫检查点的抑制剂和前述的TGFβ抑制剂在制备用于治疗和/或预防肿瘤的药物中的用途。
另一方面,本申请提供了前述的药物组合或前述的药物组合物用于治疗和/或预防肿瘤。
另一方面,本申请提供了一种抑制肿瘤生长的方法,所述方法包括使肿瘤接触前述的药物组合或前述的药物组合物。
在某些实施方式中,所述方法包括使肿瘤接触前述的PI3K抑制剂和前述的免疫检查点的抑制剂。在某些实施方式中,所述方法包括使肿瘤接触前述的PI3K抑制剂和前述的 TGFβ抑制剂。在某些实施方式中,所述方法包括使肿瘤接触前述的PI3K抑制剂、前述的免疫检查点的抑制剂和前述的TGFβ抑制剂。
另一方面,本申请提供了一种抑制肿瘤生长的方法,所述方法包括使肿瘤接触前述的PI3K抑制剂与前述的PD-L1/TGFβ双重抑制剂。
另一方面,本申请提供了一种药盒,其包括:(1)第一容器,以及位于所述第一容器中的前述的PI3K抑制剂;(2)第二容器,以及位于所述第二容器中的前述的PD-L1/TGFβ双重抑制剂。
另一方面,本申请提供了一种药盒,其可以包括:(1)第一容器,以及位于所述第一容器中的前述的PI3K抑制剂;(2)第二容器,以及位于所述第二容器中的前述的免疫检查点的抑制剂或前述的TGFβ抑制剂。
在某些实施方式中,所述药盒还包括(3)第三容器,以及位于第三容器中的前述的免疫检查点的抑制剂或前述的TGFβ抑制剂,所述第三容器中的药剂与第二容器中的药剂不同。
在某些实施方式中,所述药盒中药物的剂型可以为口服剂型或注射剂型。
在某些实施方式中,所述的药盒还可以含有说明书。
不欲被任何理论所限,下文中的实施例仅仅是为了阐释本申请的药物组合、制备方法和用途等,而不用于限制本申请发明的范围。
实施例
材料和方法
本动物实验按照AAALAC要求进行,并经过必凯动物实验中心IACUC批准进行此动物实验。从北京维通利华实验动物技术有限公司采购6-8周龄的Balb/c小鼠(16-20克),每只老鼠皮下接种50万个A20细胞,接种在老鼠的背部右后侧。当肿瘤的体积生长到约60mm 3左右开始随机分配至对照组和实验组。
当观察到病态或体重减轻≥20%时,使小鼠安乐死。用卡尺每周两次测量肿瘤直至最终处死。当肿瘤大小达到约2000mm 3或存在动物健康问题(肿瘤面积的20%溃疡)时,动物将被安乐死并记录死亡日期。从二维肿瘤测量结果估计实体肿瘤体积并根据以下等式进行计算:
TV:Tumor Volume.肿瘤体积。TV(mm 3)=[长度(mm)x宽度 2(mm 2)]/2
然后给定日的组的百分比中值消退(regression)通过取得针对该组中每只动物在该日计算的个体百分比消退的中值来获得。计算的日期是在计算ΔT/ΔC(即,实验组和实验组之间的肿瘤体积从基线变化的中值的比例)的那天确定的,除非中值百分比消退不代表该组的活 性。在那种情况下,当中值百分比消退最大时的第一天确定为这一天。如果肿瘤体积降至在治疗开始时肿瘤体积的50%,则将消退定义为部分的(PR)。当肿瘤体积低于14mm 3或无法记录时,认为已经实现了完全消退(CR)。
RTV:Relative Tumor Volume.相对肿瘤体积。RTV=V t/V 0。V t:一个实验周期结束时的(treatment组)肿瘤体积。V 0:实验开始时的肿瘤体积
TGI:Tumor Growth Inhibition value.TGI=[1-RTV(实验组)/RTV(对照组)]*100%
统计学分析
对与基线相比的肿瘤体积变化进行具有因子治疗和天数(重复)的双向ANOVA类型。在显著治疗*天数相互作用或治疗效果的情况下,随后进行带有Bonferroni-Holm校正多重性的对比分析以在第8天至第27天的每一天比较所有的实验组与对照组。针对每只动物并且在每天计算肿瘤体积相对于基线的变化,其通过从指定的观察日的肿瘤体积中减去第一次治疗当天的肿瘤体积计算。因为在组之间观察到差异的异质性,所以对于ANOVA类型模型(SAS Institute Inc.(2008)SAS/STAT 9.2用户指南,Cary NC)选择了具有组=选项的复合对称(CS)协方差结构。所有统计分析均使用SAS版本v9.2软件进行。小于5%的概率(p<0.05)被认为是显著的。
实施例1 PI3K抑制剂与不同剂量的TGFβ/PDL1双特异性抗体的联合用药研究
实施例1.1 CN401(Tenalisib)与CN202(WBP1126)(5mg/kg)在A20小鼠B细胞淋巴瘤模型中联合用药研究(试验设计见表2)
6-8周龄balb/c小鼠(18-20g)购自北京维通利华实验动物技术有限公司,每只老鼠皮下接种50万个A20细胞,接种在老鼠的背部右后侧。当肿瘤的体积生长到约62mm 3时开始给药治疗。其中,CN401以150mg/kg每天两次口服给药,持续3周。
CN202是通过腹腔注射给药的,每周3次,剂量为5mg/kg,持续3周。对照组接受不含活性产品的载体(0.5%MC和PBS)。每周测量三次肿瘤大小和体重。本动物实验按照AAALAC要求进行,并经过必凯动物实验中心IACUC批准进行此动物实验。
表2
组别 数量 给药方式
1 8 空白对照组(PBS i.p.tiw×3weeks+0.5%MC p.o.bid×3weeks)
2 8 CN401 150mg/kg,p.o.bid×3weeks
3 8 CN202 5mg/kg,i.p.tiw×3weeks
4 8 CN401 150mg/kg,p.o.bid×3weeks+CN202 5mg/kg,i.p.tiw×3weeks
肿瘤生长曲线和体重生长曲线的结果见图1A-图1F;在第22天与空白组相比,治疗组的TGI、p值和完全反应小鼠的数量显示在表3中。
结果:在本研究中,CN401与CN202联合在A20小鼠B细胞淋巴瘤模型中对肿瘤抑制有协同作用(p=0.0001,TGI=89%,CR=4/8),小鼠对给药方案表现出良好的耐受性。
表3
Figure PCTCN2022075642-appb-000014
实施例1.2 CN401与CN202(15mg/kg)在A20小鼠B细胞淋巴瘤模型中联合用药研究(试验设计见表4)
参照实施例1.1的试验方法,不同之处在于CN202的给药剂量由5mg/kg替换为15mg/kg。
表4
组别 数量 给药方式
1 8 空白对照组(PBS i.p.tiw×3weeks+0.5%MC p.o.bid×3weeks)
2 8 CN401 150mg/kg,p.o.bid×3weeks
3 8 CN202 15mg/kg,i.p.tiw×3weeks
4 8 CN401 150mg/kg,p.o.bid×3weeks+CN202 15mg/kg,i.p.tiw×3weeks
肿瘤生长曲线和体重生长曲线的结果见图2A-图2F;在第22天与空白组相比,治疗组的TGI、p值和完全反应小鼠的数量显示在表5中。
结果:在本研究中,CN401与CN202联合在A20小鼠B细胞淋巴瘤模型中对肿瘤抑制有协同作用(p=0.023,TGI=71%,CR=5/8),小鼠对给药方案表现出良好的耐受性。
如图3A-3B所示,CN401与不同剂量的的CN202(5mg/kg,15mg/kg)联合均对肿瘤抑制产生了协同作用,并且对小鼠体重无显著影响,表现出良好的耐受性。
表5
Figure PCTCN2022075642-appb-000015
Figure PCTCN2022075642-appb-000016
实施例2 TGFβ/PDL1双特异性抗体与不同形式的PI3K抑制的联合用药研究
实施例2.1 CN202(WBP1126)与CN401(Tenalisib)的联合用药研究(试验设计见表6)
6-8周龄balb/c小鼠(18-20g)购自北京维通利华实验动物技术有限公司,每只老鼠皮下接种50万个A20细胞,接种在老鼠的背部右后侧。当肿瘤的体积生长到约80mm 3时开始给药治疗。其中,CN202是通过腹腔注射给药的,每周3次,剂量为5mg/kg,持续3周。CN401以150mg/kg每天两次口服给药,持续3周。对照组接受不含活性产品的载体(0.5%MC和PBS)。每周测量三次肿瘤大小和体重。本动物实验按照AAALAC要求进行,并经过必凯动物实验中心IACUC批准进行此动物实验。
表6
组别 数量 给药方式
1 7 空白对照组(PBS i.p.tiw×3weeks+0.5%MC p.o.bid×3weeks)
2 7 CN202 5mg/kg i.p.tiw×3weeks
3 7 CN401 150mg/kg p.o.bid×3weeks
4 7 CN202 5mg/kg i.p.tiw×3weeks+CN401 150mg/kg p.o.bid×3weeks
肿瘤生长曲线和体重生长曲线的结果见图4A-图4F;在第21天与空白组相比,治疗组的TGI、p值和完全反应小鼠的数量显示在表7中。
结果:在本研究中,CN202与CN401联合在A20小鼠B细胞淋巴瘤模型中对肿瘤抑制有协同作用(p=0.0009,TGI=85%,CR=6/7),小鼠对给药方案表现出良好的耐受性。
表7
Figure PCTCN2022075642-appb-000017
实施例2.2 CN202(WBP1126)与Alpelisib的联合用药研究(试验设计见表8)
参照实施例2.1的试验方法,不同之处在于PI3K抑制剂由CN401(Tenalisib)的给药方式以150mg/kg每天两次口服给药,持续3周替换为以50mg/kg每天一次口服给药Alpelisib,持续3 周。
表8
组别 数量 给药计划
1 7 空白对照组(PBS i.p.tiw×3weeks+0.5%MC p.o.bid×3weeks)
2 7 CN202 5mg/kg i.p.tiw×3weeks
3 7 Alpelisib 50mg/kg p.o.qd×3weeks
4 7 CN202 5mg/kg i.p.tiw×3weeks+Alpelisib 50mg/kg p.o.qd×3weeks
肿瘤生长曲线和体重生长曲线的结果见图5A-图5F;在第21天与空白组相比,治疗组的TGI、p值和完全反应小鼠的数量显示在表9中。
结果:在本研究中,CN202与Alpelisib联合在A20小鼠B细胞淋巴瘤模型中对肿瘤抑制有协同作用(p=0.0002,TGI=85%and CR=3/7),小鼠对给药方案同样表现出良好的耐受性。
表9
Figure PCTCN2022075642-appb-000018
实施例3 CN401与抗PD-L1抗体在A20小鼠B细胞淋巴瘤模型中联合用药研究(试验设计见表10)
6-8周龄balb/c小鼠(18-20g)购自北京维通利华实验动物技术有限公司,每只老鼠皮下接种50万个A20细胞,接种在老鼠的背部右后侧。当肿瘤的体积生长到约62mm 3时开始给药治疗。CN401以150mg/kg每天两次口服给药,持续3周。其中,抗PD-L1抗体(Atezolizumab)是通过腹腔注射给药的,每周2次,剂量为5mg/kg,持续3周。对照组接受不含活性产品的载体(0.5%MC和PBS)。每周测量三次肿瘤大小和体重。本动物实验按照AAALAC要求进行,并经过必凯动物实验中心IACUC批准进行此动物实验。
表10
Figure PCTCN2022075642-appb-000019
Figure PCTCN2022075642-appb-000020
肿瘤生长曲线和体重生长曲线的结果见图6A-图6F;在第22天与空白组相比,治疗组的TGI、p值和完全反应小鼠的数量显示在表11中。
结果:在本研究中,CN401与抗PD-L1抗体联合在A20小鼠B细胞淋巴瘤模型中对肿瘤抑制有协同作用(p=0.026,TGI=63%,CR=3/8),小鼠对给药方案表现出良好的耐受性。
表11
Figure PCTCN2022075642-appb-000021
实施例4 CN202与白蛋白紫杉醇(nab-pac)联合用药以及CN401、CN202与白蛋白紫杉醇(nab-pac)联合用药在EMT-6肿瘤模型中联合用药研究(试验设计见表12)
6-8周龄的balb/c小鼠(18-20g)购自北京维通利华实验动物技术有限公司,每只小鼠接种50万个EMT-6细胞(小鼠三阴性乳腺癌细胞),接种于小鼠的胸部左侧从上往下数第2-3对乳腺脂肪垫。当肿瘤的体积生长到约87mm 3时开始分组给药治疗。CN401以150mg/kg每天两次口服给药,持续4周。CN202和白蛋白紫杉醇通过腹腔注射给药。CN202每周给药3次,剂量为15mg/kg,持续2周;同样的剂量,CN202每周1次,持续2周。白蛋白紫杉醇每周给药2次,剂量为10mg/kg,持续2周。对照组接受不含活性产品的载体(0.5%MC和PBS)。每周测量三次肿瘤大小和体重。本动物实验按照AAALAC要求进行,并经过必凯动物实验中心IACUC批准进行此动物实验。
表12
Figure PCTCN2022075642-appb-000022
Figure PCTCN2022075642-appb-000023
肿瘤生长曲线和体重生长曲线的结果见图7-8。
结果:本研究发现在EMT-6小鼠三阴性乳腺癌原位模型中,CN202与白蛋白紫杉醇联合用药对肿瘤抑制有一定的协同作用(p=0.06,TGI=48%(在14天),CR=3/8(在80天));CN401,CN202与白蛋白紫杉醇联合用药对肿瘤抑制有很好的协同作用(p=0.02,TGI=56%(在14天),CR=4/8(在80天))。小鼠对给药方案表现出良好的耐受性。肿瘤生长曲线和体重生长曲线的结果见图7A-7B,根据必凯动物实验中心IACUC规定,个体肿瘤体积≥2000mm 3需实施安乐死,结果见图8A-8G;在第14天与空白组相比,治疗组的TGI、p值和完全反应小鼠的数量(观察至80天)显示在表13中。
表13
Figure PCTCN2022075642-appb-000024

Claims (105)

  1. 一种药物组合,包含磷酸肌醇3-激酶(PI3K)抑制剂和第二治疗剂,其中所述第二治疗剂为免疫检查点的抑制剂,TGFβ抑制剂,双功能免疫检查点/TGFβ抑制剂或它们的组合。
  2. 根据权利要求1所述的药物组合,其中所述免疫检查点的抑制剂包括能够阻断程序性死亡1(PD-1)与程序性死亡配体1(PD-L1)相互作用的试剂。
  3. 根据权利要求1-2中任一项所述的药物组合,其中所述免疫检查点的抑制剂包括程序性死亡配体1(PD-L1)和/或程序性死亡1(PD-1)的抑制剂。
  4. 根据权利要求1-3中任一项所述的药物组合,其中所述双功能免疫检查点/TGFβ抑制剂包括PD-L1/TGFβ双重抑制剂,或PD-1/TGFβ双重抑制剂。
  5. 根据权利要求1-4中任一项所述的药物组合,所述药物组合包括:
    1)PD-L1抑制剂和PI3K抑制剂的组合;
    2)TGFβ抑制剂和PI3K抑制剂的组合;
    3)PD-1抑制剂、TGFβ抑制剂和PI3K抑制剂的组合;
    4)PD-L1抑制剂、TGFβ抑制剂和PI3K抑制剂的组合;
    5)PD-L1/TGFβ双重抑制剂和PI3K抑制剂的组合;或
    6)PD-1/TGFβ双重抑制剂和PI3K抑制剂的组合。
  6. 根据权利要求1-5中任一项所述的药物组合,其中所述免疫检查点的抑制剂包括核酸,抗体或其抗原结合片段,融合蛋白或化合物。
  7. 根据权利要求6所述的药物组合,其中所述抗体选自:人抗体、人源化抗体、嵌合抗体、多特异性抗体、单克隆抗体和多克隆抗体。
  8. 根据权利要求6-7中任一项所述的药物组合,其中所述抗原结合片段选自:Fab、Fab’、F(ab’) 2、Fv、scFv、双抗体、Fd、dAb、VHH、大抗体和互补决定区(CDR)片段。
  9. 根据权利要求6-8中任一项所述的药物组合,其中所述抗体或其抗原结合片段包含含有HCDR1,HCDR2和HCDR3的重链可变区(VH)。
  10. 根据权利要求6-9中任一项所述的药物组合,其中所述抗体或其抗原结合片段还包含或不包含CH1结构域。
  11. 根据权利要求10所述的药物组合,其中所述抗体或其抗原结合片段还包含或不包含CH2和CH3结构域。
  12. 根据权利要求6-9中任一项所述的药物组合,其中所述抗体或其抗原结合片段还包含抗体重链恒定区。
  13. 根据权利要求12所述的药物组合,其中所述抗体重链恒定区为人抗体重链恒定区。
  14. 根据权利要求13所述的药物组合,其中所述人抗体重链恒定区选自源自下组的恒定区:IgG1、IgG2、IgG3、IgG4和它们的变体。
  15. 根据权利要求6-14中任一项所述的药物组合,其中所述抗体或其抗原结合片段还包含含有LCDR1,LCDR2,LCDR3的轻链可变区(VL)。
  16. 根据权利要求15所述的药物组合,其中所述抗体或其抗原结合片段还包含轻链恒定区(CL)。
  17. 根据权利要求1-16中任一项所述的药物组合,其中所述免疫检查点的抑制剂包含抗PD-L1抗体或其抗原结合片段,或抗PD-1抗体或其抗原结合片段。
  18. 根据权利要求17所述的药物组合,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包含HCDR1,HCDR2,HCDR3,它们各自分别与以下分子的HCDR1,HCDR2,HCDR3具有至少约80%的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A(RG7446)或durvalumab(MEDI-4736)。
  19. 根据权利要求17-18中任一项所述的药物组合,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区分别与以下分子的重链可变区具有至少约80%的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab。
  20. 根据权利要求17-19中任一项所述的药物组合物,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链,其中所述重链分别与以下分子的重链具有至少约80%的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab。
  21. 根据权利要求17-20中任一项所述的药物组合,其中所述抗PD-L1抗体或其抗原结合片段还包含抗体的轻链可变区,其中所述轻链可变区包含LCDR1,LCDR2,LCDR3,它们各自分别与以下分子的LCDR1,LCDR2,LCDR3具有至少约80%的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab。
  22. 根据权利要求17-21中任一项所述的药物组合物,其中所述抗PD-L1抗体或其抗原结合片段还包含抗体的轻链可变区,其中所述轻链可变区分别与以下分子的轻链可变区具有至少约80%的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab。
  23. 根据权利要求17-22中任一项所述的药物组合物,其中所述抗PD-L1抗体或其抗原结合片段还包含抗体的轻链,其中所述轻链分别与以下分子的轻链具有至少约80%的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab。
  24. 根据权利要求17-23中任一项所述的药物组合,其中所述抗PD-L1抗体或其抗原结合片 段包括:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab,或其抗原结合片段,或其变体或生物类似物,或其组合。
  25. 根据权利要求17所述的药物组合,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包括一个或多个选自下组的重链可变区CDRs(HCDRs):(a)具有与SEQ ID NO:1所示氨基酸序列至少约70%序列同一性的HCDR1;(b)具有与SEQ ID NO:2至少约70%序列同一性的HCDR2;和(c)具有与SEQ ID NO:3至少约70%序列同一性的HCDR3。
  26. 根据权利要求25所述的药物组合,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包括一个或多个选自下组的HCDRs:(a)具有SEQ ID NO:1所示氨基酸序列的HCDR1或者通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:1所示氨基酸序列不超过2个氨基酸差异的HCDR1;(b)具有SEQ ID NO:2所示氨基酸序列的HCDR2或者通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:2所示氨基酸序列不超过2个氨基酸差异的HCDR2;和(c)具有SEQ ID NO:3所示氨基酸序列的HCDR3或者通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:3所示氨基酸序列不超过2个氨基酸差异的HCDR3。
  27. 根据权利要求17所述的药物组合,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包含HCDR1,HCDR2,HCDR3,所述HCDR1包含与SEQ ID NO:1所示氨基酸序列至少约70%序列同一性的氨基酸序列,所述HCDR2包含与SEQ ID NO:2所示氨基酸序列至少约70%序列同一性的氨基酸序列,所述HCDR3包含与SEQ ID NO:3所示氨基酸序列至少约70%序列同一性的氨基酸序列。
  28. 根据权利要求27所述的药物组合,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包含HCDR1,HCDR2,HCDR3,所述HCDR1包含SEQ ID NO:1所示的氨基酸序列或通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:1所示的氨基酸序列不超过2个氨基酸差异的氨基酸序列;所述HCDR2包含SEQ ID NO:2所示的氨基酸序列或通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:2所示的氨基酸序列不超过2个氨基酸差异的氨基酸序列;所述HCDR3包含SEQ ID NO:3所示的氨基酸序列或通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:3所示的氨基酸序列不超过2个氨基酸差异的氨基酸序列。
  29. 根据权利要求28所述的药物组合,其中所述抗PD-L1抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包含选自下组的氨基酸序列:(a)SEQ ID NO:4所示的 氨基酸序列;(b)与SEQ ID NO:4所示的氨基酸序列至少约85%,90%,95%或者99%序列同一性的氨基酸序列;和(c)通过加成、消除或取代反应所得到的与SEQ ID NO:4所示的氨基酸序列具有1个或更多差异的氨基酸序列。
  30. 根据权利要求1-29中任一项所述的药物组合,其中所述TGFβ抑制剂结合TGFβ1、TGFβ2或TGFβ3。
  31. 根据权利要求1-30中任一项所述的药物组合,其中所述TGFβ抑制剂结合TGFβ或转化生长因子β受体(TGFβR)。
  32. 根据权利要求1-31中任一项所述的药物组合,其中所述TGFβ抑制剂结合TGFβI型受体(TGFβRI),TGFβII型受体(TGFβRII)或TGFβIII型受体(TGFβRIII)。
  33. 根据权利要求1-32中任一项所述的药物组合,其中所述TGFβ抑制剂包括:1)抗TGFβ抗体或其抗原结合片段;2)抗TGFβR抗体或其抗原结合片段;3)小分子TGFβ抑制剂;4)包含转化生长因子β受体II(TGFβRII)或其功能活性片段、或其变体或生物类似物的蛋白质,或它们的组合。
  34. 根据权利要求33所述的药物组合,其中所述抗TGFβ抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区包含HCDR1,HCDR2,HCDR3,它们各自分别与Fresolimumab分子的HCDR1,HCDR2,HCDR3具有至少约80%的序列同一性。
  35. 根据权利要求33-34中任一项所述的药物组合,其中所述抗TGFβ抗体或其抗原结合片段包含抗体的重链可变区,其中所述重链可变区与Fresolimumab分子的重链可变区具有至少约80%的序列同一性。
  36. 根据权利要求33-35中任一项所述的药物组合,其中所述抗TGFβ抗体或其抗原结合片段包含抗体的重链,其中所述重链与Fresolimumab分子的重链具有至少约80%的序列同一性。
  37. 根据权利要求33-36中任一项所述的药物组合,其中所述抗TGFβ抗体或其抗原结合片段包含抗体的轻链可变区,其中所述轻链可变区包含LCDR1,LCDR2,LCDR3,它们各自分别与Fresolimumab分子的LCDR1,LCDR2,LCDR3具有至少约80%的序列同一性。
  38. 根据权利要求33-37中任一项所述的药物组合,其中所述抗TGFβ抗体或其抗原结合片段包含抗体的轻链可变区,其中所述轻链可变区与Fresolimumab分子的轻链可变区具有至少约80%的序列同一性。
  39. 根据权利要求33-38中任一项所述的药物组合,其中所述抗TGFβ抗体或其抗原结合片段 包含抗体的轻链,其中所述轻链与Fresolimumab分子的轻链具有至少约80%的序列同一性。
  40. 根据权利要求33-39中任一项所述的药物组合,其中所述抗TGFβ抗体或其抗原结合片段包括:Fresolimumab,或其抗原结合片段,或其变体或生物类似物。
  41. 根据权利要求33-40中任一项所述的药物组合,其中所述小分子TGFβ抑制剂包括以下化合物:SB525334、SD-208、SB431542、LY2109761、LY2157299、GW788388、RepSox、SIS3、LDN-193189、EW-7197、LY364947、或其组合。
  42. 根据权利要求33-41中任一项所述的药物组合,其中所述TGFβRII包括具有野生型人TGFβ受体2型同种型A序列的多肽,或具有野生型人TGFβ受体2型同种型B序列的多肽,或与其氨基酸序列具有基本相同的序列的多肽。
  43. 根据权利要求33-42中任一项所述的药物组合,其中所述TGFβRII或其功能活性片段包括人TGFβRII胞外域(ECD)或者与其氨基酸序列基本相同的序列。
  44. 根据权利要求43所述的药物组合,其中所述TGFβRII或其功能活性片段包含:
    (a)SEQ ID NO:6所示的氨基酸序列;
    (b)具有与SEQ ID NO:6所示的氨基酸序列至少约85%序列同一性的氨基酸序列;或
    (c)具有与SEQ ID NO:6所示的氨基酸序列相比,添加,缺失和/或取代一个或多个氨基酸的氨基酸序列。
  45. 根据权利要求1-44中任一项所述的药物组合,其中所述双功能免疫检查点/TGFβ抑制剂为融合蛋白。
  46. 根据权利要求4-45中任一项所述的药物组合,其中所述PD-L1/TGFβ双重抑制剂或PD-1/TGFβ双重抑制剂为融合蛋白。
  47. 根据权利要求4-46中任一项所述的药物组合,其中所述PD-L1//TGFβ双重抑制剂包含PD-L1靶向部分和TGFβ受体结构域,所述TGFβ受体结构域包含转化生长因子β受体II(TGFβRII)或其功能活性片段。
  48. 根据权利要求4-47中任一项所述的药物组合,其中所述PD-L1//TGFβ双重抑制剂包含多肽,其中所述多肽至少包含:(i)抗PD-L1抗体的重链可变区;和(ii)TGFβRII或其功能活性片段。
  49. 根据权利要求48所述的药物组合,其中所述抗PD-L1抗体重链可变区包含HCDR1,HCDR2,HCDR3;其中所述HCDR1包含与SEQ ID NO:1所示氨基酸序列至少约70%序列同一性的氨基酸序列,所述HCDR2包含与SEQ ID NO:2所示氨基酸序列至少约70% 序列同一性的氨基酸序列,且所述HCDR3包含与SEQ ID NO:3所示氨基酸序列至少约70%序列同一性的氨基酸序列;或
    其中所述HCDR1,HCDR2,HCDR3分别与以下分子的HCDR1,HCDR2,HCDR3具有至少约80%的序列同一性:Atezolizumab、Avelumab、BMS-936559、MPDL3280A(RG7446)或durvalumab(MEDI-4736)。
  50. 根据权利要求49所述的药物组合,其中所述抗PD-L1抗体重链可变区包含HCDR1,HCDR2,HCDR3,所述HCDR1包含SEQ ID NO:1所示的氨基酸序列或通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:1所示的氨基酸序列不超过2个氨基酸差异的氨基酸序列;所述HCDR2包含SEQ ID NO:2所示的氨基酸序列或通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:2所示的氨基酸序列不超过2个氨基酸差异的氨基酸序列;所述HCDR3包含SEQ ID NO:3所示的氨基酸序列或通过氨基酸加成、消除或者取代反应所得到的具有与SEQ ID NO:3所示的氨基酸序列不超过2个氨基酸差异的氨基酸序列。
  51. 根据权利要求50所述的药物组合,其中所述抗PD-L1抗体重链可变区包含选自下组的氨基酸序列:(a)SEQ ID NO:4所示的氨基酸序列;(b)与SEQ ID NO:4所示的氨基酸序列至少约85%,90%,95%或者99%序列同一性的氨基酸序列;(c)通过加成、消除或取代反应所得到的与SEQ ID NO:4所示的氨基酸序列具有1个或更多差异的氨基酸序列;和(d)与以下分子的重链可变区具有至少约80%的序列同一性的氨基酸序列:Atezolizumab、Avelumab、BMS-936559、MPDL3280A或durvalumab。
  52. 根据权利要求47所述的药物组合,其中所述TGFβRII或其功能活性片段包含:
    (a)SEQ ID NO:6所示的氨基酸序列;
    (b)具有与SEQ ID NO:6所示的氨基酸序列至少约85%序列同一性的氨基酸序列;或
    (c)具有与SEQ ID NO:6所示的氨基酸序列相比,添加,缺失和/或取代一个或多个氨基酸的氨基酸序列。
  53. 根据权利要求47所述的药物组合,其中所述多肽还包含连接子,所述连接子将所述抗PD-L1的抗体或其抗原结合片段的重链可变区的C末端与所述TGFβRII或其功能活性片段的N末端相连接。
  54. 根据权利要求47所述的药物组合,所述多肽还包括CH2、CH3结构域,所述多肽自N末端至C末端依次包含抗PD-L1抗体的重链可变区(VH)、CH2、CH3结构域和TGFβRII或其功能活性片段,所述CH3结构域的C末端与所述TGFβRII或其功能活性片段N末端通 过连接子相连接。
  55. 根据权利要求54所述的药物组合,其中所述CH2、CH3结构域源自IgG。
  56. 根据权利要求53-55中任一项所述的药物组合,其中所述连接子为肽连接子。
  57. 根据权利要求56所述的药物组合,其中所述肽连接子的氨基酸序列为(G 4S) x,其中x为3-6中的任意整数。
  58. 根据权利要求57所述的药物组合,其中所述肽连接子包含选自下组氨基酸序列:(a)SEQ ID NO:5所示的氨基酸序列;(b)具有与SEQ ID NO:5所示的氨基酸序列至少约85%,90%,95%或者99%序列同一性的氨基酸序列;和(c)通过加成、消除或取代反应所得到的与SEQ ID NO:5所示的氨基酸序列具有1个或更多差异的氨基酸序列。
  59. 根据权利要求48-58所述的药物组合,其中所述多肽包含选自下组的氨基酸序列:(a)SEQ ID NO:7所示的氨基酸序列;(b)具有与SEQ ID NO:7所示的氨基酸序列至少约85%,90%,95%或者99%序列同一性的氨基酸序列;(c)通过加成、消除或取代反应所得到的与SEQ ID NO:7所示的氨基酸序列具有1个或更多差异的氨基酸序列。
  60. 根据权利要求1-59中任一项所述的药物组合,其中所述PD-L1//TGFβ双重抑制剂包含两条权利要求48-59中任一项所述多肽。
  61. 根据权利要求1-60中任一项所述的药物组合,其中所述PI3K抑制剂为PI3Kδ/γ双重抑制剂。
  62. 根据权利要求1-61中任一项所述的药物组合,其中所述PI3K抑制剂包括式(IA-I)、(IA-II)、(IA-III)、或(IA-IV)所示的化合物:
    Figure PCTCN2022075642-appb-100001
    或它们的对映异构体、对映异构体的混合物、两种或更多种非对映异构体的混合物、同位素变体、药学上可接受的盐、溶剂化物、水合物或前药,其中
    每次出现的R独立地选自氢、卤素、-OR a、CN、经取代或未经取代的C 1-6烷基、经取代或未经取代的C 2-6烯基、经取代或未经取代的C 2-6炔基、经取代或未经取代的C 3-8环烷基和经取代或未经取代的杂环基团;
    R 1和R 2可相同或不同并且独立地选自氢、卤素和经取代或未经取代的C 1-6烷基,或与共有原子直接结合的R 1和R 2可连接形成=O基或经取代或未经取代的饱和或不饱和3-10元环,包括与R 1和R 2结合的碳原子,所述环可任选地包括一个或多个相同或不同且选自O、NR a和S的杂原子;
    Cy 1为选自经取代或未经取代的C 3-8环烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环基团、经取代或未经取代的C 6-20芳基和经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基的单环基团;
    每次出现的R a可相同或不同且独立地选自氢、卤素、羟基、氰基、经取代或未经取代的C 1-6烷基;-NR cR d,其中R c,R d独立地选自氢、卤素、羟基、氰基、经取代或未经取代的C 1-6烷基和C 1-6烷氧基,和-OR c,其中R c为经取代或未经取代的C 1-6烷基;
    n为1-4的整数;并且
    q为0、1或2;
    每次出现的X独立地选自CR 3或N;
    并且每次出现的R 3独立地选自氢、羟基、卤素、羧基、氰基、硝基、经取代或未经取代的C 1-8烷基、经取代或未经取代的C 1-8烷氧基、经取代或未经取代的C 2-10烯基、经取代或未经取代的C 2-12炔基、经取代或未经取代的C 6-20芳基、经取代或未经取代的C 6- 20芳基C 1-8烷基、经取代或未经取代的C 3-20环烷基、经取代或未经取代的C 3-20环烷基C 1-8烷基、经取代或未经取代的C 3-8环烯基C 1-8烷基、经取代或未经取代的C 3-8环烯基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基C 1-8烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环基C 1-8烷基环、经取代或未经取代的胍、-C00R x、-C(O)R x、-C(S)R x、-C(O)NR xR y、-C(O)ONR xR y,-NR yR z、-NR xCONR yR z,-N(R x)SOR y、-N(Rx)SO 2R y、-(=N-N(R x)R y)、-NR xC(O)OR y、-NR xR y、-NR xC(O)R y-、-NR xC(S)R y、-NR xC(S)NR yR z、-SONR xR y-、-SO 2NR xR y-、-OR x、-OR xC(O)NR yR z、-OR xC(O)OR y-、-OC(O)R x、-OC(O)NR xR y、-R xNR yC(O)R z、-R xOR y、-R xC(O)OR y、-R xC(O)NR yR z、-R xC(O)R x、-R xOC(O)R x、-SR x、-SOR x、-SO 2R x和-ONO 2,其中以上每个基团中的R x、R y和R z可为氢、经取代或未经取代的C 1-8烷基、经取代或未经取代的C 1-8烷氧基、经取代或未经取代的C 2-10烯基、经取代或未经取代的C 2-12炔基、经取代或未经取代的C 6-20芳基、经取代或未经取代的C 6-20芳基C 1-8烷基、经 取代或未经取代的C 3-20环烷基、经取代或未经取代的C 3-20环烷基C 1-8烷基、经取代或未经取代的C 3-8环烯基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基C 1-8烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环基C 1-8烷基环或经取代或未经取代的氨基,或R x、R y和R z的任两个可连接形成经取代或未经取代的饱和或不饱和3-10元环,所述环可任选地包括可能相同或不同且选自O、NR x或S的杂原子,其中R x为氢或经取代或未经取代的烷基。
  63. 根据权利要求1-62中任一项所述的药物组合,其中所述PI3K抑制剂包括式(IA-V)所示的化合物:
    Figure PCTCN2022075642-appb-100002
    或其对映异构体、对映异构体的混合物、两种或更多种非对映异构体的混合物、或同位素变体;或其药学上可接受的盐、溶剂化物、水合物或前药,其中
    每次出现的R独立地选自氢、卤素、-OR a、CN、经取代或未经取代的C 1-6烷基、经取代或未经取代的C 2-6烯基、经取代或未经取代的C 2-6炔基、经取代或未经取代的C 3-8环烷基和经取代或未经取代的杂环基团;
    R 1和R 2可相同或不同并且独立地选自氢、卤素和经取代或未经取代的C 1-6烷基,或与共有原子直接结合的R 1和R 2可连接形成=O基或经取代或未经取代的饱和或不饱和3-10元环,包括与R 1和R 2结合的碳原子,所述环可任选地包括一个或多个相同或不同且选自O、NR a和S的杂原子;
    每次出现的X独立地选自CR 3或N;并且
    每次出现的R 3独立地选自氢、羟基、卤素、羧基、氰基、硝基、经取代或未经取代的C 1-8烷基、经取代或未经取代的C 1-8烷氧基、经取代或未经取代的C 2-10烯基、经取代或未经取代的C 2-12炔基、经取代或未经取代的C 6-20芳基、经取代或未经取代的C 6-20芳基C 1-8烷基、经取代或未经取代的C 3-20环烷基、经取代或未经取代的C 3-20环烷基C 1-8烷 基、经取代或未经取代的C 3-8环烯基C 1-8烷基、经取代或未经取代的C 3-8环烯基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基C 1-8烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环基C 1-8烷基环、经取代或未经取代的胍、-C00R x、-C(O)R x、-C(S)R x、-C(O)NR xR y、-C(O)ONR xR y,-NR yR z、-NR xCONR yR z,-N(R x)SOR y、-N(Rx)SO 2R y、-(=N-N(R x)R y)、-NR xC(O)OR y、-NR xR y、-NR xC(O)R y-、-NR xC(S)R y、-NR xC(S)NR yR z、-SONR xR y-、-SO 2NR xR y-、-OR x、-OR xC(O)NR yR z、-OR xC(O)OR y-、-OC(O)R x、-OC(O)NR xR y、-R xNR yC(O)R z、-R xOR y、-R xC(O)OR y、-R xC(O)NR yR z、-R xC(O)R x、-R xOC(O)R x、-SR x、-SOR x、-SO 2R x和-ONO 2,其中以上每个基团中的R x、R y和R z可为氢、经取代或未经取代的C 1-8烷基、经取代或未经取代的C 1-8烷氧基、经取代或未经取代的C 2-10烯基、经取代或未经取代的C 2-12炔基、经取代或未经取代的C 6-20芳基、经取代或未经取代的C 6-20芳基C 1-8烷基、经取代或未经取代的C 3-20环烷基、经取代或未经取代的C 3-20环烷基C 1-8烷基、经取代或未经取代的C 3-8环烯基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基C 1-8烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环基C 1-8烷基环或经取代或未经取代的氨基,或R x、R y和R z的任两个可连接形成经取代或未经取代的饱和或不饱和3-10元环,所述环可任选地包括可能相同或不同且选自O、NR x或S的杂原子,其中R x为氢或经取代或未经取代的烷基;
    每次出现的R 5为氢、C 1-6烷基或卤素;
    n为0、1、2、3或4;并且
    P为0、1、2、3、4或5。
  64. 根据权利要求1-63中任一项所述的药物组合,其中所述PI3K抑制剂包括式(IA-VI)所示的化合物:
    Figure PCTCN2022075642-appb-100003
    或其对映异构体、对映异构体的混合物、两种或更多种非对映异构体的混合物、或同位素变体;或其药学上可接受的盐、溶剂化物、水合物或前药,其中
    每次出现的R独立地选自氢、卤素、-OR a、CN、经取代或未经取代的C 1-6烷基、经取代或未经取代的C 2-6烯基、经取代或未经取代的C 2-6炔基、经取代或未经取代的C 3-8环烷基和经取代或未经取代的杂环基团;
    R 1和R 2可相同或不同并且独立地选自氢、卤素和经取代或未经取代的C 1-6烷基,或与共有原子直接结合的R 1和R 2可连接形成=O基或经取代或未经取代的饱和或不饱和3-10元环,包括与R 1和R 2结合的碳原子,所述环可任选地包括一个或多个相同或不同且选自O、NR a和S的杂原子;
    每次出现的X独立地选自CR 3或N;并且
    每次出现的R 3独立地选自氢、羟基、卤素、羧基、氰基、硝基、经取代或未经取代的C 1-8烷基、经取代或未经取代的C 1-8烷氧基、经取代或未经取代的C 2-10烯基、经取代或未经取代的C 2-12炔基、经取代或未经取代的C 6-20芳基、经取代或未经取代的C 6-20芳基C 1-8烷基、经取代或未经取代的C 3-20环烷基、经取代或未经取代的C 3-20环烷基C 1-8烷基、经取代或未经取代的C 3-8环烯基C 1-8烷基、经取代或未经取代的C 3-8环烯基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基C 1-8烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环基C 1-8烷基环、经取代或未经取代的胍、-C00R x、-C(O)R x、-C(S)R x、-C(O)NR xR y、-C(O)ONR xR y,-NR yR z、-NR xCONR yR z,-N(R x)SOR y、-N(Rx)SO 2R y、-(=N-N(R x)R y)、-NR xC(O)OR y、-NR xR y、-NR xC(O)R y-、-NR xC(S)R y、-NR xC(S)NR yR z、-SONR xR y-、-SO 2NR xR y-、-OR x、-OR xC(O)NR yR z、-OR xC(O)OR y-、-OC(O)R x、-OC(O)NR xR y、-R xNR yC(O)R z、-R xOR y、-R xC(O)OR y、-R xC(O)NR yR z、-R xC(O)R x、-R xOC(O)R x、-SR x、-SOR x、-SO 2R x和- ONO 2,其中以上每个基团中的R x、R y和R z可为氢、经取代或未经取代的C 1-8烷基、经取代或未经取代的C 1-8烷氧基、经取代或未经取代的C 2-10烯基、经取代或未经取代的C 2-12炔基、经取代或未经取代的C 6-20芳基、经取代或未经取代的C 6-20芳基C 1-8烷基、经取代或未经取代的C 3-20环烷基、经取代或未经取代的C 3-20环烷基C 1-8烷基、经取代或未经取代的C 3-8环烯基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基、经取代或未经取代的具有一个或多个选自N、O和S的杂原子的5至14元杂芳基C 1-8烷基、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环、经取代或未经取代的具有至少一个选自N、O和S的杂原子的3至15元杂环基C 1-8烷基环或经取代或未经取代的氨基,或R x、R y和R z的任两个可连接形成经取代或未经取代的饱和或不饱和3-10元环,所述环可任选地包括可能相同或不同且选自O、NR x或S的杂原子,其中R x为氢或经取代或未经取代的烷基;
    每次出现的R 5为氢、C 1-6烷基或卤素;
    n为0、1、2、3或4;并且
    P为0、1、2、3、4或5。
  65. 根据权利要求1-64中任一项所述的药物组合,其中R为氢、卤素、经取代或未经取代的C 1-6烷基或OR a
  66. 根据权利要求65所述的药物组合,其中所述R a为烷基。
  67. 根据权利要求62-66中任一项所述的药物组合,其中所述Cy 1选自:
    Figure PCTCN2022075642-appb-100004
  68. 根据权利要求62-67中任一项所述的药物组合,其中所述R 1和R 2各自独立地表示氢或经取代或未经取代的C 1-6烷基。
  69. 根据权利要求62-68中任一项所述的药物组合,其中所述R 3为碘代、氰基、经取代或未经取代的烷基、经取代或未经取代的炔基、经取代或未经取代的芳基、经取代或未经取代的杂芳基。
  70. 根据权利要求62-69中任一项所述的药物组合,其中X为CR 3并且每次出现的R 3独立为氢、卤素、羟基或NH 2
  71. 根据权利要求62-70中任一项所述的化合物,其中所述R 3为氢。
  72. 根据权利要求1-71中任一项所述的药物组合,其中所述PI3K抑制剂包括:
    2-[(6-氨基-9H-嘌呤-9-基)甲基]-6-溴-3-苯基-4H-色烯-4-酮;
    2-[(6-氨基-9H-嘌呤-9-基)甲基]-3-苯基-4H-色烯-4-酮;
    2-((9H-嘌呤-6-基硫代)甲基)-3-苯基-4H-色烯-4-酮;
    2-[(9H-嘌呤-6-基硫代)甲基]-6-溴-3-苯基-4H-色烯-4-酮;
    2-((4-氨基-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-溴-3-苯基-4!1-色烯-4-酮;
    2-[(6-氨基-9H-嘌呤-9-基)甲基]-6-溴-3-(4-氟苯基)-4H-色烯-4-酮;
    2-[(6-氨基-9H-嘌呤-9-基)甲基]-3-(4-氟苯基)-4H-色烯-4-酮;
    2-[(6-氨基-9H-嘌呤-9-基)甲基]-6-溴-3-邻甲苯基-4H-色烯-4-酮;
    2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-6-溴-3-苯基-4H-色烯-4-酮;
    2-(1-(9H-嘌呤-6-基硫代)乙基)-6-溴-3-苯基-4H-色烯-4-酮;
    2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-3-苯基-4H-色烯-4-酮;
    (S)-2-(1-(9H-嘌呤-6-基氨基)乙基)-6-溴-3-苯基-4H-色烯-4-酮;
    2-((9H-嘌呤-6-基氨基)甲基)-6-溴-3-苯基-4H-色烯-4-酮;
    2-(1-(4-氨基-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-溴-3-苯基-4H-色烯-4-酮;
    2-((6-氨基-9H-嘌呤-9-基)甲基)-6-甲氧基-3-苯基-4H-色烯-4-酮;
    2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-6-溴-3-(2-氟苯基)-4H-色烯-4-酮;
    2-((6-氨基-9H-嘌呤-9-基)甲基)-6-溴-3-(2-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
    2-(1-(6-氨基-9H-嘌呤-9-基)丙基)-3-苯基-4H-色烯-4-酮;
    2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((6-氨基-9H-嘌呤-9-基)甲基)-3-(2-氟苯基)-4H-色烯-4-酮;
    2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-3-(2-氟苯基)-4H-色烯-4-酮;
    2-(1-(6-氨基-9H-嘌呤-9-基)丙基)-3-(2-氟苯基)-4H-色烯-4-酮;
    2-(1-(6-氨基-9H-嘌呤-9-基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(6-氨基-9H-嘌呤-9-基)丙基)-3-(4-氟苯基)-4H-色烯-4-酮;
    2-(1-(6-氨基-9H-嘌呤-9-基)丙基)-6-氟-3-苯基-4H-色烯-4-酮;
    2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-3-(4-氟苯基)-4H-色烯-4-酮;
    2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-6-氟-3-苯基-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
    2-((9H-嘌呤-6-基氨基)甲基)-3-苯基-4H-色烯-4-酮;
    2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-3-邻甲苯基-4H-色烯-4-酮;
    2-((9H-嘌呤-6-基氨基)甲基)-3-(2-氟苯基)-4H-色烯-4-酮;
    2-((9H-嘌呤-6-基氨基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
    (S)-2-(1-(9H-嘌呤-6-基氨基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-6-氟-3-(2-氟苯基)-4H-色烯-4-酮;
    2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-3-(3,5-二氟苯基)-4H-色烯-4-酮;
    2-(1-(6-氨基-9H-嘌呤-9-基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((4-氨基-3-(3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
    2-((4-氨基-3-(3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
    2-((4-氨基-3-(3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((4-氨基-3-(3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
    (R)-2-(1-(9H-嘌呤-6-基氨基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    (S)-2-(1-(9H-嘌呤-6-基氨基)乙基)-6-氟-3-苯基-4H-色烯-4-酮;
    2-((4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
    2-(1-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
    2-(1-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-氟-3-苯基-4H-色烯-4-酮;
    2-(1-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-氟-3-苯基-4H-色烯-4-酮;
    2-(1-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-碘-1H-吡唑并[3,4-d]嘧啶-1-基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((4-氨基-3-(吡啶-3-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
    2-((4-氨基-3-(3-羟基丙-1-炔基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
    2-((4-氨基-3-(1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
    2-((4-氨基-3-(3-(羟基甲基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
    2-((4-氨基-3-(1H-吲唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
    2-((4-氨基-3-(3-氟苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
    2-((4-氨基-3-(3-羟基丙基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
    N-(3-(4-氨基-1-((4-氧代-3-苯基-4H-色烯-2-基)甲基)-1H-吡唑并[3,4-d]嘧啶-3-基)苯基)乙酰胺;
    2-((4-氨基-3-(3-氟-5-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
    2-((4-氨基-3-(3-氟-5-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
    2-((4-氨基-3-(3-氟-5-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((4-氨基-3-(3-氟-5-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-羟基-3-甲基丁-1-炔基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
    2-(1_(4-氨基-3-(1H-吡唑-4-基)-1H-吡唑并[3,4-(1]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    (S)-2-(1-(9H-嘌呤-6-基氨基)乙基)-3-苯基-4H-色烯-4-酮;
    (S)-2-(1-(9H-嘌呤-6-基氨基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((4-氨基-3-(1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-氟-5-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-氟-5-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(1H-吲唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3,5-二甲基-1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(2-(羟基甲基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(4-氟-3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(4-氟-3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-羟基丙-1-炔基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-氟-4-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-氟-4-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-氯-5-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-氯-5-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-(三氟甲氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(4-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(4-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((6-氨基-9H-嘌呤-9-基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(4-氟-2-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(4-氟-2-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((4-氨基-3-(3-氨基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
    2-((4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-苯基-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(2-氨基嘧啶-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(1H-吲哚-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(4-氯-3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(4-氯-3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(2-氯-5-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(2-氯-5-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3,4-二甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3,4-二羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(1H-吲哚-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-甲基-1H-吲哚-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    叔丁基-(5-(4-氨基-1-(1-(3-(3-氟苯基)-4-氧代-4H-色烯-2-基)乙基)-1H-吡唑并[3,4-d]嘧啶-3-基)噻吩-2-基)氨基甲酸甲酯;
    2-(1-(4-氨基-3-(5-(氨基甲基)噻吩-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-氟-3-苯基-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-苯基-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-氟-3-苯基-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-甲基-1H-吲唑-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    N-(4-(4-氨基-1-(1-(3-(3-氟苯基)-4-氧代-4H-色烯-2-基)乙基)-1H-吡唑并[3,4-d]嘧啶-3-基)苯基)乙酰胺;
    2-(1-(4-氨基-3-(4-氨基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(2,3-二氢苯并呋喃-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-乙基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-甲基-1H-吲哚-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(2-甲氧基嘧啶-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    4-(4-氨基-1-(1-(3-(3-氟苯基)-4-氧代-4H-色烯-2-基)乙基)-1H-吡唑并[3,4-d]嘧啶-3-基)噻吩-2-甲醛;
    2-(1-(4-氨基-3-(5-(羟基甲基)噻吩-3-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(2-甲基-1H-苯并[d]咪唑-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-甲基-1H-吲哚-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((6-氨基-9H-嘌呤-9-基)甲基)-6-氟-3-苯基-4H-色烯-4-酮;2-((6-氨基-9H-嘌呤-9-基)甲基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((4-氨基-3-(3-氟-5-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((4-氨基-3-(3-氟-5-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((9H-嘌呤-6-基氨基)甲基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((9H-嘌呤-6-基氨基)甲基)-6-氟-3-苯基-4H-色烯-4-酮;(R)-2-(1-(9H-嘌呤-6-基氨基)乙基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((4-氨基-3-(1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-氟-3-苯基-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3,5-二氟-4-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3,5-二氟-4-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((4-氨基-3-(3,5-二氟-4-甲氧基苯基)-1H-吡唑并[3,4-(1]嘧啶-1-基)甲基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
    2-((4-氨基-3-(3,5-二氟-4-羟基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)甲基)-6-氟-3-(3-氟苯基)-4H-色烯-4-酮;
    (+)-2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    (-)-2-(1-(4-氨基-3-(3-甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3,5-二甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(4-甲氧基-3,5-二甲基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯 基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(2-氟-5-异丙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(2,3-二氢苯并[b][1,4]二噁英-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(1-苄基-1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(2-甲基吡啶-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3,4-二氢-2H-苯并[b][1,4]二氧杂卓-7-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(6-吗啉基吡啶-3-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(二苯并[b,d]呋喃-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(4-苯氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(4-(苄氧基)-3-氯苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-氯-4-异丙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-(二甲基氨基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(4-乙氧基-3-氟苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(4-异丙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(4-(三氟甲氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(3-(4-乙酰基苯基)-4-氨基-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4- 酮;
    2-(1-(4-氨基-3-(4-(苄氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(4-(二甲基氨基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(4-(甲磺酰基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-乙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(苯并[b]噻吩-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(5-氯噻吩-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3,5-二甲基异噁唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-丙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2~(1-(4-氨基-3-(呋喃-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(4-乙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-氯-4-甲氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-氟-4-异丙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(6-氟吡啶-3-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(嘧啶-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-(甲氧基甲基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(6-羟基萘-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-异丙氧基苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(1-甲基-1H-吡唑-4-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(1,3-二甲基-1H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(2,3-二甲基-2H-吲唑-6-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(6-甲氧基萘-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(苯并[b]噻吩-3-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(2,4-二甲氧基嘧啶-5-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(6-乙氧基萘-2-基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    3-(4-氨基-1-(1-(3-(3-氟苯基)-4-氧代-4H-色烯-2-基)乙基)-1H-吡唑并[3,4-d]嘧啶-3-基)-N-环丙基苯甲酰胺;
    2-(1-(4-氨基-3-(3-(吗啉-4-羰基)苯基)-1H-吡唑并[3,4-(1]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    2-(1-(4-氨基-3-(3-(二氟甲氧基)苯基)-1H-吡唑并[3,4-d]嘧啶-1-基)乙基)-3-(3-氟苯基)-4H-色烯-4-酮;
    5-(4-氨基-1-(1-(3-(3-氟苯基)-4-氧代-4H-色烯-2-基)乙基)-1H-吡唑并[3,4-d]嘧啶-3-基)呋喃-2-甲醛;
    和/或它们的对映异构体、对映异构体的混合物、两种或更多种非对映异构体的混合物、同位素变体、药学上可接受的盐、溶剂化物、水合物或前药。
  73. 根据权利要求1-72中任一项所述的药物组合,其中所述PI3K抑制剂包括:2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮,或其对映异构体、对映异构体的混合物、 两种或更多种非对映异构体的混合物、或同位素变体;或其药学上可接受的盐、溶剂化物、水合物或前药。
  74. 根据权利要求1-73中任一项所述的药物组合,其中所述PI3K抑制剂包括(S)-2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮或其药学上可接受的盐、溶剂化物、水合物或前药。
  75. 根据权利要求1-74中任一项所述的药物组合,其包括PD-L1/TGFβ双重抑制剂和PI3K抑制剂。
  76. 根据权利要求75中任一项所述的药物组合,其中所述PD-L1/TGFβ双重抑制剂包含抗PD-L1抗体或其抗原结合片段和TGFβRII或其功能活性片段。
  77. 根据权利要求76中任一项所述的药物组合,其中所述PD-L1/TGFβ双重抑制剂包含2条多肽,所述多肽包含选自下组的氨基酸序列:(a)SEQ ID NO:7所示的氨基酸序列;(b)具有与SEQ ID NO:7所示的氨基酸序列至少85%,90%,95%或者99%序列同一性的氨基酸序列;(c)通过加成、消除或取代反应所得到的与SEQ ID NO:7所示的氨基酸序列具有1个或更多差异的氨基酸序列。
  78. 根据权利要求75所述的药物组合,其中所述PI3K抑制剂为PI3Kδ/γ双重抑制剂。
  79. 根据权利要求78所述的药物组合,其中所述PI3K抑制剂为(S)-2-(1-(9H-嘌呤-6-基氨基)丙基)-3-(3-氟苯基)-4H-色烯-4-酮或其药学上可接受的盐、溶剂化物、水合物或前药。
  80. 根据权利要求1-79中任一项所述的药物组合,所述第二治疗剂与所述PI3K抑制剂在所述药物组合中不互相混合。
  81. 根据权利要求1-80中任一项所述的药物组合,所述第二治疗剂与所述PI3K抑制剂各自独立地存在于单独的容器中。
  82. 根据权利要求1-81中任一项所述的药物组合,所述的药物组合还可以进一步地包括第三活性物质。
  83. 根据权利要求82所述的药物组合,所述的第三活性物质可以选自长春新碱、长春碱、长春地辛、依托泊苷、多西他赛、紫杉醇、伊立替康、长春瑞滨、米托蒽醌、长春氟宁、拓扑替康中的任意一种或其任意地组合。
  84. 药物组合物,所述药物组合包括权利要求1-83中任一项所述的PI3K抑制剂和第二治疗剂,以及任选的一种或多种药学上可接受的载体或赋形剂。
  85. 根据权利要求84所述的药物组合物,其中所述第二治疗剂为权利要求46-60中任一项所述的PD-L1/TGFβ双重抑制剂。
  86. 根据权利要求84-85中任一项所述的药物组合物,其中所述PI3K抑制剂为权利要求61-74中任一项所述的PI3Kδ/γ双重抑制剂。
  87. 根据权利要求84-86中任一项所述的药物组合物,其中所述第二治疗剂与所述PI3K抑制剂存在于单一或分开的剂型中。
  88. 一种治疗和/或预防肿瘤的方法,其包括向有需要的受试者施用:有效量的权利要求1-83中任一项所述的药物组合,或权利要求84-87中任一项所述的药物组合物。
  89. 根据权利要求88所述的方法,其包括向有需要的受试者施用:有效量的权利要求61-74中任一项所述的PI3K抑制剂;以及权利要求46-60中任一项所述的PD-L1/TGFβ双重抑制剂。
  90. 根据权利要求89所述的方法,其中所述PI3K抑制剂与所述PD-L1/TGFβ双重抑制剂同时施用。
  91. 根据权利要求90所述的方法,其中所述PI3K抑制剂在所述PD-L1/TGFβ双重抑制剂之后施用。
  92. 根据权利要求89所述的方法,其中所述PI3K抑制剂在所述PD-L1/TGFβ双重抑制剂之前施用。
  93. 根据权利要求89所述的方法,还包括向有需要的受试者施用有效量的权利要求41所述的小分子TGFβ抑制剂。
  94. 根据权利要求88-93中任一项所述的方法,其中所述肿瘤包括实体瘤和非实体瘤。
  95. 根据权利要求88-94中任一项所述的方法,其中所述肿瘤包括PI3K表达异常的肿瘤。
  96. 根据权利要求88-95中任一项所述的方法,其中所述肿瘤包括PD-L1表达异常的肿瘤。
  97. 根据权利要求88-96中任一项所述的方法,其中所述肿瘤包括TGFβ表达异常的肿瘤。
  98. 根据权利要求88-97中任一项所述的方法,所述肿瘤包括消化道肿瘤、黑素瘤或淋巴瘤。
  99. 权利要求1-83中任一项所述的药物组合或权利要求84-87中任一项所述的药物组合物在制备用于治疗和/或预防肿瘤的药物中的用途。
  100. 权利要求1-83中任一项所述的药物组合或权利要求84-87中任一项所述的药物组合物用于治疗和/或预防肿瘤。
  101. 一种抑制肿瘤生长的方法,所述方法包括使肿瘤接触权利要求1-83中任一项所述的药物组合或权利要求84-87中任一项所述的药物组合物。
  102. 一种抑制肿瘤生长的方法,所述方法包括使肿瘤接触权利要求61-74中任一项所述 的PI3K抑制剂与权利要求46-60中任一项所述的PD-L1/TGFβ双重抑制剂。
  103. 一种药盒,其包括:(1)第一容器,以及位于所述第一容器中的权利要求61-74中任一项所述的PI3K抑制剂;(2)第二容器,以及位于所述第二容器中权利要求46-60中任一项所述的PD-L1/TGFβ双重抑制剂。
  104. 一种药盒,其可以包括:(1)第一容器,以及位于所述第一容器中的权利要求61-74中任一项所述的PI3K抑制剂;(2)第二容器,以及位于所述第二容器中的权利要求17-29中任一项所述的免疫检查点的抑制剂或权利要求30-44中任一项所述的TGFβ抑制剂。
  105. 根据权利要求104所述的药盒,所述药盒还包括(3)第三容器,以及位于第三容器中的权利要求17-29中任一项所述的免疫检查点的抑制剂或权利要求30-44中任一项所述的TGFβ抑制剂,所述第三容器中的药剂与第二容器中的药剂不同。
PCT/CN2022/075642 2021-02-09 2022-02-09 治疗肿瘤的方法和组合 WO2022171121A1 (zh)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP22752281.0A EP4292596A1 (en) 2021-02-10 2022-02-09 Method and combination for treating tumors
US18/264,394 US20240102634A1 (en) 2021-02-09 2022-02-09 A light valve surface image and light beam projector
CN202280012531.5A CN117222413A (zh) 2021-02-10 2022-02-09 治疗肿瘤的方法和组合

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN202110184081.5 2021-02-10
CN202110184081 2021-02-10
CN202110410557.2 2021-04-13
CN202110410557 2021-04-13
CN202210099148 2022-01-27
CN202210099148.X 2022-01-27

Publications (1)

Publication Number Publication Date
WO2022171121A1 true WO2022171121A1 (zh) 2022-08-18

Family

ID=82838279

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/075642 WO2022171121A1 (zh) 2021-02-09 2022-02-09 治疗肿瘤的方法和组合

Country Status (4)

Country Link
EP (1) EP4292596A1 (zh)
CN (3) CN114452403A (zh)
TW (1) TW202241494A (zh)
WO (1) WO2022171121A1 (zh)

Citations (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4751180A (en) 1985-03-28 1988-06-14 Chiron Corporation Expression using fused genes providing for protein product
WO1988009344A1 (en) 1987-05-21 1988-12-01 Creative Biomolecules, Inc. Targeted multifunctional proteins
US4935233A (en) 1985-12-02 1990-06-19 G. D. Searle And Company Covalently linked polypeptide cell modulators
WO1999054440A1 (en) 1998-04-21 1999-10-28 Micromet Gesellschaft Für Biomedizinische Forschung Mbh CD19xCD3 SPECIFIC POLYPEPTIDES AND USES THEREOF
WO2003042402A2 (en) 2001-11-13 2003-05-22 Dana-Farber Cancer Institute, Inc. Agents that modulate immune cell activation and methods of use thereof
WO2006121168A1 (en) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
WO2007005874A2 (en) 2005-07-01 2007-01-11 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)
WO2008156712A1 (en) 2007-06-18 2008-12-24 N. V. Organon Antibodies to human programmed death receptor pd-1
US7488802B2 (en) 2002-12-23 2009-02-10 Wyeth Antibodies against PD-1
WO2009101611A1 (en) 2008-02-11 2009-08-20 Curetech Ltd. Monoclonal antibodies for tumor treatment
WO2009114335A2 (en) 2008-03-12 2009-09-17 Merck & Co., Inc. Pd-1 binding proteins
WO2010036959A2 (en) 2008-09-26 2010-04-01 Dana-Farber Cancer Institute Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor
WO2010077634A1 (en) 2008-12-09 2010-07-08 Genentech, Inc. Anti-pd-l1 antibodies and their use to enhance t-cell function
WO2010089411A2 (en) 2009-02-09 2010-08-12 Universite De La Mediterranee Pd-1 antibodies and pd-l1 antibodies and uses thereof
WO2011066342A2 (en) 2009-11-24 2011-06-03 Amplimmune, Inc. Simultaneous inhibition of pd-l1/pd-l2
WO2011082400A2 (en) 2010-01-04 2011-07-07 President And Fellows Of Harvard College Modulators of immunoinhibitory receptor pd-1, and methods of use thereof
WO2011159877A2 (en) 2010-06-18 2011-12-22 The Brigham And Women's Hospital, Inc. Bi-specific antibodies against tim-3 and pd-1 for immunotherapy in chronic immune conditions
WO2011161699A2 (en) 2010-06-25 2011-12-29 Aurigene Discovery Technologies Limited Immunosuppression modulating compounds
US20120114649A1 (en) 2008-08-25 2012-05-10 Amplimmune, Inc. Delaware Compositions of pd-1 antagonists and methods of use
US20130034559A1 (en) 2009-11-24 2013-02-07 Medlmmune Limited Targeted Binding Agents Against B7-H1
WO2013079174A1 (en) 2011-11-28 2013-06-06 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof
US20150272936A1 (en) * 2012-11-08 2015-10-01 Rhizen Pharmaceuticals Sa Pharmaceutical compositions containing a pde4 inhibitor and a pi3 delta or dual pi3 delta-gamma kinase inhibitor
US20150361070A1 (en) * 2014-06-13 2015-12-17 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
WO2017007658A1 (en) * 2015-07-07 2017-01-12 Rigel Pharmaceuticals, Inc. A combination for immune mediated cancer treatment
US20180222982A1 (en) * 2015-07-29 2018-08-09 Novartis Ag Combination therapies comprising antibody molecules to pd-1
WO2020039097A1 (en) * 2018-08-24 2020-02-27 Biomedical Research Foundation Of The Academy Of Athens (Brfaa) 2,6-bis(((1h-benzo[d]imidazol-2-yl)thio)methyl)pyridine and n2,n6-dibenzylpyridine-2,6-dicarboxamide derivatives and related compounds as phosphoinositide 3-kinase (pi3k) inhibitors for treating cancer
WO2020072445A1 (en) * 2018-10-01 2020-04-09 Verastem, Inc. Combination therapies
US20200289520A1 (en) * 2017-12-06 2020-09-17 Rhizen Pharmaceuticals Sa Composition and method for treating peripheral t-cell lymphoma and cutaneous t-cell lymphoma
CN111712243A (zh) * 2018-01-10 2020-09-25 Array生物制药公司 治疗癌症的方法和组合疗法
KR102195221B1 (ko) * 2019-12-31 2020-12-24 서울대학교산학협력단 포스파티딜이노시톨 3-키나아제 억제제 및 프로그램화 세포 사멸 단백질 1 억제제를 포함하는, 삼중음성 유방암의 방사선 병용 치료용 약학적 조성물

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IT201600111877A1 (it) * 2016-11-07 2018-05-07 Biouniversa Srl Anti-BAG3 antibodies in combination with inhibitors of immune check-point for therapeutic use
WO2018127699A1 (en) * 2017-01-06 2018-07-12 Bicyclerd Limited Compounds for treating cancer
US11802153B2 (en) * 2017-10-18 2023-10-31 Forty Seven, Inc. Anti-CD47 agent-based ovarian cancer therapy

Patent Citations (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4751180A (en) 1985-03-28 1988-06-14 Chiron Corporation Expression using fused genes providing for protein product
US4935233A (en) 1985-12-02 1990-06-19 G. D. Searle And Company Covalently linked polypeptide cell modulators
WO1988009344A1 (en) 1987-05-21 1988-12-01 Creative Biomolecules, Inc. Targeted multifunctional proteins
WO1999054440A1 (en) 1998-04-21 1999-10-28 Micromet Gesellschaft Für Biomedizinische Forschung Mbh CD19xCD3 SPECIFIC POLYPEPTIDES AND USES THEREOF
WO2003042402A2 (en) 2001-11-13 2003-05-22 Dana-Farber Cancer Institute, Inc. Agents that modulate immune cell activation and methods of use thereof
US20100028330A1 (en) 2002-12-23 2010-02-04 Medimmune Limited Methods of upmodulating adaptive immune response using anti-pd1 antibodies
US7488802B2 (en) 2002-12-23 2009-02-10 Wyeth Antibodies against PD-1
WO2006121168A1 (en) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
US8008449B2 (en) 2005-05-09 2011-08-30 Medarex, Inc. Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
WO2007005874A2 (en) 2005-07-01 2007-01-11 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)
US8354509B2 (en) 2007-06-18 2013-01-15 Msd Oss B.V. Antibodies to human programmed death receptor PD-1
WO2008156712A1 (en) 2007-06-18 2008-12-24 N. V. Organon Antibodies to human programmed death receptor pd-1
WO2009101611A1 (en) 2008-02-11 2009-08-20 Curetech Ltd. Monoclonal antibodies for tumor treatment
WO2009114335A2 (en) 2008-03-12 2009-09-17 Merck & Co., Inc. Pd-1 binding proteins
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
US8609089B2 (en) 2008-08-25 2013-12-17 Amplimmune, Inc. Compositions of PD-1 antagonists and methods of use
US20120114649A1 (en) 2008-08-25 2012-05-10 Amplimmune, Inc. Delaware Compositions of pd-1 antagonists and methods of use
WO2010036959A2 (en) 2008-09-26 2010-04-01 Dana-Farber Cancer Institute Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor
WO2010077634A1 (en) 2008-12-09 2010-07-08 Genentech, Inc. Anti-pd-l1 antibodies and their use to enhance t-cell function
US8217149B2 (en) 2008-12-09 2012-07-10 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
WO2010089411A2 (en) 2009-02-09 2010-08-12 Universite De La Mediterranee Pd-1 antibodies and pd-l1 antibodies and uses thereof
US20120039906A1 (en) 2009-02-09 2012-02-16 INSER (Institut National de la Recherche Medicale) PD-1 Antibodies and PD-L1 Antibodies and Uses Thereof
WO2011066342A2 (en) 2009-11-24 2011-06-03 Amplimmune, Inc. Simultaneous inhibition of pd-l1/pd-l2
US20130034559A1 (en) 2009-11-24 2013-02-07 Medlmmune Limited Targeted Binding Agents Against B7-H1
US8779108B2 (en) 2009-11-24 2014-07-15 Medimmune, Limited Targeted binding agents against B7-H1
WO2011082400A2 (en) 2010-01-04 2011-07-07 President And Fellows Of Harvard College Modulators of immunoinhibitory receptor pd-1, and methods of use thereof
WO2011159877A2 (en) 2010-06-18 2011-12-22 The Brigham And Women's Hospital, Inc. Bi-specific antibodies against tim-3 and pd-1 for immunotherapy in chronic immune conditions
WO2011161699A2 (en) 2010-06-25 2011-12-29 Aurigene Discovery Technologies Limited Immunosuppression modulating compounds
WO2013079174A1 (en) 2011-11-28 2013-06-06 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof
US20140341917A1 (en) 2011-11-28 2014-11-20 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof
US20150272936A1 (en) * 2012-11-08 2015-10-01 Rhizen Pharmaceuticals Sa Pharmaceutical compositions containing a pde4 inhibitor and a pi3 delta or dual pi3 delta-gamma kinase inhibitor
US20150361070A1 (en) * 2014-06-13 2015-12-17 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
WO2017007658A1 (en) * 2015-07-07 2017-01-12 Rigel Pharmaceuticals, Inc. A combination for immune mediated cancer treatment
US20180222982A1 (en) * 2015-07-29 2018-08-09 Novartis Ag Combination therapies comprising antibody molecules to pd-1
US20200289520A1 (en) * 2017-12-06 2020-09-17 Rhizen Pharmaceuticals Sa Composition and method for treating peripheral t-cell lymphoma and cutaneous t-cell lymphoma
CN111712243A (zh) * 2018-01-10 2020-09-25 Array生物制药公司 治疗癌症的方法和组合疗法
WO2020039097A1 (en) * 2018-08-24 2020-02-27 Biomedical Research Foundation Of The Academy Of Athens (Brfaa) 2,6-bis(((1h-benzo[d]imidazol-2-yl)thio)methyl)pyridine and n2,n6-dibenzylpyridine-2,6-dicarboxamide derivatives and related compounds as phosphoinositide 3-kinase (pi3k) inhibitors for treating cancer
WO2020072445A1 (en) * 2018-10-01 2020-04-09 Verastem, Inc. Combination therapies
KR102195221B1 (ko) * 2019-12-31 2020-12-24 서울대학교산학협력단 포스파티딜이노시톨 3-키나아제 억제제 및 프로그램화 세포 사멸 단백질 1 억제제를 포함하는, 삼중음성 유방암의 방사선 병용 치료용 약학적 조성물

Non-Patent Citations (36)

* Cited by examiner, † Cited by third party
Title
"Bioreversible Carriers in Drug Design", 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS
A1-LAZIKANI ET AL., JMOL BIOL, vol. 273, 1997, pages 927 - 48
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403
ALTSCHUL ET AL., NUCLEIC ACIDS RES., vol. 25, 1977, pages 3389
BENIGNI ET AL., J. AM. SOC. NEPHROL, vol. 14, 2003, pages 1816
BERGE ET AL.: "describes pharmaceutically acceptable salts in detail v& J.", PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 19
BORDER ET AL., NATURE, vol. 346, 1990, pages 371 - 374
BORDER ET AL., NATURE, vol. 360, 1992, pages 361 - 364
CHEADLE ET AL., MOL IMMUNOL, vol. 29, 1992, pages 21 - 30
CLAYTON ET AL., J EXP MED., vol. 196, no. 6, 2002, pages 753 - 19 63
DALL'ACQUA ET AL., BIOCHEM., vol. 37, 1998, pages 9266 - 9273
FOSTER, F.M. ET AL., J CELL SCI, vol. 116, 2003, pages 3037 - 3040
FUNG-LEUNG, CELL SIGNAL, vol. 23, no. 4, 2011, pages 603 - 8
HAMID, O. ET AL., NEW ENGLAND JOURNAL OF MEDICINE, vol. 369, no. 2, 2013, pages 134 - 44
HIGUCHI, T. ET AL.: "Pro-drugs as Novel Delivery Systems", A. C. S. SYMPOSIUM SERIES, vol. 14
HUSTON ET AL.: "Protein Engineering of Antibody Binding Sites: Recovery of SpecificActivity in an Anti-Digoxin Single-Chain Fv Analogue Produced in Escherichiacoli", PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 5883
ISAKA ET AL., KIDNEYINT., vol. 55, 1999, pages 465 - 475
KABAT ET AL.: "Immunology protein sequence", 1991, UNITED STATES NATIONAL INSTITUTION OF HEALTH
KATSO ET AL., ANNU. REV. CELL DEV. BIOL., vol. 17, 2001, pages 615 - 675
KNIGHT ET AL., CELL, vol. 125, no. 4, 2006, pages 733 - 47
LI ET AL., ANNU. REV. IMMUNOL, vol. 24, 2006, pages 99 - 146
MANFRED E. WOLFF: "Design of Prodrugs", 1985, ELSELVIER, pages: 7 - 9
MATHIEU DONDELINGER ET AL.: "Understanding the Significance and Imperial of Antibody Numbering and Antigen-Binding Surface/Residue Definition", FRONT.IMMUNOL., 16 October 2018 (2018-10-16)
MUYLDERMANS S., J BIOTECHNOL, vol. 74, 2001, pages 277 - 302
NGUYEN V. K. ET AL., THE EMBO JOURNAL, vol. 19, 2000, pages 921 - 930
NOBUHIRO TSUCHIYA, TOSHIAKI YOSHIKAWA, NORIHIRO FUJINAMI, KEIGO SAITO, SHOICHI MIZUNO, YU SAWADA, ITARU ENDO, TETSUYA NAKATSURA: "Immunological efficacy of glypican-3 peptide vaccine in patients with advanced hepatocellular carcinoma", ONCOIMMUNOLGY, vol. 6, no. 10, 3 October 2017 (2017-10-03), US , pages 1 - 11, XP055742966, ISSN: 2162-4011, DOI: 10.1080/2162402X.2017.1346764 *
OKKENHAUG ET AL., SCIENCE, vol. 297, no. 5583, 2002, pages 1031 - 34
RAAGWHITLOW, FASEB, vol. 9, no. 1, 1995, pages 73 - 80
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 2001, COLD SPRING HARBOR LABORATORY PRESS
SHARMA ET AL., DIABETES, vol. 45, pages 522
VANHAESEBROECK ET AL., CURRENT TOPIC MICROBIOL. IMMUNOL, vol. 347, 2010, pages 1 - 19
VANLANDSCHOOT P. ET AL., ANTIVIRAL RESEARCH, vol. 92, 2011, pages 389 - 407
WANG MANNI; LIU YU; CHENG YUAN; WEI YUQUAN; WEI XIAWEI: "Immune checkpoint blockade and its combination therapy with small-molecule inhibitors for cancer treatment", BIOCHIMICA ET BIOPHYSICA ACTA (BBA) - REVIEWS ON CANCER, vol. 1871, no. 2, 1 April 2019 (2019-04-01) - 31 December 2018 (2018-12-31), NL , pages 199 - 224, XP085692939, ISSN: 0304-419X, DOI: 10.1016/j.bbcan.2018.12.002 *
WARD ET AL.: "Binding Actida of a Repertoire ofSingle Immunoglobulin", VARIABLE DOMAINS SECRETED FROM ESCHERICHIA COLI, NATURE, vol. 341, 1989, pages 544 - 546
XIN ET AL., TRANSPLANTATION, vol. 15, 2004, pages 1433
ZHOU, J. ET AL., BLOOD, vol. 110, 2007, pages 1607 - 1611

Also Published As

Publication number Publication date
CN114452403A (zh) 2022-05-10
TW202241494A (zh) 2022-11-01
CN117222413A (zh) 2023-12-12
CN115105600A (zh) 2022-09-27
EP4292596A1 (en) 2023-12-20
CN114452403A8 (zh) 2023-01-31

Similar Documents

Publication Publication Date Title
JP6877420B2 (ja) toll様受容体アゴニストを含む抗体コンジュゲート
JP6588461B2 (ja) 抗血管新生剤及びox40結合アゴニストを含む併用療法
JP2020143149A (ja) Pd−1に対する抗体分子およびその使用
JP2021119167A (ja) Pd−1に対する抗体分子を含む組み合わせ治療
US20200277378A1 (en) Combination therapies
AU2024201912A1 (en) Antibodies specific to human poliovirus receptor (PVR)
KR20190140472A (ko) 톨-유사 수용체 작용제를 포함하는 항체 접합체 및 병용 요법
WO2016187220A2 (en) Anti-ror1 antibodies
TW201625692A (zh) 抗體藥物結合物
KR20210028544A (ko) 인간 ror1에 대한 항체를 포함하는 항체 약물 접합체 및 이의 용도
CN112566661B (zh) 喹啉衍生物与抗体的药物组合
CA3123511A1 (en) Dosing regimen and pharmaceutical combination comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
BR112021011900A2 (pt) Anticorpos para pmel17 e conjugados dos mesmos
WO2022171121A1 (zh) 治疗肿瘤的方法和组合
JP2023103352A (ja) 抗原結合タンパク質
US20240139198A1 (en) Method and combination for treating tumors
KR20220123105A (ko) 항-갈렉틴-9 항체 및 그것의 용도
WO2022075482A1 (ja) がん治療用医薬
RU2788092C2 (ru) Молекулы антител к pd-1 и их применения
KR20210063070A (ko) 항-b7-h3 항체를 포함하는 항체-약물 접합체 및 이의 용도
CN115038466A (zh) 联合治疗及其用途和方法
KR20210028556A (ko) 인간 ror1에 대한 항체를 포함하는 항체 약물 접합체 및 이의 용도

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: 22752281

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 18276394

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2022752281

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2022752281

Country of ref document: EP

Effective date: 20230911