WO2023079428A1 - Combination therapies using tlr7/8 agonist - Google Patents

Combination therapies using tlr7/8 agonist Download PDF

Info

Publication number
WO2023079428A1
WO2023079428A1 PCT/IB2022/060461 IB2022060461W WO2023079428A1 WO 2023079428 A1 WO2023079428 A1 WO 2023079428A1 IB 2022060461 W IB2022060461 W IB 2022060461W WO 2023079428 A1 WO2023079428 A1 WO 2023079428A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
cancer
amino
imidazo
combination
Prior art date
Application number
PCT/IB2022/060461
Other languages
French (fr)
Inventor
Sudhakar Reddy Chintharlapalli
Ruth Ingeborg Johanna SEELIGE
Stina Lisa URBAN
Hae Jung WON
Original Assignee
Pfizer Inc.
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 Pfizer Inc. filed Critical Pfizer Inc.
Publication of WO2023079428A1 publication Critical patent/WO2023079428A1/en

Links

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
    • 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
    • A61K31/4353Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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
    • A61K39/39541Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against normal tissues, cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C07K16/2818Immunoglobulins [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 CD28 or CD152
    • 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
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered

Definitions

  • the present disclosure relates to combination therapies useful for the treatment of cancers.
  • Certain embodiments relate to a combination therapy which comprises a PD-1 axis binding antagonist, in combination with a Toll-like receptor 7/8 (TLR7/8) agonist or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising such compounds or salts.
  • TLR7/8 Toll-like receptor 7/8
  • the disclosure also relates to associated methods of treatment, pharmaceutical combinations, and pharmaceutical uses. The methods and combinations are useful for any indication for which the therapeutic is itself useful in the detection, treatment and/or prevention of a disease, disorder, or other condition of a subject.
  • NMIBC Non-muscle invasive bladder cancer
  • balder cancer patients generally undergo transurethral resection of the bladder tumor (TURBT) and Intravesical (IVe) adjuvant chemotherapy (mitomycin) or immunotherapy with Bacillus Calmette-Guerin (BCG) to prevent recurrence.
  • TURBT transurethral resection of the bladder tumor
  • IVe Intravesical adjuvant chemotherapy
  • mitomycin mitomycin
  • BCG Bacillus Calmette-Guerin
  • kits comprising one or more of the combinations of the disclosure.
  • the disclosure provides a method for treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of a Programmed Death 1 protein (PD-1 ) axis binding antagonist, in combination with a compound of Formula I: or a pharmaceutically acceptable salt thereof, wherein
  • PD-1 Programmed Death 1 protein
  • R 1 and R 2 are independently Ci 3 alkyl; or
  • R 1 and R 2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
  • R 4 is C1-6 alkyl, or (CH2)nO(CH2)mCH3, wherein said C1 -6 alkyl or any carbon of the (CH2)nO(CH2)mCH3 group is substituted with 0 to 3 F as valency allows;
  • R 5 is 0-3 alkyl, or OC1 -3 alkyl, wherein the C1-3 alkyl is substituted by 0 to 3 F;
  • R 6 is H, or C1-3 alkyl, wherein the C1-3 alkyl is substituted with 0 to 3 F; m is 0 to 2; and n is 1 to 3.
  • the compound is of Formula I, wherein:
  • R 1 and R 2 are independently Ci 2 alkyl; or
  • R 1 and R 2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
  • R 4 is C3-5 alkyl, or (CH2)nO(CH2)mCH 3 ;
  • R 5 is C1-2 alkyl
  • R6 is H; m is 1 ; and n is 1 .
  • the compound of Formula I is selected from:
  • the compound of Formula I is preferably 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5- c]pyridin-1-yl)methyl)-2-methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof.
  • the preferred compound is also known as PF- 07225570 and has the following structure A:
  • the PD-1 axis binding antagonist comprises a PD-1 binding antagonist, or a PD-L1 binding antagonist.
  • the PD-1 binding antagonist is an anti-PD-1 antibody.
  • the anti-PD-1 antibody is sasanlimab, nivolumab, pembrolizumab, pidilizumab, cemiplimab, tislelizumab, spartalizumab, sintilimab, MEDI- 0680, BGB-108, AGEN2034, genolimzumab, CBT-502, camrelizumab, or any combination thereof.
  • the preferred anti-PD-1 antibody is sasanlimab.
  • the PD-L1 binding antagonist is an anti-PD-L1 antibody.
  • the anti-PD-L1 antibody is BMS-936559, KN035, atezolizumab, durvalumab, avelumab, or any combination thereof.
  • the cancer being treated is bladder cancer.
  • the cancer being treated is non-muscle invasive bladder cancer.
  • the non-muscle invasive bladder cancer is Bacillus Calmette-Guerin (BCG) un-responsive non-muscle invasive bladder cancer.
  • the PD-1 axis binding antagonist is administered by intravenous infusion or subcutaneous delivery, and the compound of Formula I, or a pharmaceutically acceptable salt thereof, is administered by intravesical delivery to bladder epithelium of the subject.
  • BCG immunotherapy in this disclosure refers to a weakened, live bacterium, bacillus Calmette-Guerin (BCG), which is FDA-approved immunotherapy and helps reduce the risk of bladder cancer recurrence by stimulating an immune response that targets the bacteria as well as any nearby bladder cancer cells.
  • BCG bacillus Calmette-Guerin
  • the compound 2-((4- amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)methyl)-2- methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof is administered with an interval of at least about 7 days (7 ⁇ 2 days).
  • the disclosure provides a combination therapy comprising a PD-1 axis binding antagonist and a TLR7/8 agonist, as provided in this disclosure, for use in the treatment of cancer in a subject in need thereof.
  • the disclosure provides a combination of a PD-1 axis binding antagonist and a TLR7/8 agonist, for use in the treatment of cancer in a subject.
  • Such combination can be used, for example, in the methods of the present disclosure.
  • the disclosure provides a medicament comprising a TLR7/8 agonist for use, in combination with a PD-1 axis binding antagonist, for treating a cancer.
  • the disclosure provides a medicament comprising a PD-1 axis binding antagonist for use, in combination with a TLR7/8 agonist in the treatment of a cancer.
  • the disclosure provides the use of a combination comprising a PD-1 axis binding antagonist and a TLR7/8 agonist for the treatment of cancer in a subject in need thereof.
  • the disclosure provides a kit that comprises a PD-1 axis binding antagonist, a TLR7/8 agonist, and a package insert, and the package insert comprises instructions for treating a subject for cancer using the medicaments.
  • a kit that comprises a PD-1 axis binding antagonist, a TLR7/8 agonist, and a package insert, and the package insert comprises instructions for treating a subject for cancer using the medicaments.
  • Such kit can be used, for example, in the methods of the present disclosure.
  • FIG. 2 shows tumor growth inhibition in MB49 subcutaneous tumor model with various combinatorial strategies of PF-07225570 and Anti-PD-1 compared to Single Agent Anti-PD-1 or Single Agent PF-07225570. Data and p-values for FIG.
  • alkyl refers to a saturated, monovalent aliphatic hydrocarbon radical including straight chain and branched chain groups having the specified number of carbon atoms.
  • the alkyl substituents disclosed herein may be specified as 1 to 6 carbon atoms (“Ci -6 alkyl”), or 1 to 3 carbon atoms (“Ci -3 alkyl”), and so forth.
  • alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, n-heptyl, n-octyl and the like.
  • alkoxy refers to a monovalent -O-alkyl group, wherein the alkyl portion has the specified number of carbon atoms.
  • Alkoxy groups typically contain 1 to 8 carbon atoms (“Ci -8 alkoxy”), or 1 to 6 carbon atoms (“Ci -6 alkoxy”), or 1 to 4 carbon atoms (“Ci -4 alkoxy”).
  • C1-4 alkoxy includes methoxy, ethoxy, isopropoxy, tert-butyloxy (i.e., -OCH3, -OCH2CH3, -OCH(CH3)2, -OC(CH3)3), and the like.
  • drug including, but not limited to, “drug,” “agent,” “component,” “composition,” “compound,” “substance,” “targeted agent,” “targeted therapeutic agent,” “therapeutic agent,” and “medicament” may be used interchangeably to refer to the small molecule compounds of the present disclosure, e.g., a TLR7/8 agonist, an anti- PD-L1 antibody, an anti-PD-1 antibody, or combinations thereof.
  • therapeutic antibody and “antibody” may be used interchangeable to refer to an antibody that is used in the treatment of a disease or a disorder.
  • a therapeutic antibody may have various mechanisms of action.
  • a therapeutic antibody may bind and neutralize the normal function of a target associated with an antigen.
  • a monoclonal antibody that blocks the activity of the protein needed for the survival of a cancer cell causes the cell's death.
  • Another therapeutic antibody may bind and activate the normal function of a target associated with an antigen.
  • a monoclonal antibody can bind to a protein on a cell and trigger an apoptosis signal.
  • Yet another monoclonal antibody may bind to a target antigen expressed only on diseased tissue; conjugation of a toxic payload (effective agent), such as a chemotherapeutic or radioactive agent, to the monoclonal antibody can create an agent for specific delivery of the toxic payload to the diseased tissue, reducing harm to healthy tissue.
  • a toxic payload such as a chemotherapeutic or radioactive agent
  • a “biologically functional fragment” of a therapeutic antibody will exhibit at least one if not some or all of the biological functions attributed to the intact antibody, the function comprising at least specific binding to the target antigen.
  • the therapeutic antibody may bind to any protein, including, without limitation, a PD-L1 , a PD-1. Accordingly, therapeutic antibodies include, without limitation, anti-PD- L1 antibodies, anti-PD-1 antibodies, or combinations thereof.
  • the terms “synergy” or “synergistic” are used to mean that the result of the combination of two or more compounds, components or targeted agents is greater than the sum of each agent together.
  • the terms “synergy” or “synergistic” also means that there is an improvement in the disease condition or disorder being treated, over the use of the two or more compounds, components or targeted agents while each compound, component or targeted agent individually. This improvement in the disease condition or disorder being treated is a “synergistic effect”.
  • a “synergistic amount” is an amount of the combination of the two compounds, components or targeted agents that results in a synergistic effect, as “synergistic” is defined herein.
  • the optimum range for the effect and absolute dose ranges of each component for the effect may be definitively measured by administration of the components over different w/w (weight per weight) ratio ranges and doses to patients in need of treatment.
  • w/w weight per weight
  • the observation of synergy in in vitro models or in vivo models can be predictive of the effect in humans and other species and in vitro models or in v/vomodels exist, as described herein, to measure a synergistic effect and the results of such studies can also be used to predict effective dose and plasma concentration ratio ranges and the absolute doses and plasma concentrations required in humans and other species by the application of pharmacokinetic/pharmacodynamic methods.
  • a "chemotherapeutic agent” is a chemical compound useful in the treatment of cancer.
  • immunotherapy refers to the treatment of a subject by a method comprising inducing, enhancing, suppressing, or otherwise modifying an immune response.
  • abnormal cell growth refers to cell growth that is independent of normal regulatory mechanisms (e.g., loss of contact inhibition). Abnormal cell growth may be benign (not cancerous), or malignant (cancerous).
  • a “disorder” is any condition that would benefit from treatment with the compounds of the present invention. This includes chronic and acute disorders or diseases including those pathological conditions which predispose the subject to the disorder in question.
  • antibody refers to an immunoglobulin molecule capable of specific binding to a target, such as a carbohydrate, polynucleotide, lipid, polypeptide, etc., through at least one antigen recognition site, located in the variable region of the immunoglobulin molecule.
  • the term encompasses a polyclonal antibody, a monoclonal antibody, a chimeric antibody, a bispecific antibody, a dual-specific antibody, bifunctional antibody, a trispecific antibody, a multispecific antibody, a bispecific heterodimeric diabody, a bispecific heterodimeric IgG, a labeled antibody, a humanized antibody, a human antibody, and fragments thereof (such as Fab, Fab’, F(ab’)2, Fv), single chain (ScFv) and domain antibodies (including, for example, shark and camelid antibodies), fusion proteins comprising an antibody, any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site, and antibody like binding peptidomimetics (ABiPs).
  • ABSiPs binding peptidomimetics
  • An antibody includes an antibody of any class, such as IgG, IgA, or IgM (or sub-class thereof), and the antibody need not be of any particular class.
  • immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgG - 1 , lgG-2, lgG-3, IgG -4, Ig A1 and lgA2.
  • the heavy-chain constant regions that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • the term "immunoglobulin” is used interchangeably with "antibody” herein.
  • the basic 4-chain antibody unit is a heterotetrameric glycoprotein composed of two identical light (L) chains and two identical heavy (H) chains.
  • An IgM antibody consists of 5 of the basic heterotetramer units along with an additional polypeptide called a J chain, and contains 10 antigen binding sites, while IgA antibodies comprise from 2-5 of the basic 4-chain units which can polymerize to form polyvalent assemblages in combination with the J chain.
  • the 4-chain unit is generally about 150,000 Daltons.
  • Each L chain is linked to an H chain by one covalent disulfide bond, while the two H chains are linked to each other by one or more disulfide bonds depending on the H chain isotype.
  • Each H and L chain also has regularly spaced intrachain disulfide bridges.
  • Each H chain has at the N-terminus, a variable domain (VH) followed by three constant domains (CH) for each of the a and y chains and four CH domains for p and £ isotypes.
  • Each L chain has at the N-terminus, a variable domain (VL) followed by a constant domain at its other end. The VL is aligned with the VH and the CL is aligned with the first constant domain of the heavy chain (CHI).
  • Particular amino acid residues are believed to form an interface between the light chain and heavy chain variable domains.
  • the pairing of a VH and VL together forms a single antigen-binding site.
  • the L chain from any vertebrate species can be assigned to one of two clearly distinct types, called kappa and lambda, based on the amino acid sequences of their constant domains.
  • immunoglobulins can be assigned to different classes or isotypes.
  • conventional (polyclonal) antibody preparations typically include a multitude of different antibodies having different amino acid sequences in their variable domains, particularly their CDRs, which are often specific for different epitopes.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al., Continuous cultures of fused cells secreting antibody of predefined specificity, Nature 1975, 256: 495; or may be made by recombinant DNA methods (e.g., U.S. Patent No. 4,816,567).
  • the "monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Making antibody fragments using phage display libraries, Nature 1991 , 352: 624-628 and Marks et al., By-passing immunization: human antibodies from V-gene libraries displayed on phage, J. Mol. Biol. 1991 , 222: 581-597, for example. See also Presta, Selection, design, and engineering of therapeutic antibodies, J. Allergy Clin. Immunol. 2005,116:731.
  • Chimeric antibody refers to an antibody in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in an antibody derived from a particular species (e.g., human) or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in an antibody derived from another species (e.g., mouse) or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity.
  • a particular species e.g., human
  • another species e.g., mouse
  • Human antibody refers to an antibody that comprises human immunoglobulin protein sequences only.
  • a human antibody may contain murine carbohydrate chains if produced in a mouse, in a mouse cell, or in a hybridoma derived from a mouse cell.
  • mouse antibody or rat antibody refer to an antibody that comprises only mouse or rat immunoglobulin sequences, respectively.
  • Humanized antibody refers to forms of antibodies that contain sequences from non-human (e.g., murine) antibodies as well as human antibodies. Such antibodies contain minimal sequence derived from non-human immunoglobulin.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • the prefix “hum,” “hu” or “h” is added to antibody clone designations when necessary to distinguish humanized antibodies from parental rodent antibodies.
  • the humanized forms of rodent antibodies will generally comprise the same CDR sequences of the parental rodent antibodies, although certain amino acid substitutions may be included to increase affinity, increase stability of the humanized antibody, or for other reasons.
  • substantially reduced denotes a sufficiently high degree of difference between two numeric values (generally one associated with a molecule and the other associated with a reference/comparator molecule) such that one of skill in the art would consider the difference between the two values to be of statistical significance within the context of the biological characteristic measured by said values (e.g., Kd values).
  • the difference between said two values is, for example, greater than about 10%, greater than about 20%, greater than about 30%, greater than about 40%, and/or greater than about 50% as a function of the value for the reference/comparator molecule.
  • the term “specifically binds to” or is “specific for” refers to measurable and reproducible interactions such as binding between a target and an antibody, which is determinative of the presence of the target in the presence of a heterogeneous population of molecules including biological molecules.
  • an antibody that specifically binds to a target (which can be an epitope) is an antibody that binds this target with greater affinity, avidity, more readily, and/or with greater duration than it binds to other targets.
  • the extent of binding of an antibody to an unrelated target is less than about 10 percent of the binding of the antibody to the target as measured, e.g., by a radioimmunoassay (RIA).
  • an antibody that specifically binds to a target has a dissociation constant (Kd) of ⁇ 1 pM, ⁇ 100 nM, ⁇ 10 nM, ⁇ 1 nM, or ⁇ 0.1 nM.
  • Kd dissociation constant
  • an antibody specifically binds to an epitope on a protein that is conserved among the protein from different species.
  • specific binding can include, but does not require exclusive binding.
  • immunoadhesin designates antibody-like molecules which combine the binding specificity of a heterologous protein (an “adhesin”) with the effector functions of immunoglobulin constant domains.
  • the immunoadhesins comprise a fusion of an amino acid sequence with the desired binding specificity which is other than the antigen recognition and binding site of an antibody (/.e., is “heterologous”), and an immunoglobulin constant domain sequence.
  • the adhesin part of an immunoadhesin molecule typically is a contiguous amino acid sequence comprising at least the binding site of a receptor or a ligand.
  • the immunoglobulin constant domain sequence in the immunoadhesin may be obtained from any immunoglobulin, such as lgG-1 , lgG-2 (including lgG2A and lgG2B), lgG-3, or lgG-4 subtypes, IgA (including IgA-1 and IgA-2), IgE, IgD or IgM.
  • the Ig fusions preferably include the substitution of a domain of a polypeptide or antibody described herein in the place of at least one variable region within an Ig molecule.
  • the immunoglobulin fusion includes the hinge, CH2 and CH3, or the hinge, CHI, CH2 and CH3 regions of an lgG-1 molecule.
  • Immunoadhesin combinations of Ig Fc and ECD of cell surface receptors are sometimes termed soluble receptors.
  • a “fusion protein” and a “fusion polypeptide” refer to a polypeptide having two portions covalently linked together, where each of the portions is a polypeptide having a different property.
  • the property may be a biological property, such as activity in vitro or in vivo.
  • the property may also be simple chemical or physical property, such as binding to a target molecule, catalysis of a reaction, etc.
  • the two portions may be linked directly by a single peptide bond or through a peptide linker but are in reading frame with each other.
  • an “antagonist” antibody or a “blocking” antibody is one that inhibits or reduces a biological activity of the antigen it binds.
  • blocking antibodies or antagonist antibodies substantially or completely inhibit the biological activity of the antigen.
  • the anti-PD-L1 antibodies of the invention block the signaling through PD-1 so as to restore a functional response by T-cells (e.g., proliferation, cytokine production, target cell killing) from a dysfunctional state to antigen stimulation.
  • agonist antibody or “activating antibody” is one that enhances or initiates signaling by the antigen to which it binds.
  • agonist antibodies cause or activate signaling without the presence of the natural ligand.
  • An small molecule “agonist” such as a TLR7/8 agonist of the present disclosure is a small molecule chemical that binds to a receptor and activates the receptor to produce a biological response.
  • the term "dysfunction" in the context of immune dysfunction refers to a state of reduced immune responsiveness to antigenic stimulation. The term includes the common elements of both exhaustion and/or anergy in which antigen recognition may occur, but the ensuing immune response is ineffective to control infection or tumor growth.
  • disfunctional also includes refractory or unresponsive to antigen recognition, specifically, impaired capacity to translate antigen recognition into down-stream T-cell effector functions, such as proliferation, cytokine production and/or target cell killing.
  • T cell anergy refers to the state of unresponsiveness to antigen stimulation resulting from incomplete or insufficient signals delivered through the T-cell receptor (e.g., increase in intracellular Ca+2 in the absence of ras- activation). T cell anergy can also result upon stimulation with antigen in the absence of co- stimulation, resulting in the cell becoming refractory to subsequent activation by the antigen even in the context of co stimulation.
  • the unresponsive state can often be overridden by the presence of lnterleukin-2. Anergic T-cells do not undergo clonal expansion and/or acquire effector functions.
  • exhaustion refers to T cell exhaustion as a state of T cell dysfunction that arises from sustained TCR signaling that occurs during many chronic infections and cancer. It is distinguished from anergy in that it arises not through incomplete or deficient signaling, but from sustained signaling. It is defined by poor effector function, sustained expression of inhibitory receptors and a transcriptional state distinct from that of functional effector or memory T cells. Exhaustion prevents optimal control of infection and tumors. Exhaustion can result from both extrinsic negative regulatory pathways (e.g., immunoregulatory cytokines) as well as cell intrinsic negative regulatory (co stimulatory) pathways.
  • extrinsic negative regulatory pathways e.g., immunoregulatory cytokines
  • Enhancing T-cell function means to induce, cause or stimulate a T-cell to have a sustained or amplified biological function, or renew or reactivate exhausted or dysfunctional T-cells.
  • enhancing T-cell function include: increased secretion of y-interferon from CD4+ or CD8+ T-cells, increased proliferation, increased survival, increased differentiation, increased antigen responsiveness (e.g., viral, pathogen, or tumor clearance) relative to such levels before the intervention.
  • the level of enhancement is as least 50%, alternatively 60%, 70%, 80%, 90%, 100%, 120%, 150%, 200%. The manner of measuring this enhancement is known to one of ordinary skill in the art.
  • Metastasis or “metastatic” is meant the spread of cancer from its primary site to other places in the body. Cancer cells can break away from a primary tumor, penetrate into lymphatic and blood vessels, circulate through the bloodstream, and grow in a distant focus (metastasize) in normal tissues elsewhere in the body. Metastasis can be local or distant. Metastasis is a sequential process, contingent on tumor cells breaking off from the primary tumor, traveling through the bloodstream, and stopping at a distant site. At the new site, the cells establish a blood supply and can grow to form a life-threatening mass. Both stimulatory and inhibitory molecular pathways within the tumor cell regulate this behavior, and interactions between the tumor cell and host cells in the distant site are also significant.
  • cancer refers to or describe the physiological condition in subjects that is typically characterized by unregulated cell growth.
  • cancer includes but is not limited to a primary cancer that originates at a specific site in the body, a metastatic cancer that has spread from the place in which it started to other parts of the body, a recurrence from the original primary cancer after remission, and a second primary cancer that is a new primary cancer in a person with a history of previous cancer of a different type from the latter one.
  • subject refers to a human or non-human subject including veterinary subjects such as, cows, sheep, cats, dogs, horses, primates, rabbits, and rodents (e.g., mice and rats) for which therapy is desired.
  • the subject is a human and may be referred to as a patient or individual, e.g., a human suffering from, at risk of suffering from, or potentially capable of suffering from cancer.
  • a human suffering from at risk of suffering from, or potentially capable of suffering from cancer.
  • Those skilled in the medical art are readily able to identify individual patients who are afflicted with cancer.
  • treat or “treating” a cancer, as used herein means to administer a combination therapy according to the present disclosure to a subject having cancer, or diagnosed with cancer, to achieve at least one positive therapeutic effect, such as, for example, reduced number of cancer cells, reduced tumor size, reduced rate of cancer cell infiltration into peripheral tissues, or reduced rate of tumor metastases or tumor growth, reversing, stopping, controlling, slowing, interrupting, arresting, alleviating, and/or inhibiting the progression or severity of a sign, symptom, disorder, condition, or disease, but does not necessarily involve a total elimination of all disease-related signs, symptoms, conditions, or disorders.
  • a positive therapeutic effect such as, for example, reduced number of cancer cells, reduced tumor size, reduced rate of cancer cell infiltration into peripheral tissues, or reduced rate of tumor metastases or tumor growth, reversing, stopping, controlling, slowing, interrupting, arresting, alleviating, and/or inhibiting the progression or severity of a sign, symptom, disorder, condition, or disease, but does not necessarily involve a total elimination
  • the term “treat” or “treating” also denotes, to arrest, delay the onset (i.e., the period prior to clinical manifestation of a disease or symptom of a disease) and/or reduce the risk of developing or worsening a symptom of a disease.
  • co-administration refers to the administration of one agent and in addition to at least one other agent, wherein said agents are part of the same or separate dosage formsand are administered via the same or different routes of administration and on the same or different administration schedule.
  • the co-administration of two or more agents can be useful for treating individuals suffering from cancer who have primary or acquired resistance to ongoing therapies.
  • the combination therapy provided herein may be useful for improving the efficacy and/or reducing the side effects of cancer therapies for individuals who do respond to such therapies.
  • the term “simultaneously,” “simultaneous administration,” “administered simultaneously,” “concurrently,” or “concurrent administration,” means that the agents are administered at the same point in time or immediately following one another, but that the agents can be administered in any order. For example, in the latter case, the two or more agents are administered at times sufficiently close that the results observed are indistinguishable from those achieved when the agents are administered at the same point in time.
  • the term simultaneous includes the administration of each agent of the combination therapy of the disclosure in the same medicament.
  • the agents of the present disclosure can be administered completely separately or in the form of one or more separate compositions.
  • the agents may be given separately at different times during the course of therapy (in a chronologically staggered manner, especially a sequence-specific manner) in such time intervals that the combination therapy is effective in treating cancer.
  • the term “sequential,” “sequentially,” “administered sequentially,” or “sequential administration” refers to the administration of each agent of the combination therapy of the invention, either alone or in a medicament, one after the other, wherein each agent can be administered in any order. Sequential administration may be particularly useful when the therapeutic agents in the combination therapy are in different dosage forms, for example, one agent is a tablet and another agent is a sterile liquid, and/or the agents are administered according to different dosing schedules, for example, one agent is administered daily, and the second agent is administered less frequently such as weekly.
  • composition refers to a preparation which is in such form as to permit the biological activity of the active ingredient to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered. Such formulations are sterile. “Pharmaceutically acceptable” carriers or excipients are those which can reasonably be administered to a subject to provide an effective dose of the active ingredient employed.
  • Suitable formulations include, for example, tablets, capsules, press coat formulations, and other easily administered formulations.
  • a "package insert” refers to instructions customarily included in commercial packages of medicaments that contain information about the indications customarily included in commercial packages of medicaments that contain information about the indications, usage, dosage, administration, contraindications, other medicaments to be combined with the packaged product, and/or warnings concerning the use of such medicaments, etc.
  • a therapeutically effective amount refers to that amount of a drug, agent, component, composition, compound, substance, targeted agent, targeted therapeutic agent, therapeutic antibody, therapeutic agent, medicament or pharmaceutical composition which is effective to achieve one or more of the following results following the administration of one or more therapies: (1) reducing the size of the tumor, (2) reducing the number of cancer cells, (3) inhibiting (/.e., slowing to some extent, preferably stopping) cancer cell infiltration into peripheral organs, (4) inhibiting (/.e., slowing to some extent, preferably stopping) tumor metastasis, (5) inhibiting (i.e., slowing to some extent, preferably stopping) tumor growth or tumor invasiveness, (6) relieving (i.e., to some extent, preferably eliminating) one or more signs or symptoms associated with the cancer, (7) decreasing the dose of other medications required to treat the disease, (8) enhancing the effect of another medication, (9) delaying the progression of the disease, (10) improving or increasing the disease-free, relapse-free, progression-free,
  • an effective amount can be administered in one or more administrations.
  • an effective amount of drug, compound, and/or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly.
  • an effective amount of a drug, compound, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition.
  • an “effective amount” may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
  • a therapeutic amount may also refer to a dosage of a drug that has been approved for use by a regulatory agency.
  • a "subtherapeutic amount,” as used herein, refers to a dosage of a drug that is significantly lower than the approved dosage, such as 10%, 20%, 30%, 40%, or 50% lower than the approved dosage.
  • treatment regimen “dosing protocol” and “dosing regimen” are used interchangeably to refer to the dose and timing of administration of each therapeutic agent in a combination of the invention.
  • Tumor as it applies to a subject diagnosed with, or suspected of having, a cancer refers to a malignant or potentially malignant neoplasm or tissue mass of any size and includes primary tumors and secondary neoplasms.
  • a solid tumor is an abnormal growth or mass of tissue that usually does not contain cysts or liquid areas. Examples of solid tumors are sarcomas, carcinomas, and lymphomas.
  • the optimum range for the effect and absolute dose ranges of each agent for the effect may be definitively measured by administration of the agents over different dose ranges, and/or dose ratios to subjects in need of treatment.
  • the observation of synergy in in vitro models or in vivo models can be predictive of the effect in humans and other species and in vitro models or in vivo models exist, as described herein, to measure a synergistic effect.
  • the results of such studies can also be used to predict effective dose and plasma concentration ratio ranges and the absolute doses and plasma concentrations required in humans and other species such as by the application of pharmacokinetic and / or pharmacodynamics methods.
  • pharmaceutically acceptable salt refers to pharmaceutically acceptable organic or inorganic salts of a compound of the invention. Some embodiments also relate to the pharmaceutically acceptable acid addition salts of the compounds described herein. Suitable acid addition salts are formed from acids which form non-toxic salts.
  • Non-limiting examples of suitable acid addition salts include, but are not limited to, the acetate, acid citrate, adipate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, bitartrate, borate, camsylate, citrate, cyclamate, edisylate, esylate, ethanesulfonate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobrom ide/brom ide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, methanesulfonate, methylsulphate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxa
  • Carriers include pharmaceutically acceptable carriers, excipients, or stabilizers that are nontoxic to the cell or subject being exposed thereto at the dosages and concentrations employed. Often the physiologically acceptable carrier is an aqueous pH buffered solution.
  • physiologically acceptable carriers include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid; low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfactants such as TWEENTM, polyethylene glycol (PEG), and PLURONICSTM.
  • buffers such as phosphate, citrate, and other organic acids
  • antioxidants including ascorbic acid
  • proteins such as serum albumin,
  • solvate describes a molecular complex comprising a compound described herein and one or more pharmaceutically acceptable solvent molecules, for example, water (hydrate) and ethanol.
  • tautomeric isomerism (‘tautomerism’) can occur. This can take the form of proton tautomerism in compounds described herein containing, for example, an imino, keto, or oxime group, or so-called valence tautomerism in compounds which contain an aromatic moiety. A single compound may exhibit more than one type of isomerism.
  • the compounds of the embodiments described herein include all stereoisomers (e.g., cis and trans isomers) and all optical isomers of compounds described herein (e.g., R and S enantiomers), as well as racemic, diastereomeric and other mixtures of such isomers. While all stereoisomers are encompassed within the scope of our claims, one skilled in the art will recognize that particular stereoisomers may be preferred.
  • TLRs Toll-like receptors
  • PAMPS pathogen-associated molecular patterns
  • TLR7 and TLR8 recognize nucleic acids and their degradation products.
  • the distribution of TLR3, TLR7, TLR8, and TLR9 is restricted to the endosomal compartments of cells and they are preferentially expressed in cells of the immune system.
  • TLR7 and TLR8 recognize single strand RNA at one ligand binding site and the ribonucleoside degradation products guanosine and uridine, respectively, (as well as small molecule ligands with related structural motifs) at a second ligand binding site.
  • TLR7 or TLR8 agonists Some small-molecule TLR7 or TLR8 agonists have been identified. Those agonists can be grouped into purine-like molecules, such as 7-thia-8-oxoguanosine (TOG, isatoribine) or the imidazoquinoline-based compounds such as imiquimod. Imiquimod is so far the only approved TLR7 agonist, marketed as a 5% cream (Aldara). It generates approximately 80% 5-year clearance of superficial basal cell carcinomas, which is the most common cancer worldwide, thus demonstrating the importance of TLR7 agonists in cancer immunotherapy.
  • TOG 7-thia-8-oxoguanosine
  • imiquimod imidazoquinoline-based compounds
  • TLR7/TLR8 small molecule agonist with dual bioactivity could provide further benefit over a more selective TLR7 agonist and would cause innate immune responses in a wider range of antigen presenting cells and other key immune cell types, including plasmacytoid and myeloid dendritic cells, monocytes, and B cells.
  • Such potent dual TLR7/8 agonists may also be effective in stimulating effective anti-tumor responses in cancer.
  • Embodiments of the present invention comprise a TLR7/8 agonist of Formula I: or a pharmaceutically acceptable salt thereof, wherein
  • R 1 and R 2 are independently Ci 3 alkyl; or
  • R 1 and R 2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
  • R 4 is Ci -6 alkyl, or (CH2)nO(CH2)mCH3, wherein said Ci -6 alkyl or any carbon of the (CH2)nO(CH2)mCH3 group is substituted with 0 to 3 F as valency allows;
  • R 5 is Ci-3 alkyl, or OC1 -3 alkyl, wherein the C1-3 alkyl is substituted by 0 to 3 F;
  • R 6 is H, or C1-3 alkyl, wherein the C1-3 alkyl is substituted with 0 to 3 F; m is 0 to 2; and n is 1 to 3.
  • the TLR7/8 agonist is of Formula I, wherein:
  • R 1 and R 2 are independently Ci 2 alkyl; or
  • R 1 and R 2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
  • R 4 is C3-5 alkyl, or (CH2)nO(CH2)mCH3;
  • R 5 is C1-2 alkyl
  • R 6 is H; m is 1 ; and n is 1 .
  • the TLR7/8 agonist may be selected from:
  • the preferred TLR7/8 agonist is 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5- c]pyridin-1-yl)methyl)-2-methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof.
  • the preferred TLR7/8 agonist is also referred as PF- 07225570 in this disclosure (or PF-5570 as referred in FIG.1-FIG.3) and has the following structure A:
  • the TLR7/8 agonist, especially the preferred PF- 07225570 or a pharmaceutically acceptable salt thereof may be administered with an interval of at least 2 days, at least 3 days, at least 4 days, at least 5 days, at least 6 days, at least 7 days, at least 10 days, at least 14 days, or at least 21 days.
  • the dose interval for the TLR7/8 agonist, especially the preferred PF- 07225570 or a pharmaceutically acceptable salt thereof may be about 2-21 days, 2-14 days, 2-10 days, 2-7 days, 3-21 days, 3-14 days, 3-10 days, 3-7 days, 5-21 days, 5-14 days, 5-10 days, or 5-7 days.
  • the dose interval for the TLR7/8 agonist is at least about 7 days (7 ⁇ 2 days), at least about 14 days (14 ⁇ 2 days), or at least about 21 days (21 ⁇ 2 days).
  • Embodiments of the present disclosure comprise a PD-1 axis binding antagonist.
  • the term “PD-1 axis binding antagonist” or “PD-1 axis antagonist” refers to a molecule that inhibits the interaction of a PD-1 axis binding partner (e.g., PD-1 , PD-L1 , PD-L2) with either one or more of its binding partners, for example so as to overcome or partially overcome T-cell dysfunction resulting from signaling on the PD-1 signaling axis — with a result being to restore, partially restore or enhance T-cell function (e.g., proliferation, cytokine production, target cell killing, survival).
  • a PD-1 axis binding partner e.g., PD-1 , PD-L1 , PD-L2
  • T-cell function e.g., proliferation, cytokine production, target cell killing, survival.
  • a PD-1 axis binding antagonist includes one or more of (i) a PD-1 antagonist, (ii) a PD-L1 antagonist, and/or (iii) a PD-L2 antagonist. i) PD-1 antagonist
  • PD-1 antagonist refers to a molecule that decreases, blocks, inhibits, abrogates or interferes with signal transduction resulting from the interaction of PD-1 with one or more of its binding partners, such as PD-L1 , PD-L2.
  • the PD-1 antagonist is a molecule that inhibits the binding of PD-1 to its binding partners.
  • the PD-1 antagonist inhibits the binding of PD-1 to PD-L1 and/or PD-L2.
  • the PD-1 antagonist inhibits the binding of PD-1 to PD-LI.
  • the PD-1 antagonist inhibits the binding of PD-1 to PD-L2.
  • the PD-1 antagonist inhibits the binding of PD-1 to both PD-L1 and PD-L2.
  • PD-1 antagonists include anti- PD-1 antibodies, antigen binding fragments thereof, immunoadhesins, fusion proteins, and other molecules that decrease, block, inhibit, abrogate or interfere with signal transduction resulting from the interaction of PD-1 with PD-L1 and/or PD-L2.
  • a PD-1 antagonist reduces the negative co-stimulatory signal mediated by or through cell surface proteins expressed on T lymphocytes mediated signaling through PD-1 so as to render a dysfunctional T-cell less non-dysfunctional.
  • the PD-1 antagonist is an anti-PD-1 antibody (aPD-1 ).
  • the PD-1 antagonist blocks binding of PD-L1 expressed on a cancer cell to PD-1 expressed on an immune cell (T cell, B cell or NKT cell) and preferably also blocks binding of PD-L2 expressed on a cancer cell to the immune-cell expressed PD-1.
  • PD-1 and its ligands include: PDCD1 , PD1 , CD279 and SLEB2 for PD-1 ; PDCD1 L1 , PDL1 , B7H1 , B7-4, CD274 and B7-H for PD-L1 ; and PDCD1 L2, PDL2, B7-DC, Btdc and CD273 for PD-L2.
  • the PD-1 antagonist may block binding of human PD- L1 to human PD-1 , and block binding of both human PD-L1 and PD-L2 to human PD-1.
  • PD-1 antagonists useful in any of the treatment methods, medicaments and uses of the present disclosure include a monoclonal antibody (mAb), or antigen binding fragment thereof, which specifically binds to PD-1 or PD-L1 , and preferably specifically binds to human PD-1 or human PD-L1.
  • the mAb may be a human antibody, a humanized antibody or a chimeric antibody, and may include a human constant region.
  • the human constant region is selected from the group consisting of lgG1 , lgG2, lgG3 and lgG4 constant regions, and in some embodiments, the human constant region is an IgG 1 or lgG4 constant region.
  • the antigen binding fragment is selected from the group consisting of Fab, Fab'-SH, F(ab')2, scFv and Fv fragments.
  • Specific anti-human PD-1 mAbs useful as the PD-1 antagonist in the treatment method, medicaments and uses of the present disclosure include: sasanlimab (RN888, PF-06801591 , Pfizer Inc.), nivolumab (OPDIVO®, ONO-4538, BMS-936558, MDX1106, Bristol-Myers Squibb Company), pembrolizumab (KEYTRUDA®, MK-3475, lambrolizumab, Merck & Co., Inc.), pidilizumab (CT-011), cemiplimab (LIBTAYO®, REGN2810, Regeneron Pharmaceuticals, Inc.), tislelizumab (BGB-A317, BeiGene Ltd./Celgene Corporation), spartalizumab (PDR001 , Novartis AG), sintilimab (IBI308, Innovent Biologies, Inc.), MEDI-0680 (AMP-5
  • WO2014204856A1 BGB-108 (CAS No.: 1859072-61-9, PCT Publication No. W02016007235A1 ), or AGEN2034 (Agenus Inc. CAS No.: 2230167-06-1 , PCT Publication No. WO2018106864A1 ), genolimzumab (CBT Pharmaceuticals, CAS No. 2135810-45-4, PCT Publication No. WO2017176965 A1 ), CBT-502 (CBT Pharmaceuticals, CAS No.: 2303884-58-2, PCT Publication No. WO2019176965 A1), camrelizumab (SHR-1210, Incyte Corporation), or any combination thereof.
  • the PD-1 antagonist is an anti-PD-1 antibody.
  • the anti-PD-1 antibody is nivolumab. In some embodiments, the anti-PD-1 antibody is pembrolizumab. In some embodiments, the anti-PD-1 antibody is sasanlimab.
  • Sasanlimab is a humanized, immunoglobulin G4 (lgG4) monoclonal antibody (mAb) that binds to the PD-1 receptor. By blocking its interaction with PD-L1 and PD-L2, PD-1 pathway-mediated inhibition of the immune response is released, leading to an anti-tumor immune response.
  • Clinical anti-tumor activity with sasanlimab has been seen in a panel of anti-PD1 sensitive solid tumor types including non-small cell lung cancer and urothelial carcinoma. Sasanlimab is described, for example, in US Patent No. US 10,155,037, which is hereby incorporated for all purposes.
  • Additional exemplary PD-1 antagonists include those described in U.S. Patent Application Publication 20130280265, U.S. Patent Application Publication 20130237580, U.S. Patent Application Publication 20130230514, U.S. Patent Application Publication 20130109843, U.S. Patent Application Publication 20130108651 , U.S. Patent Application Publication 20130017199, U.S. Patent Application Publication 20120251537, U.S. Patent Application Publication 20110271358, European Patent EP2170959B1 , in PCT Publication No. WO 2011/066342, PCT Publication No. WO 2015/035606, PCT Publication No. WO
  • exemplary PD-1 antagonists are described in Curran et al., PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors, PNAS, 2010, 107, 4275; Topalian etal., Safety, activity, and immune correlates of anti-PD-1 antibody in cancer, New Engl. J. Med. 2012, 366, 2443; Brahmer et al., Safety and activity of anti-PD-L1 antibody in patients with advanced cancer, New Engl. J. Med.
  • the term “PD-L1 antagonist,” as used herein, refers to a molecule that decreases, blocks, inhibits, abrogates or interferes with signal transduction resulting from the interaction of PD-L1 with either one or more of its binding partners, such as PD-1 , B7-1.
  • the PD-1 axis antagonist comprises a PD-L1 antagonist.
  • the PD-L1 antagonist inhibits the binding of PD-L1 to its binding partners.
  • the PD-L1 antagonist inhibits the binding of PD-L1 to PD-1 .
  • the PD-L1 antagonist inhibits the binding of PD-L1 to PD-1 and/or B7-1.
  • the PD-L1 antagonist inhibits the binding of PD-L1 to both PD-1 and B7-1.
  • the PD-L1 antagonists include anti-PD-L1 antibodies, antigen binding fragments thereof, immunoadhesins, fusion proteins, and other molecules that decrease, block, inhibit, abrogate or interfere with signal transduction resulting from the interaction of PD-L1 with one or more of its binding partners, such as PD-1 , and/or B7-1.
  • a PD-L1 antagonist reduces the negative costimulatory signal mediated by or through cell surface proteins expressed on T lymphocytes mediated signaling through PD-L1 so as render a dysfunctional T-cell less non-dysfunctional.
  • a PD-L1 antagonist is an anti-PD-L1 antibody (OPD-L1).
  • the PD-1 antagonist is an anti-PD-1 antibody.
  • the anti-PD-1 antibody useful for this disclosure include BMS-936559 (Bristol-Myers Squibb Company, CAS No. 1422185-22-5, PCT Publication No. WO2013019906 A1), KN035 (envafolimab, Jiangsu Alphamab biopharmaceuticals Co., Ltd., CAS No. 2102192-68-5, Zhang F, Wei H, Wang X, Bai Y, Wang P, Wu J, et al. Structural basis of a novel PD-L1 nanobody for immune checkpoint blockade. Cell Discov.
  • Atezolizumab (TECENTRIQ®, MPDL3280A, Roche Holding AG), durvalumab (IMFINZI®, AstraZeneca PLC), and/or avelumab (BAVENCIO®, Merck & Co., Inc. and Pfizer, Inc.).
  • Additional exemplary PD-L1 antagonists include those described in U.S. Patent Application Publication 20090055944, U.S. Patent Application Publication 20100203056, U.S. Patent Application Publication 20120039906, U.S. Patent Application Publication 20130045202, U.S. Patent Application Publication
  • PD-L2 antagonist refers to a molecule that decreases, blocks, inhibits, abrogates or interferes with signal transduction resulting from the interaction of PD-L2 with either one or more of its binding partners, such as PD-1.
  • a PD-L2 antagonist is a molecule that inhibits the binding of PD-L2 to its binding partners.
  • the PD-L2 antagonist inhibits binding of PD-L2 to PD-1.
  • the PD-L2 antagonists include anti- PD-L2 antibodies, antigen binding fragments thereof, immunoadhesins, fusion proteins, and other molecules that decrease, block, inhibit, abrogate or interfere with signal transduction resulting from the interaction of PD-L2 with either one or more of its binding partners, such as PD-1.
  • a PD-L2 antagonist reduces the negative co-stimulatory signal mediated by or through cell surface proteins expressed on T lymphocytes mediated signaling through PD-L2 so as render a dysfunctional T-cell less non-dysfunctional.
  • a PD-L2 antagonist is a PD-L2 immunoadhesin.
  • Monoclonal, polyclonal, and humanized antibodies can be prepared (e.g., Sheperd and Dean (eds.) Monoclonal Antibodies, 2000; Kontermann and Dubel (eds.) Antibody Engineering, 2001 ; Harlow and Lane, Antibodies A Laboratory Manual, 1988, pp. 139-243; Carpenter, et al., Non-Fc receptor-binding humanized anti-CD3 antibodies induce apoptosis of activated human T cells, J. Immunol. 2000, 165:6205; He, et al., Humanization and pharmacokinetics of a monoclonal antibody with specificity for both E-and P-selectin,J. Immunol.
  • mice An alternative to humanization is to use human antibody libraries displayed on phage or human antibody libraries in transgenic mice (Vaughan et al., Human antibodies with sub-nanomolar affinities isolated from a large non-immunized phage display library, Nature Biotechnol. 1996, 14:309-314; Barbas , Synthetic human antibodies, Nature Medicine 1995, 1 :837-839; Mendez et al., Functional transplant of megabase human immunoglobulin loci recapitulates human antibody response in mice, Nature Genetics 1997, 15:146-156; Hoogenboom and Chames, Natural and designer binding sites made by phage display technology, Immunol.
  • Animals can be immunized with cells bearing the antigen of interest. Splenocytes can then be isolated from the immunized animals, and the splenocytes can be fused with a myeloma cell line to produce a hybridoma (e.g., Meyaard, L., et.
  • a hybridoma e.g., Meyaard, L., et.
  • Antibodies can be conjugated, e.g., to small drug molecules, enzymes, liposomes, polyethylene glycol (PEG). Antibodies are useful for therapeutic, diagnostic, kit or other purposes, and include antibodies coupled, e.g., to dyes, radioisotopes, enzymes, or metals, e.g., colloidal gold (e.g., Le Doussal et al., Enhanced in vivo targeting of an asymmetric bivalent hapten to double-antigen-positive mouse B cells with monoclonal antibody conjugate cocktails, J. Immunol.
  • colloidal gold e.g., Le Doussal et al.
  • Methods for flow cytometry including fluorescence activated cell sorting (FACS), are available (e.g., Owens, etal., Flow Cytometry Principles for Clinical Laboratory Practice, 1994; Givan Flow Cytometry, 2nd ed.; 2001 ; Shapiro, Practical Flow Cytometry, 2003).
  • Fluorescent reagents suitable for modifying nucleic acids, including nucleic acid primers and probes, polypeptides, and antibodies, for use, e.g., as diagnostic reagents, are available (Molecular Probes, Catalogue, 2003; Sigma-Aldrich, Catalogue, 2003.
  • GenBank Vector NTI® Suite (Informax, Inc, Bethesda, MD); GCG Wisconsin Package (Accelrys, Inc., San Diego, CA); DeCypher® (TimeLogic Corp., Crystal Bay, Nevada); Menne, etal., A comparison of signal sequence prediction methods using a test set of signal peptides, Bioinformatics 2000, 16: 741-742; Menne, K.M.L., et. al.
  • the disclosure provides therapeutic methods and uses comprising administering to the subject the combinations as described herein, optionally in further combination with other therapeutic agents.
  • the disclosure provides a method for treating a cancer in a subject comprising administering to the subject a combination therapy of the disclosure.
  • the disclosure provides a method for treating a cancer comprising administering to a subject in need thereof an amount of a PD-1 axis binding antagonist and an amount of a TLR7/8 agonist, wherein the amounts together are effective in treating cancer, and wherein the TLR7/8 agonist is a compound of Formula I, or a pharmaceutically acceptable salt thereof, disclosed herein.
  • the subject is a human.
  • the method involves the use of an anti-PD-L1 antibody in combination with a TLR7/8 agonist described herein.
  • the method involves the use of an anti-PD-1 antibody selected from sasanlimab, nivolumab, pembrolizumab, pidilizumab, cemiplimab, tislelizumab, spartalizumab, sintilimab, MEDI-0680, BGB-108, AGEN2034 genolimzumab, CBT-502, camrelizumab, or any combination thereof, in combination with 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1-yl)methyl)-2- methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof.
  • an anti-PD-1 antibody selected from sasanlimab, nivolumab, pembrolizumab, pidilizumab, cemiplimab, tislelizumab, spartalizum
  • the method involves the use of sasanlimab, in combination with 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H- imidazo[4,5-c]pyridin-1 -yl)methyl)-2-methylpropane- 1 ,3-diol, or a pharmaceutically acceptable salt thereof.
  • the treatment results in sustained response in the individual after cessation of the treatment.
  • the methods of this disclosure may find use in treating conditions where enhanced immunogenicity is desired such as increasing tumor immunogenicity for the treatment of cancer. As such, a variety of cancers may be treated, or their progression may be delayed.
  • the individual has cancer that is resistant (has been demonstrated to be resistant) to one or more PD-1 axis binding antagonists.
  • resistance to PD-1 axis binding antagonist includes recurrence of cancer or refractory cancer. Recurrence may refer to the reappearance of cancer, in the original site or a new site, after treatment.
  • resistance to PD-1 axis binding antagonist includes progression of the cancer during treatment with the PD-1 axis binding antagonist.
  • resistance to PD-1 axis binding antagonist includes cancer that does not response to treatment. The cancer may be resistant at the beginning of treatment or it may become resistant during treatment. In some embodiments, the cancer is at early stage or at late stage.
  • the subject has received one, two, three, four, five or more prior cancer treatments. In other embodiments, the subject is treatment- naive. In some embodiments, the subject has progressed on other cancer treatments.
  • the prior cancer treatment comprised an immunotherapy. In other embodiments, the prior cancer treatment comprised a chemotherapy.
  • the tumor has reoccurred. In some embodiments, the tumor is metastatic. In other embodiments, the tumor is not metastatic.
  • the subject has received a prior therapy to treat the tumor and the tumor is relapsed or refractory. In some embodiments, the subject has received a prior immuno-oncology therapy to treat the tumor and the tumor is relapsed or refractory. In some embodiments, the subject has received more than one prior therapy to treat the tumor and the subject is relapsed or refractory.
  • the cancer is a solid tumor.
  • the disclosure is related to a method for treating cancer, wherein the cancer may be brain cancer, head/neck cancer (including squamous cell carcinoma of the head and neck (SCCHN)), prostate cancer, bladder cancer (including non-muscle invasive bladder cancer (NMIBC), or urothelial carcinoma, also known as transitional cell carcinoma (TCC)), lung cancer (including squamous cell carcinoma, small cell lung cancer (SCLC), and non-small cell lung cancer (NSCLC)), breast cancer, ovarian cancer, bone cancer, colorectal cancer, kidney cancer, liver cancer (including hepatocellular carcinoma (HCC)), pancreatic cancer, esophageal cancer (including sauamous cell carcinoma (SCCGC)), gastric cancer, gastroesophageal junction cancer, thyroid cancer, cervical cancer, uterine cancer, and/or renal cancer.
  • the cancer is bladder cancer.
  • the method of the present disclosure is for treating non-muscle invasive bladder cancer (NMIBC).
  • the combination therapy of the disclosure comprises administration of a PD-1 axis binding antagonist in combination with a TLR7/8 agonist.
  • each of the PD-1 axis binding antagonist and the TLR7/8 agonist may be administered in any suitable manner known in the art.
  • the PD-1 axis binding antagonist and the TLR7/8 agonist are administered simultaneously or sequentially in either order.
  • the disclosure provides a combination comprising: (i) a PD-1 axis binding antagonist described herein; and (ii) a TLR7/8 agonist of Formula I, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer in a subject.
  • the PD-1 axis binding antagonist is a PD-1 antagonist.
  • the PD-1 axis binding antagonist is a PD-L1 antagonist.
  • the disclosure provides a combination comprising: (i) an anti-PD-1 antibody; and (ii) 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5- c]pyridin-1-yl)methyl)-2-methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer in a subject.
  • the disclosure provides a combination comprising: (i) sasanlimab; and (ii) 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5- c]pyridin-1-yl)methyl)-2-methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof.
  • the combination is for use in the treatment of cancer in a subject.
  • Administration of the compounds of the disclosure may be affected by any method that enables delivery of the compounds to the site of action. These methods include oral routes, intraduodenal routes, parenteral injection (including intravenous, subcutaneous (sc), intramuscular, intravesical delivery such as intravesical instillation, intravascular or infusion), topical, and rectal administration.
  • parenteral injection including intravenous, subcutaneous (sc), intramuscular, intravesical delivery such as intravesical instillation, intravascular or infusion
  • topical and rectal administration.
  • the methods of administration of the agents and combinations herein may include oral, intravenous, intramuscular subcutaneous, topical, transdermal, intraperitoneal, by implantation, by inhalation, intrathecal, intraventricular, intravesical delivery such as intravesical instillation, or intranasal administration.
  • the method of administration for the TLR7/8 agonist is by intravesical instillation.
  • Dosage regimens may be adjusted to provide the optimum desired response. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the dose and dosing regimen is adjusted in accordance with methods well- known in the therapeutic arts. That is, the maximum tolerable dose can be readily established, and the effective amount providing a detectable therapeutic benefit to a patient may also be determined, as can the temporal requirements for administering each agent to provide a detectable therapeutic benefit to the patient. Accordingly, while certain dose and administration regimens are exemplified herein, these examples in no way limit the dose and administration regimen that may be provided to a patient in practicing the present disclosure.
  • the agents may be administered at their approved dosages. Treatment is continued as long as clinical benefit is observed or until unacceptable toxicity or disease progression occurs. Nevertheless, in certain embodiments, the combination therapies of the present disclosure may advantageously utilize lower dosages of the administered therapeutic agents, thus avoiding possible toxicities or complications associated with the various monotherapies.
  • the dosages of the agents administered are significantly lower than the approved dosage, e.g., a subtherapeutic dosage of the TLR7/8 agonist is administered in combination with a subtherapeutic dosage of a PD-1 axis binding antagonist.
  • a lower dosage of the agent may be desirable than when the agent alone is administered to a subject, a synergistic therapeutic effect may be achieved through the use of combination therapy which, in turn, permits use of a lower dose of the agent to achieve the desired therapeutic effect.
  • the dosages may be lower and may also be applied less frequently, which may diminish the incidence or severity of side-effects. This is in accordance with the desires and requirements of the subjects to be treated.
  • the amount of the agent of the disclosure administered will be dependent on the subject being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician.
  • An effective amount of the PD-1 axis binding antagonist and the TLR7/8 agonist may be administered for prevention or treatment of disease.
  • the appropriate dosage of the TLR7/8 agonist, and PD-1 axis binding antagonist may be determined based on the type of disease to be treated, the TLR7/8 agonist, the PD-1 axis binding antagonist, the severity and course of the disease, the clinical condition of the subject, the subject's clinical history and response to the treatment, and the discretion of the attending physician.
  • combination treatment with TLR7/8 agonist, and PD- 1 axis binding antagonist are synergistic, whereby an efficacious dose of the TLR7/8 agonist, PD-1 axis binding antagonist, in the combination is reduced relative to efficacious dose of the each of the TLR7/8 agonist, PD-1 axis binding antagonist, as a single agent.
  • the TLR7/8 agonist such as 2-((4-amino-2- (ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1-yl)methyl)-2-methylpropane- 1 ,3-diol, or a pharmaceutically acceptable salt thereof, can be administered with an interval of about at least 7 days (7 ⁇ 2 days), at least 14 days (14 ⁇ 2 days), or at least 21 days (21 ⁇ 2 days).
  • the efficacy observed see Table 1 to Table 5, and FIG. 1 to FIG. 3) for a single-dose treatment or with a weekly dose interval for a small molecule compound is very surprising. Such weekly dose interval, or longer interval, may provide more convenience and less toxicity to a patient being treated.
  • the disclosure provides a kit comprising: (i) a pharmaceutical composition comprising a PD-1 axis binding antagonist described herein; and (ii) a pharmaceutical composition comprising a TLR7/8 agonist described herein and a pharmaceutically acceptable carrier.
  • the kit further comprises package insert comprising instructions for using the TLR7/8 agonist in conjunction with PD-1 axis binding antagonist (e.g., anti-PD-1 or anti-PD-L1 antibody), treat or delay progression of cancer in a subject or to enhance immune function of a subject having cancer.
  • PD-1 axis binding antagonist e.g., anti-PD-1 or anti-PD-L1 antibody
  • any of the PD-1 axis binding antagonists and the TLR7/8 agonists described herein may be included in the kits.
  • the PD-1 binding antagonist is sasanlimab
  • the TLR7/8 agonist is 2-((4-amino-2- (ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1-yl)methyl)-2-methylpropane- 1 ,3-diol, or a pharmaceutically acceptable salt thereof.
  • the PD-1 axis binding antagonist and the TLR7/8 agonist and are stored in the same container or in separate containers. Suitable containers include, for example, bottles, vials, bags and syringes.
  • the container may be formed from a variety of materials such as glass, plastic (such as polyvinyl chloride or polyolefin), or metal alloy.
  • the container holds the formulation and the label on, or associated with, the container may indicate directions for use.
  • the kit may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • the kit further includes one or more of another agent (e.g., a chemotherapeutic agent, and anti-neoplastic agent).
  • Suitable containers for the one or more agent include, for example, bottles, vials, bags and syringes.
  • EB1 A method for treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of a Programmed Death 1 protein (PD-1) axis binding antagonist in combination with a compound of Formula I: or a pharmaceutically acceptable salt thereof, wherein
  • PD-1 Programmed Death 1 protein
  • R 1 and R 2 are independently Ci -3 alkyl; or
  • R 1 and R 2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
  • R 4 is C1-6 alkyl, or (CH2)nO(CH2)mCH3, wherein said C1-6 alkyl or any carbon of the (CH2)nO(CH2)mCH3 group is substituted with 0 to 3 F as valency allows;
  • R 5 is C1-3 alkyl, or OC1 -3 alkyl, wherein the C1-3 alkyl is substituted by 0 to 3 F;
  • R 6 is H, or C1-3 alkyl, wherein the C1-3 alkyl is substituted with 0 to 3 F; m is 0 to 2; and n is 1 to 3.
  • EB2 The method of EB1 , wherein:
  • R 1 and R 2 are independently Ci 2 alkyl; or
  • R 1 and R 2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
  • R 4 is C3-5 alkyl, or (CH2)nO(CH2)mCH 3 ;
  • R 5 is C1-2 alkyl
  • R 6 is H; m is 1 ; and n is 1 .
  • EB3 The method of EB1 , wherein the compound of Formula I is:
  • EB4 The method of any one of EB1 to EB3, wherein the compound of Formula I is 2- ((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1-yl)methyl)-2- methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof.
  • EB5 The method of any one of EB1 to EB4, wherein the PD-1 axis binding antagonist is an anti-PD-1 antibody selected from the group consisting of sasanlimab, nivolumab, pembrolizumab, pidilizumab, cemiplimab, tislelizumab, spartalizumab, sintilimab, MEDI- 0680, BGB-108, AGEN2034, genolimzumab, CBT-502, camrelizumab, and any combination thereof.
  • the PD-1 axis binding antagonist is an anti-PD-1 antibody selected from the group consisting of sasanlimab, nivolumab, pembrolizumab, pidilizumab, cemiplimab, tislelizumab, spartalizumab, sintilimab, MEDI- 0680, BGB-108, AGEN2034, genolimzum
  • EB6 The method of EB5, wherein the anti-PD-1 antibody is sasanlimab.
  • EB7 The method of any one of EB1 to EB4, wherein the PD-1 axis binding antagonist is an anti-PD-L1 antibody selected from the group consisting of BMS-936559, KN035, atezolizumab, durvalumab, avelumab, and any combination thereof.
  • the PD-1 axis binding antagonist is an anti-PD-L1 antibody selected from the group consisting of BMS-936559, KN035, atezolizumab, durvalumab, avelumab, and any combination thereof.
  • E8 The method of any one of EB1 to EB7, wherein the cancer is bladder cancer.
  • E9 The method of EB8, wherein the bladder cancer is non-muscle invasive bladder cancer (NMIBC).
  • NMIBC non-muscle invasive bladder cancer
  • E10 The method of EB9, wherein the NMIBC is Bacillus Calmette-Guerin (BCG) unresponsive NMIBC.
  • BCG Bacillus Calmette-Guerin
  • E11 The method of any one of EB1 to EB10, wherein the PD-1 axis binding antagonist is administered by intravenous infusion or subcutaneous delivery, and the compound of Formula I, or a pharmaceutically acceptable salt thereof, is administered by intravesical delivery to bladder epithelium of the subject.
  • E12 The method of any one of EB1 to EB11 , wherein a therapeutically effective amount of BCG immunotherapy is further administered.
  • EB13 The method of any one of EB4 to EB12, wherein 2-((4-amino-2-(ethoxymethyl)- 6,7-dimethyl-1 H-imidazo[4,5-c]pyridin- 1 -yl)methyl)-2-methylpropane- 1 ,3-diol, or a pharmaceutically acceptable salt thereof, is administered with a dosing interval of about at least 7 days (7 ⁇ 2 days).
  • EB14 A combination comprising a Programmed Death 1 protein (PD-1 ) axis binding antagonist and a compound of Formula I , or a pharmaceutically acceptable salt thereof, wherein the PD-1 axis binding antagonist comprises an anti-PD-1 antibody selected from the group consisting of sasanlimab, nivolumab, pembrolizumab, pidilizumab, cemiplimab, tislelizumab, spartalizumab, sintilimab, MEDI-0680, BGB-108, AGEN2034, genolimzumab, CBT-502, camrelizumab, and any combination thereof, or an anti-PD-L1 antibody selected from the group consisting of BMS-936559, KN035, atezolizumab, durvalumab, avelumab, and any combination thereof ; and wherein the compound of Formula I is: or a pharmaceutically acceptable salt thereof, wherein
  • R 1 and R 2 are independently Ci -3 alkyl; or
  • R 1 and R 2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
  • R 4 is Ci -6 alkyl, or (CH2)nO(CH2)mCH3, wherein said Ci -6 alkyl or any carbon of the (CH2)nO(CH2)mCH3 group is substituted with 0 to 3 F as valency allows;
  • R 5 is C1 -3 alkyl, or OC1 -3 alkyl, wherein the C1 -3 alkyl is substituted by 0 to 3 F;
  • R 6 is H, or C1-3 alkyl, wherein the C1-3 alkyl is substituted with 0 to 3 F;
  • m is 0 to 2; and
  • n is 1 to 3.
  • EB15 The combination of EB14, wherein:
  • R 1 and R 2 are independently Ci 2 alkyl; or
  • R 1 and R 2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
  • R 4 is C3-5 alkyl, or (CH2)nO(CH2)mCH 3 ;
  • R 5 is C1-2 alkyl
  • R 6 is H; m is 1 ; and n is 1 .
  • EB16 The combination of EB14 or EB15, wherein the PD-1 axis binding antagonist is sasanlimab, and the compound of Formula I is 2-((4-amino-2-(ethoxymethyl)-6,7- dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)methyl)-2-methylpropane- 1 ,3-diol, or a pharmaceutically acceptable salt thereof.
  • EB17 The combination of any one of EB14 to EB16, for use in the treatment of cancer.
  • EB18 The combination for use in the treatment of cancer of EB 17, wherein said cancer is bladder cancer
  • EB19 The combination for use in the treatment of cancer of EB18, wherein the bladder cancer is NMIBC.
  • EB20 The combination for use in the treatment of cancer of EB19, wherein the NMIBC is BCG un-responsive NMIBC.
  • EB21 The combination for use in the treatment of cancer of any one of EB18 to EB20, wherein 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1- yl)methyl)-2-methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof, is administered with a dosing interval of about at least 7 days).
  • EB22 The method of any of EB4 to EB13, wherein the compound 2-((4-amino-2- (ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1-yl)methyl)-2-methylpropane- 1 ,3-diol, or a pharmaceutically acceptable salt thereof, is administered with a dosing interval of at least about 14 days.
  • EB23 The method of any of EB1 to EB13, wherein the compound 2-((4-amino-2- (ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1-yl)methyl)-2-methylpropane- 1 ,3-diol, or a pharmaceutically acceptable salt thereof, is administered with a dosing interval of at least about 21 days.
  • EB24 The combination for use in the treatment of cancer of any one of EB18 to EB20, wherein the compound 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5- c]pyridin-1-yl)methyl)-2-methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof, is administered with a dosing interval of at least about 14 days.
  • EB25 The combination for use in the treatment of cancer of any one of EB18 to EB20, and EB24, wherein the compound 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H- imidazo[4,5-c]pyridin-1 -yl)methyl)-2-methylpropane- 1 ,3-diol, or a pharmaceutically acceptable salt thereof, is administered with a dosing interval of at least about 21 days.
  • Example 1 In vivo efficacy induced by combinatorial therapy of PF-07225570 and anti-PD-1 in MB49 syngeneic tumor model
  • PF-07225570 and murine surrogate anti-PD-1 (PF-06937004) in a syngeneic mouse bladder cancer model.
  • MB49 cells were implanted subcutaneously and PF-07225570 was tested through intra-tumor (IT) delivery to mimic local administration while anti-PD-1 was dosed Intra-peritoneally (IP) for systemic exposure.
  • I intra-tumor
  • IP Intra-peritoneally
  • RPMI Roswell Park Memorial Institute
  • DMEM Dulbecco’s Modified Eagle Medium
  • FBS Fetal bovine serum
  • DPBS Dulbecco’s Phosphate buffer solution
  • EDTA Ethylenediaminetetraacetic.
  • *PF-06937004 is a chimeric mAb (human anti-PD-1 CDR) on mouse lgG1 (mlgG1) frame. It binds to mouse PD-1 with an affinity of about 685 nM.
  • PF-06937004 is an anti-mouse PD-1 mlgG1 having the same heavy chain variable region (VH) and light chain variable region (VL) sequence of the PD1 -F2 clone described in PCT Publication No. WO 2004/056875 A1 , which is hereby incorporated for all purposes. No substantial mutation has been introduced into the mouse fragment crystallizable (Fc) regions.
  • Organ system evaluated Measurement of subcutaneous tumor growth and mouse body weight
  • mice Mus musculus/C57BI/6J mice
  • mice/group Female 10 mice/group
  • Q1 Dx1 Single dose; Q3Dx2: Dose every 3 days for a total of 2 doses; Q3Dx3: Dose every 3 days for atotal of 3 doses; QWx2: Dose every week foratotal of 2 doses.
  • Studylog 4.0 was used to acquire mouse body weights and tumor volumes.
  • GraphPad Prism 9.0.0 were used for analysis of mouse tumor volumes, body weights and survival.
  • Mice were treated with isotype or anti-PD-1 (IP) or with vehicle or PF- 07225570 (IT).
  • IP isotype or anti-PD-1
  • IT vehicle or PF- 07225570
  • anti-PD-1 was dosed on a Q3Dx3 schedule, and PF-07225570 treatment was altered testing Q3Dx3, Q3Dx2, Q1 Dx1 , or QWx2 dose strategy.
  • Body weight and tumor volume were measured twice a week throughout the study.
  • TGI was calculated with the following formula: 1 -(Tend-Tbegin/Cend-Cbegin) where Tend is final average tumor volume of treated group, Tbegin is average starting volume of treated group, Cend is final average volume of control group, and Cbegin is average starting volume of control group.
  • TGI was calculated on the first day an animal was lost in any group due to excessive tumor volume (for Vehicle + Isotype controls, TGI was assessed at day 20 and for single agent controls, TGI was assessed at day 24).
  • TGI was assessed at day 24.
  • Anti-PD-1 treatment remained consistent with Q3Dx3 IP dosing and PF- 07225570 IT treatment was altered to explore how reducing frequency (Q3Dx3, Q3Dx2, Q1 Dx1) or increasing length of time (QWx2) between dosing would affect TGI, number of responders or survival compared to isotype + vehicle controls or single agent treatments.
  • TGI tumor growth factor receptor 1
  • p-values are summarized in Table 1 and FIG. 1. No significant body weight loss was observed in any group.
  • Significant single agent efficacy of anti-PD-1 and PF-07225570 was observed in this model when using vehicle + isotype as the control using TGI calculated at day 20 post implant.
  • TGI and p-values are summarized in Table 2 (anti-PD-1 single agent as the control), Table 3 (PF-07225570 single agent as the control), and FIG.
  • Anti-PD-1 in MB-49 Syngeneic Model Compared to Vehicle + Isotype Control p-values determined by ANCOVA and TGI were determined on day 20 with Vehicle + Isotype as control using Pfizer-internal TGI analyzer software. Number of animals per group 10.
  • Responders were defined as having two consecutive tumor measurements that were lower than previous measurements after randomization and dosing began. The number of responders and p-values are summarized in Table 4.
  • Table 4 Number of Responders in Combinatorial Study of PF-07225570 and
  • single agent PF-07225570 and single agent anti-PD-1 provided statistically significant TGI and survival benefit compare to vehicle + isotype controls. While the number of responders was not significantly different comparing single agent therapy to vehicle + isotype controls, all combination therapies, regardless of PF- 07225570 dose regimen tested, resulted in an increased number of responders, significant TGI at day 20 post implant, and significant survival advantage compared to vehicle + isotype treated controls.
  • the combination of anti-PD-1 and PF-07225570 dosed Q3Dx3 provided significant survival and TGI at day 24 post implant over anti-PD- 1 and PF-07225570 single agent therapy.

Abstract

The present disclosure relates to combination therapies useful for the treatment of cancers. The combination therapy comprises a PD-1 axis binding antagonist, in combination with a Toll-like receptor 7/8 (TLR7/8) agonist or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising such compounds or salts. In one preferred combination therapy, the TLR7/8 agonist is 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1H-imidazo[4,5-c]pyridin-1-yl)methyl)-2-methylpropane-1,3-diol, or a pharmaceutically acceptable salt thereof, and the cancer to be treated is non-muscle invasive bladder cancer.

Description

COMBINATION THERAPIES USING TLR7/8 AGONIST
FIELD
The present disclosure relates to combination therapies useful for the treatment of cancers. Certain embodiments relate to a combination therapy which comprises a PD-1 axis binding antagonist, in combination with a Toll-like receptor 7/8 (TLR7/8) agonist or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising such compounds or salts. The disclosure also relates to associated methods of treatment, pharmaceutical combinations, and pharmaceutical uses. The methods and combinations are useful for any indication for which the therapeutic is itself useful in the detection, treatment and/or prevention of a disease, disorder, or other condition of a subject.
BACKGROUND
Bladder cancer accounts for approximately 6.6% of all cancer cases and 70-75% are Non-muscle invasive bladder cancer (NMIBC). There is an increasing need for alternative therapies targeting NMIBC.
Currently, balder cancer patients generally undergo transurethral resection of the bladder tumor (TURBT) and Intravesical (IVe) adjuvant chemotherapy (mitomycin) or immunotherapy with Bacillus Calmette-Guerin (BCG) to prevent recurrence. While BCG is well-entrenched, there is a need for options that delay progression to muscle invasive disease and prevent recurrence. In addition, a worldwide BCG shortage has increased the need for BCG alternatives.
There remains a need for new therapies for the treatment of cancers such as bladder cancers. Preferred combination therapies of the present invention show greater efficacy than treatment with either therapeutic agent alone.
SUMMARY
This disclosure relates to therapeutic methods, combinations, and combinations for use in the treatment of cancer. Also provided are combination therapies comprising the compounds of the disclosure, in combination with other therapeutic agents or therapies. The present disclosure also provides kits comprising one or more of the combinations of the disclosure. In one embodiment, the disclosure provides a method for treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of a Programmed Death 1 protein (PD-1 ) axis binding antagonist, in combination with a compound of Formula I:
Figure imgf000003_0001
or a pharmaceutically acceptable salt thereof, wherein
R1 and R2 are independently Ci 3 alkyl; or
R1 and R2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
R3 is
Figure imgf000003_0002
R4 is C1-6 alkyl, or (CH2)nO(CH2)mCH3, wherein said C1 -6 alkyl or any carbon of the (CH2)nO(CH2)mCH3 group is substituted with 0 to 3 F as valency allows;
R5 is 0-3 alkyl, or OC1 -3 alkyl, wherein the C1-3 alkyl is substituted by 0 to 3 F;
R6 is H, or C1-3 alkyl, wherein the C1-3 alkyl is substituted with 0 to 3 F; m is 0 to 2; and n is 1 to 3.
In some embodiments of the methods as described herein, the compound is of Formula I, wherein:
R1 and R2 are independently Ci 2 alkyl; or
R1 and R2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
Figure imgf000004_0001
R4 is C3-5 alkyl, or (CH2)nO(CH2)mCH3;
R5 is C1-2 alkyl;
R6 is H; m is 1 ; and n is 1 .
In some embodiments of the methods as described herein, the compound of Formula I is selected from:
2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)methyl)- 2-methylpropane-1 ,3-diol,
2-((4-amino-2-(ethoxymethyl)-1 /7-imidazo[4,5-c]qu inolin- 1-yl)methyl)-2- methylpropane-1 ,3-diol,
2-((4-amino-2-(ethoxymethyl)-7,8-dihydrocyclopenta[b]imidazo[4,5-d]pyridin-
1 (6H)-yl)methyl)-2-methylpropane-1 ,3-diol,
2-((4-amino-2-(ethoxymethyl)-6,7,8,9-tetrahydro-1 H-imidazo[4,5-c]quinolin-1 - yl)methyl)-2-methylpropane-1 ,3-diol,
3-(4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)-2-
(methoxymethyl)-2-methylpropan-1 -ol,
(R)-3-(4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin- 1 -y l)-2- (methoxymethyl)-2-methylpropan-1 -ol,
(S)-3-(4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)-2- (methoxymethyl)-2-methylpropan-1 -ol,
2-((4-amino-6,7-dimethyl-2-(2,2,2-trifluoroethyl)-1 H-imidazo[4,5-c]pyridin- 1 - yl)methyl)-2-methylpropane-1 ,3-diol,
2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)methyl)- 2-ethylpropane-1 ,3-diol,
2-((4-amino-2-butyl-1 H-imidazo[4,5-c]quinolin-1-yl)methyl)-2-methylpropane-1 ,3- diol,
2-((4-amino-2-butyl-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1-yl)methyl)-2- methylpropane-1 ,3-diol, and
2-((4-amino-2-pentyl-1 H-imidazo[4, 5-c]quinolin-1 -yl)methyl)-2-methylpropane-
1 ,3-diol; or a pharmaceutically acceptable salt thereof.
In one preferred embodiment of the methods as described herein, the compound of Formula I is preferably 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5- c]pyridin-1-yl)methyl)-2-methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof. The preferred compound is also known as PF- 07225570 and has the following structure A:
Figure imgf000005_0001
A
In some embodiments of the methods as described herein, the PD-1 axis binding antagonist comprises a PD-1 binding antagonist, or a PD-L1 binding antagonist.
In some embodiments, the PD-1 binding antagonist is an anti-PD-1 antibody. In a more specific embodiment, the anti-PD-1 antibody is sasanlimab, nivolumab, pembrolizumab, pidilizumab, cemiplimab, tislelizumab, spartalizumab, sintilimab, MEDI- 0680, BGB-108, AGEN2034, genolimzumab, CBT-502, camrelizumab, or any combination thereof.
In one preferred embodiment of the methods as described herein, the preferred anti-PD-1 antibody is sasanlimab.
In some embodiments, the PD-L1 binding antagonist is an anti-PD-L1 antibody. In a more specific embodiment, the anti-PD-L1 antibody is BMS-936559, KN035, atezolizumab, durvalumab, avelumab, or any combination thereof.
In one preferred embodiment of the methods as described herein, the cancer being treated is bladder cancer. In one more preferred embodiment, the cancer being treated is non-muscle invasive bladder cancer. In one even more preferred aspect, the non-muscle invasive bladder cancer is Bacillus Calmette-Guerin (BCG) un-responsive non-muscle invasive bladder cancer.
In some embodiments, the PD-1 axis binding antagonist is administered by intravenous infusion or subcutaneous delivery, and the compound of Formula I, or a pharmaceutically acceptable salt thereof, is administered by intravesical delivery to bladder epithelium of the subject.
In one embodiment of the methods as described herein, wherein a therapeutically effective amount of BCG immunotherapy may be further administered. BCG immunotherapy in this disclosure refers to a weakened, live bacterium, bacillus Calmette-Guerin (BCG), which is FDA-approved immunotherapy and helps reduce the risk of bladder cancer recurrence by stimulating an immune response that targets the bacteria as well as any nearby bladder cancer cells.
In one embodiment of the methods as described herein, the compound 2-((4- amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)methyl)-2- methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof, is administered with an interval of at least about 7 days (7± 2 days).
In one aspect, the disclosure provides a combination therapy comprising a PD-1 axis binding antagonist and a TLR7/8 agonist, as provided in this disclosure, for use in the treatment of cancer in a subject in need thereof.
In one aspect, the disclosure provides a combination of a PD-1 axis binding antagonist and a TLR7/8 agonist, for use in the treatment of cancer in a subject. Such combination can be used, for example, in the methods of the present disclosure.
In one aspect, the disclosure provides a medicament comprising a TLR7/8 agonist for use, in combination with a PD-1 axis binding antagonist, for treating a cancer.
In one aspect, the disclosure provides a medicament comprising a PD-1 axis binding antagonist for use, in combination with a TLR7/8 agonist in the treatment of a cancer.
In one aspect, the disclosure provides the use of a combination comprising a PD-1 axis binding antagonist and a TLR7/8 agonist for the treatment of cancer in a subject in need thereof.
In one aspect, the disclosure provides a kit that comprises a PD-1 axis binding antagonist, a TLR7/8 agonist, and a package insert, and the package insert comprises instructions for treating a subject for cancer using the medicaments. Such kit can be used, for example, in the methods of the present disclosure.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 shows tumor growth inhibition in MB49 subcutaneous tumor model with various combinatorial strategies of PF-07225570 and Anti-PD-1 compared to Vehicle + Isotype control. Data and p-values for FIG. 1 are presented in Table 1. Number of animals per group = 10. The statistical significance of differences between experimental groups was assessed by ANCOVA using Pfizer-internal TGI analyzer software. **, p<0.01 ; *“*, p<0.0001 Data is depicted as mean ± SEM. FIG. 2 shows tumor growth inhibition in MB49 subcutaneous tumor model with various combinatorial strategies of PF-07225570 and Anti-PD-1 compared to Single Agent Anti-PD-1 or Single Agent PF-07225570. Data and p-values for FIG. 2 are presented in Table 2 and Table 3. Number of animals per group = 10. The statistical significance of differences between experimental groups was assessed by ANCOVA using Pfizer-internal TGI analyzer software. # p=0.0592; *, p<0.05. Data is depicted as mean ± SEM.
FIG. 3 shows survival in MB49 subcutaneous tumor model with various combinatorial strategies of PF-07225570 and Anti-PD-1. Data and p-values for FIG. 3 are presented in Table 5. Number of animals per group = 10. Statistical significance in survival determined using Logrank Mantel-cox test compared to Vehicle + Isotype using GraphPad Prism. *,p<0.05, **, p<0.01 ; ****, p<0.0001.
DETAILED DESCRIPTION
Each of the embodiments described below can be combined with any other embodiment described herein not inconsistent with the embodiment with which it is combined.
I. Definitions
As used herein, the singular form "a," "an," and "the" include plural references unless indicated otherwise. For example, "a" substituent includes one or more substituents. Where the plural form is used for compounds, salts, and the like, this is taken to mean also a single compound, salt, or the like.
The term “about” when used to modify a numerically defined parameter means that the parameter may vary by as much as 10% above or below the stated numerical value for that parameter.
The term "alkyl" refers to a saturated, monovalent aliphatic hydrocarbon radical including straight chain and branched chain groups having the specified number of carbon atoms. The alkyl substituents disclosed herein may be specified as 1 to 6 carbon atoms (“Ci -6 alkyl”), or 1 to 3 carbon atoms (“Ci -3 alkyl”), and so forth. Examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, n-heptyl, n-octyl and the like.
The term “alkoxy” refers to a monovalent -O-alkyl group, wherein the alkyl portion has the specified number of carbon atoms. Alkoxy groups typically contain 1 to 8 carbon atoms (“Ci -8 alkoxy”), or 1 to 6 carbon atoms (“Ci -6 alkoxy”), or 1 to 4 carbon atoms (“Ci -4 alkoxy”). For example, C1-4 alkoxy includes methoxy, ethoxy, isopropoxy, tert-butyloxy (i.e., -OCH3, -OCH2CH3, -OCH(CH3)2, -OC(CH3)3), and the like.
As used herein, terms, including, but not limited to, “drug,” “agent,” “component,” “composition," “compound," “substance," “targeted agent," “targeted therapeutic agent," “therapeutic agent,” and “medicament” may be used interchangeably to refer to the small molecule compounds of the present disclosure, e.g., a TLR7/8 agonist, an anti- PD-L1 antibody, an anti-PD-1 antibody, or combinations thereof.
As used herein, terms “therapeutic antibody” and “antibody” may be used interchangeable to refer to an antibody that is used in the treatment of a disease or a disorder. A therapeutic antibody may have various mechanisms of action. A therapeutic antibody may bind and neutralize the normal function of a target associated with an antigen. For example, a monoclonal antibody that blocks the activity of the protein needed for the survival of a cancer cell causes the cell's death. Another therapeutic antibody may bind and activate the normal function of a target associated with an antigen. For example, a monoclonal antibody can bind to a protein on a cell and trigger an apoptosis signal. Yet another monoclonal antibody may bind to a target antigen expressed only on diseased tissue; conjugation of a toxic payload (effective agent), such as a chemotherapeutic or radioactive agent, to the monoclonal antibody can create an agent for specific delivery of the toxic payload to the diseased tissue, reducing harm to healthy tissue. A “biologically functional fragment” of a therapeutic antibody will exhibit at least one if not some or all of the biological functions attributed to the intact antibody, the function comprising at least specific binding to the target antigen.
The therapeutic antibody may bind to any protein, including, without limitation, a PD-L1 , a PD-1. Accordingly, therapeutic antibodies include, without limitation, anti-PD- L1 antibodies, anti-PD-1 antibodies, or combinations thereof.
The terms “synergy” or “synergistic” are used to mean that the result of the combination of two or more compounds, components or targeted agents is greater than the sum of each agent together. The terms “synergy” or “synergistic” also means that there is an improvement in the disease condition or disorder being treated, over the use of the two or more compounds, components or targeted agents while each compound, component or targeted agent individually. This improvement in the disease condition or disorder being treated is a “synergistic effect”. A “synergistic amount” is an amount of the combination of the two compounds, components or targeted agents that results in a synergistic effect, as “synergistic” is defined herein. Determining a synergistic interaction between one or two components, the optimum range for the effect and absolute dose ranges of each component for the effect may be definitively measured by administration of the components over different w/w (weight per weight) ratio ranges and doses to patients in need of treatment. However, the observation of synergy in in vitro models or in vivo models can be predictive of the effect in humans and other species and in vitro models or in v/vomodels exist, as described herein, to measure a synergistic effect and the results of such studies can also be used to predict effective dose and plasma concentration ratio ranges and the absolute doses and plasma concentrations required in humans and other species by the application of pharmacokinetic/pharmacodynamic methods.
A "chemotherapeutic agent" is a chemical compound useful in the treatment of cancer.
The term "immunotherapy" refers to the treatment of a subject by a method comprising inducing, enhancing, suppressing, or otherwise modifying an immune response.
The terms “abnormal cell growth” as used herein, unless otherwise indicated, refers to cell growth that is independent of normal regulatory mechanisms (e.g., loss of contact inhibition). Abnormal cell growth may be benign (not cancerous), or malignant (cancerous).
A “disorder” is any condition that would benefit from treatment with the compounds of the present invention. This includes chronic and acute disorders or diseases including those pathological conditions which predispose the subject to the disorder in question.
The term “antibody,” as used herein, refers to an immunoglobulin molecule capable of specific binding to a target, such as a carbohydrate, polynucleotide, lipid, polypeptide, etc., through at least one antigen recognition site, located in the variable region of the immunoglobulin molecule. As used herein, the term encompasses a polyclonal antibody, a monoclonal antibody, a chimeric antibody, a bispecific antibody, a dual-specific antibody, bifunctional antibody, a trispecific antibody, a multispecific antibody, a bispecific heterodimeric diabody, a bispecific heterodimeric IgG, a labeled antibody, a humanized antibody, a human antibody, and fragments thereof (such as Fab, Fab’, F(ab’)2, Fv), single chain (ScFv) and domain antibodies (including, for example, shark and camelid antibodies), fusion proteins comprising an antibody, any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site, and antibody like binding peptidomimetics (ABiPs). An antibody includes an antibody of any class, such as IgG, IgA, or IgM (or sub-class thereof), and the antibody need not be of any particular class. Depending on the antibody amino acid sequence of the constant region of its heavy chains, immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgG - 1 , lgG-2, lgG-3, IgG -4, Ig A1 and lgA2. The heavy-chain constant regions that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
The term "immunoglobulin" (Ig) is used interchangeably with "antibody" herein. The basic 4-chain antibody unit is a heterotetrameric glycoprotein composed of two identical light (L) chains and two identical heavy (H) chains. An IgM antibody consists of 5 of the basic heterotetramer units along with an additional polypeptide called a J chain, and contains 10 antigen binding sites, while IgA antibodies comprise from 2-5 of the basic 4-chain units which can polymerize to form polyvalent assemblages in combination with the J chain. In the case of IgGs, the 4-chain unit is generally about 150,000 Daltons. Each L chain is linked to an H chain by one covalent disulfide bond, while the two H chains are linked to each other by one or more disulfide bonds depending on the H chain isotype. Each H and L chain also has regularly spaced intrachain disulfide bridges. Each H chain has at the N-terminus, a variable domain (VH) followed by three constant domains (CH) for each of the a and y chains and four CH domains for p and £ isotypes. Each L chain has at the N-terminus, a variable domain (VL) followed by a constant domain at its other end. The VL is aligned with the VH and the CL is aligned with the first constant domain of the heavy chain (CHI). Particular amino acid residues are believed to form an interface between the light chain and heavy chain variable domains. The pairing of a VH and VL together forms a single antigen-binding site. For the structure and properties of the different classes of antibodies, e.g., Daniel P. Sties, Abba I. Terr and Tristram G. Parsolw (eds), Basic and Clinical Immunology, 8th Edition, 1994, page 71 and Chapter 6. The L chain from any vertebrate species can be assigned to one of two clearly distinct types, called kappa and lambda, based on the amino acid sequences of their constant domains. Depending on the amino acid sequence of the constant domain of their heavy chains (CH), immunoglobulins can be assigned to different classes or isotypes.
"Monoclonal antibody" or “mAb” or “Mab,” as used herein, refers to a population of substantially homogeneous antibodies, i.e., the antibody molecules comprising the population are identical in amino acid sequence except for possible naturally occurring mutations that may be present in minor amounts. In contrast, conventional (polyclonal) antibody preparations typically include a multitude of different antibodies having different amino acid sequences in their variable domains, particularly their CDRs, which are often specific for different epitopes. The modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al., Continuous cultures of fused cells secreting antibody of predefined specificity, Nature 1975, 256: 495; or may be made by recombinant DNA methods (e.g., U.S. Patent No. 4,816,567). The "monoclonal antibodies" may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Making antibody fragments using phage display libraries, Nature 1991 , 352: 624-628 and Marks et al., By-passing immunization: human antibodies from V-gene libraries displayed on phage, J. Mol. Biol. 1991 , 222: 581-597, for example. See also Presta, Selection, design, and engineering of therapeutic antibodies, J. Allergy Clin. Immunol. 2005,116:731.
"Chimeric antibody" refers to an antibody in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in an antibody derived from a particular species (e.g., human) or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in an antibody derived from another species (e.g., mouse) or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity.
“Human antibody” refers to an antibody that comprises human immunoglobulin protein sequences only. A human antibody may contain murine carbohydrate chains if produced in a mouse, in a mouse cell, or in a hybridoma derived from a mouse cell. Similarly, “mouse antibody” or “rat antibody” refer to an antibody that comprises only mouse or rat immunoglobulin sequences, respectively.
"Humanized antibody" refers to forms of antibodies that contain sequences from non-human (e.g., murine) antibodies as well as human antibodies. Such antibodies contain minimal sequence derived from non-human immunoglobulin. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence. The humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. The prefix “hum,” “hu” or “h” is added to antibody clone designations when necessary to distinguish humanized antibodies from parental rodent antibodies. The humanized forms of rodent antibodies will generally comprise the same CDR sequences of the parental rodent antibodies, although certain amino acid substitutions may be included to increase affinity, increase stability of the humanized antibody, or for other reasons.
The phrase "substantially reduced," "substantially different," or “substantially inhibit,” as used herein, denotes a sufficiently high degree of difference between two numeric values (generally one associated with a molecule and the other associated with a reference/comparator molecule) such that one of skill in the art would consider the difference between the two values to be of statistical significance within the context of the biological characteristic measured by said values (e.g., Kd values). The difference between said two values is, for example, greater than about 10%, greater than about 20%, greater than about 30%, greater than about 40%, and/or greater than about 50% as a function of the value for the reference/comparator molecule.
As use herein, the term "specifically binds to" or is "specific for" refers to measurable and reproducible interactions such as binding between a target and an antibody, which is determinative of the presence of the target in the presence of a heterogeneous population of molecules including biological molecules. For example, an antibody that specifically binds to a target (which can be an epitope) is an antibody that binds this target with greater affinity, avidity, more readily, and/or with greater duration than it binds to other targets. In one embodiment, the extent of binding of an antibody to an unrelated target is less than about 10 percent of the binding of the antibody to the target as measured, e.g., by a radioimmunoassay (RIA). In certain embodiments, an antibody that specifically binds to a target has a dissociation constant (Kd) of < 1 pM, < 100 nM, < 10 nM, < 1 nM, or < 0.1 nM. In certain embodiments, an antibody specifically binds to an epitope on a protein that is conserved among the protein from different species. In another embodiment, specific binding can include, but does not require exclusive binding.
As used herein, the term "immunoadhesin" designates antibody-like molecules which combine the binding specificity of a heterologous protein (an "adhesin") with the effector functions of immunoglobulin constant domains. Structurally, the immunoadhesins comprise a fusion of an amino acid sequence with the desired binding specificity which is other than the antigen recognition and binding site of an antibody (/.e., is "heterologous"), and an immunoglobulin constant domain sequence. The adhesin part of an immunoadhesin molecule typically is a contiguous amino acid sequence comprising at least the binding site of a receptor or a ligand. The immunoglobulin constant domain sequence in the immunoadhesin may be obtained from any immunoglobulin, such as lgG-1 , lgG-2 (including lgG2A and lgG2B), lgG-3, or lgG-4 subtypes, IgA (including IgA-1 and IgA-2), IgE, IgD or IgM. The Ig fusions preferably include the substitution of a domain of a polypeptide or antibody described herein in the place of at least one variable region within an Ig molecule. In a particularly preferred embodiment, the immunoglobulin fusion includes the hinge, CH2 and CH3, or the hinge, CHI, CH2 and CH3 regions of an lgG-1 molecule. For the production of immunoglobulin fusions, see also US Patent No. 5,428,130 issued June 27, 1995. Immunoadhesin combinations of Ig Fc and ECD of cell surface receptors are sometimes termed soluble receptors.
A "fusion protein" and a "fusion polypeptide" refer to a polypeptide having two portions covalently linked together, where each of the portions is a polypeptide having a different property. The property may be a biological property, such as activity in vitro or in vivo. The property may also be simple chemical or physical property, such as binding to a target molecule, catalysis of a reaction, etc. The two portions may be linked directly by a single peptide bond or through a peptide linker but are in reading frame with each other.
An "antagonist” antibody or a "blocking" antibody is one that inhibits or reduces a biological activity of the antigen it binds. In some embodiments, blocking antibodies or antagonist antibodies substantially or completely inhibit the biological activity of the antigen. The anti-PD-L1 antibodies of the invention block the signaling through PD-1 so as to restore a functional response by T-cells (e.g., proliferation, cytokine production, target cell killing) from a dysfunctional state to antigen stimulation.
An "agonist antibody" or “activating antibody” is one that enhances or initiates signaling by the antigen to which it binds. In some embodiments, agonist antibodies cause or activate signaling without the presence of the natural ligand.
An small molecule “agonist” such as a TLR7/8 agonist of the present disclosure is a small molecule chemical that binds to a receptor and activates the receptor to produce a biological response. The term "dysfunction" in the context of immune dysfunction, refers to a state of reduced immune responsiveness to antigenic stimulation. The term includes the common elements of both exhaustion and/or anergy in which antigen recognition may occur, but the ensuing immune response is ineffective to control infection or tumor growth.
The term "dysfunctional", as used herein, also includes refractory or unresponsive to antigen recognition, specifically, impaired capacity to translate antigen recognition into down-stream T-cell effector functions, such as proliferation, cytokine production and/or target cell killing.
The term "anergy" refers to the state of unresponsiveness to antigen stimulation resulting from incomplete or insufficient signals delivered through the T-cell receptor (e.g., increase in intracellular Ca+2 in the absence of ras- activation). T cell anergy can also result upon stimulation with antigen in the absence of co- stimulation, resulting in the cell becoming refractory to subsequent activation by the antigen even in the context of co stimulation. The unresponsive state can often be overridden by the presence of lnterleukin-2. Anergic T-cells do not undergo clonal expansion and/or acquire effector functions.
The term "exhaustion" refers to T cell exhaustion as a state of T cell dysfunction that arises from sustained TCR signaling that occurs during many chronic infections and cancer. It is distinguished from anergy in that it arises not through incomplete or deficient signaling, but from sustained signaling. It is defined by poor effector function, sustained expression of inhibitory receptors and a transcriptional state distinct from that of functional effector or memory T cells. Exhaustion prevents optimal control of infection and tumors. Exhaustion can result from both extrinsic negative regulatory pathways (e.g., immunoregulatory cytokines) as well as cell intrinsic negative regulatory (co stimulatory) pathways.
"Enhancing T-cell function" means to induce, cause or stimulate a T-cell to have a sustained or amplified biological function, or renew or reactivate exhausted or dysfunctional T-cells. Examples of enhancing T-cell function include: increased secretion of y-interferon from CD4+ or CD8+ T-cells, increased proliferation, increased survival, increased differentiation, increased antigen responsiveness (e.g., viral, pathogen, or tumor clearance) relative to such levels before the intervention. In some embodiments, the level of enhancement is as least 50%, alternatively 60%, 70%, 80%, 90%, 100%, 120%, 150%, 200%. The manner of measuring this enhancement is known to one of ordinary skill in the art. As used herein, "metastasis" or “metastatic” is meant the spread of cancer from its primary site to other places in the body. Cancer cells can break away from a primary tumor, penetrate into lymphatic and blood vessels, circulate through the bloodstream, and grow in a distant focus (metastasize) in normal tissues elsewhere in the body. Metastasis can be local or distant. Metastasis is a sequential process, contingent on tumor cells breaking off from the primary tumor, traveling through the bloodstream, and stopping at a distant site. At the new site, the cells establish a blood supply and can grow to form a life-threatening mass. Both stimulatory and inhibitory molecular pathways within the tumor cell regulate this behavior, and interactions between the tumor cell and host cells in the distant site are also significant.
The term “cancer,” “cancerous,” or “malignant” refers to or describe the physiological condition in subjects that is typically characterized by unregulated cell growth. The term “cancer” includes but is not limited to a primary cancer that originates at a specific site in the body, a metastatic cancer that has spread from the place in which it started to other parts of the body, a recurrence from the original primary cancer after remission, and a second primary cancer that is a new primary cancer in a person with a history of previous cancer of a different type from the latter one.
The term “subject” or “a subject in need thereof” refers to a human or non-human subject including veterinary subjects such as, cows, sheep, cats, dogs, horses, primates, rabbits, and rodents (e.g., mice and rats) for which therapy is desired. In one preferred embodiment, the subject is a human and may be referred to as a patient or individual, e.g., a human suffering from, at risk of suffering from, or potentially capable of suffering from cancer. Those skilled in the medical art are readily able to identify individual patients who are afflicted with cancer.
The term “treat” or “treating” a cancer, as used herein means to administer a combination therapy according to the present disclosure to a subject having cancer, or diagnosed with cancer, to achieve at least one positive therapeutic effect, such as, for example, reduced number of cancer cells, reduced tumor size, reduced rate of cancer cell infiltration into peripheral tissues, or reduced rate of tumor metastases or tumor growth, reversing, stopping, controlling, slowing, interrupting, arresting, alleviating, and/or inhibiting the progression or severity of a sign, symptom, disorder, condition, or disease, but does not necessarily involve a total elimination of all disease-related signs, symptoms, conditions, or disorders. Within the meaning of the present disclosure, the term “treat” or “treating” also denotes, to arrest, delay the onset (i.e., the period prior to clinical manifestation of a disease or symptom of a disease) and/or reduce the risk of developing or worsening a symptom of a disease.
As used herein, “in combination with,” “combined administration,” or “coadministration” refers to the administration of one agent and in addition to at least one other agent, wherein said agents are part of the same or separate dosage formsand are administered via the same or different routes of administration and on the same or different administration schedule. In some embodiments, the co-administration of two or more agents can be useful for treating individuals suffering from cancer who have primary or acquired resistance to ongoing therapies. The combination therapy provided herein may be useful for improving the efficacy and/or reducing the side effects of cancer therapies for individuals who do respond to such therapies.
As used herein, the term “simultaneously,” "simultaneous administration,” "administered simultaneously,” “concurrently,” or "concurrent administration,” means that the agents are administered at the same point in time or immediately following one another, but that the agents can be administered in any order. For example, in the latter case, the two or more agents are administered at times sufficiently close that the results observed are indistinguishable from those achieved when the agents are administered at the same point in time. The term simultaneous includes the administration of each agent of the combination therapy of the disclosure in the same medicament.
The agents of the present disclosure can be administered completely separately or in the form of one or more separate compositions. For example, the agents may be given separately at different times during the course of therapy (in a chronologically staggered manner, especially a sequence-specific manner) in such time intervals that the combination therapy is effective in treating cancer.
As used herein, the term “sequential,” “sequentially,” “administered sequentially,” or “sequential administration” refers to the administration of each agent of the combination therapy of the invention, either alone or in a medicament, one after the other, wherein each agent can be administered in any order. Sequential administration may be particularly useful when the therapeutic agents in the combination therapy are in different dosage forms, for example, one agent is a tablet and another agent is a sterile liquid, and/or the agents are administered according to different dosing schedules, for example, one agent is administered daily, and the second agent is administered less frequently such as weekly. The term “advanced,” as used herein, as it relates to breast cancer, includes locally advanced (non-metastatic) disease and metastatic disease.
The term “pharmaceutical composition” refers to a preparation which is in such form as to permit the biological activity of the active ingredient to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered. Such formulations are sterile. “Pharmaceutically acceptable” carriers or excipients are those which can reasonably be administered to a subject to provide an effective dose of the active ingredient employed.
The combinations provided herein may be formulated by a variety of methods apparent to those of skill in the art of pharmaceutical formulation. The various release properties described above may be achieved in a variety of different ways. Suitable formulations include, for example, tablets, capsules, press coat formulations, and other easily administered formulations.
A "package insert" refers to instructions customarily included in commercial packages of medicaments that contain information about the indications customarily included in commercial packages of medicaments that contain information about the indications, usage, dosage, administration, contraindications, other medicaments to be combined with the packaged product, and/or warnings concerning the use of such medicaments, etc.
In reference to the treatment of cancer, a therapeutically effective amount refers to that amount of a drug, agent, component, composition, compound, substance, targeted agent, targeted therapeutic agent, therapeutic antibody, therapeutic agent, medicament or pharmaceutical composition which is effective to achieve one or more of the following results following the administration of one or more therapies: (1) reducing the size of the tumor, (2) reducing the number of cancer cells, (3) inhibiting (/.e., slowing to some extent, preferably stopping) cancer cell infiltration into peripheral organs, (4) inhibiting (/.e., slowing to some extent, preferably stopping) tumor metastasis, (5) inhibiting (i.e., slowing to some extent, preferably stopping) tumor growth or tumor invasiveness, (6) relieving (i.e., to some extent, preferably eliminating) one or more signs or symptoms associated with the cancer, (7) decreasing the dose of other medications required to treat the disease, (8) enhancing the effect of another medication, (9) delaying the progression of the disease, (10) improving or increasing the disease-free, relapse-free, progression-free, and/or overall survival, duration, or rate, (11) increasing the response rate, the durability of response, or number of patients who respond or are in remission, (12) decreasing the hospitalization rate, (13) decreasing the hospitalization lengths, (14) the size of the tumor is maintained and does not increase or increases by less than 10%, preferably less than 5%, preferably less than 4%, preferably less than 2%; (15) an increase in the number of patients in remission, (16) increasing the length or duration of remission, (17) decreasing the recurrence rate of cancer; (18) decreasing the time to recurrence of cancer, and (19) an amelioration of cancer related symptoms and/or quality of life.
An effective amount can be administered in one or more administrations. For purposes of this disclosure, an effective amount of drug, compound, and/or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly.
As is understood in the clinical context, an effective amount of a drug, compound, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition. Thus, an “effective amount” may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
A therapeutic amount may also refer to a dosage of a drug that has been approved for use by a regulatory agency. A "subtherapeutic amount," as used herein, refers to a dosage of a drug that is significantly lower than the approved dosage, such as 10%, 20%, 30%, 40%, or 50% lower than the approved dosage.
The terms “treatment regimen,” “dosing protocol” and “dosing regimen” are used interchangeably to refer to the dose and timing of administration of each therapeutic agent in a combination of the invention.
“Tumor” as it applies to a subject diagnosed with, or suspected of having, a cancer refers to a malignant or potentially malignant neoplasm or tissue mass of any size and includes primary tumors and secondary neoplasms. A solid tumor is an abnormal growth or mass of tissue that usually does not contain cysts or liquid areas. Examples of solid tumors are sarcomas, carcinomas, and lymphomas.
Determining a synergistic interaction between two or more agents, the optimum range for the effect and absolute dose ranges of each agent for the effect may be definitively measured by administration of the agents over different dose ranges, and/or dose ratios to subjects in need of treatment. However, the observation of synergy in in vitro models or in vivo models can be predictive of the effect in humans and other species and in vitro models or in vivo models exist, as described herein, to measure a synergistic effect. The results of such studies can also be used to predict effective dose and plasma concentration ratio ranges and the absolute doses and plasma concentrations required in humans and other species such as by the application of pharmacokinetic and / or pharmacodynamics methods.
The term "pharmaceutically acceptable salt," as used herein, refers to pharmaceutically acceptable organic or inorganic salts of a compound of the invention. Some embodiments also relate to the pharmaceutically acceptable acid addition salts of the compounds described herein. Suitable acid addition salts are formed from acids which form non-toxic salts. Non-limiting examples of suitable acid addition salts, i.e., salts containing pharmacologically acceptable anions, include, but are not limited to, the acetate, acid citrate, adipate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, bitartrate, borate, camsylate, citrate, cyclamate, edisylate, esylate, ethanesulfonate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobrom ide/brom ide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, methanesulfonate, methylsulphate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, pyroglutamate, saccharate, stearate, succinate, tannate, tartrate, p-toluenesulfonate, trifluoroacetate and xinofoate salts.
For a review on suitable salts, see Handbook of Pharmaceutical Salts: Properties, Selection, and Use by Stahl and Wermuth (Wiley-VCH, 2002). Methods for making pharmaceutically acceptable salts of compounds described herein are known to one of skill in the art.
"Carriers," as used herein, include pharmaceutically acceptable carriers, excipients, or stabilizers that are nontoxic to the cell or subject being exposed thereto at the dosages and concentrations employed. Often the physiologically acceptable carrier is an aqueous pH buffered solution. Examples of physiologically acceptable carriers include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid; low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfactants such as TWEEN™, polyethylene glycol (PEG), and PLURONICS™. The compounds described herein may also exist in unsolvated and solvated forms. Accordingly, some embodiments relate to the hydrates and solvates of the compounds described herein. The term “solvate,” as used herein, describes a molecular complex comprising a compound described herein and one or more pharmaceutically acceptable solvent molecules, for example, water (hydrate) and ethanol.
Compounds described herein containing one or more asymmetric carbon atoms can exist as two or more stereoisomers. Where structural isomers are interconvertible via a low energy barrier, tautomeric isomerism (‘tautomerism’) can occur. This can take the form of proton tautomerism in compounds described herein containing, for example, an imino, keto, or oxime group, or so-called valence tautomerism in compounds which contain an aromatic moiety. A single compound may exhibit more than one type of isomerism.
The compounds of the embodiments described herein include all stereoisomers (e.g., cis and trans isomers) and all optical isomers of compounds described herein (e.g., R and S enantiomers), as well as racemic, diastereomeric and other mixtures of such isomers. While all stereoisomers are encompassed within the scope of our claims, one skilled in the art will recognize that particular stereoisomers may be preferred.
Exemplary methods and materials are described herein, although methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the invention. The materials, methods, and examples are illustrative only and not intended to be limiting.
II. TLR7/8 Agonists
Toll-like receptors (TLRs) are a family of transmembrane proteins that recognize structurally conserved molecules that are derived from and unique to pathogens, referred to as pathogen-associated molecular patterns (PAMPS). As such, TLRs function in the mammalian immune system as front-line sensors of pathogen- associated molecular patterns, detecting the presence of invading pathogens.
The human genome contains 10 known TLRs, of these TLRS, TLR7, TLR8, and TLR9 recognize nucleic acids and their degradation products. The distribution of TLR3, TLR7, TLR8, and TLR9 is restricted to the endosomal compartments of cells and they are preferentially expressed in cells of the immune system. In the activated dimeric receptor configuration TLR7 and TLR8 recognize single strand RNA at one ligand binding site and the ribonucleoside degradation products guanosine and uridine, respectively, (as well as small molecule ligands with related structural motifs) at a second ligand binding site.
Some small-molecule TLR7 or TLR8 agonists have been identified. Those agonists can be grouped into purine-like molecules, such as 7-thia-8-oxoguanosine (TOG, isatoribine) or the imidazoquinoline-based compounds such as imiquimod. Imiquimod is so far the only approved TLR7 agonist, marketed as a 5% cream (Aldara). It generates approximately 80% 5-year clearance of superficial basal cell carcinomas, which is the most common cancer worldwide, thus demonstrating the importance of TLR7 agonists in cancer immunotherapy.
A TLR7/TLR8 (TLR7/8) small molecule agonist with dual bioactivity could provide further benefit over a more selective TLR7 agonist and would cause innate immune responses in a wider range of antigen presenting cells and other key immune cell types, including plasmacytoid and myeloid dendritic cells, monocytes, and B cells. Such potent dual TLR7/8 agonists may also be effective in stimulating effective anti-tumor responses in cancer.
Methods of preparing the TLR7/8 agonists that are required for the combination therapy purposes of the present disclosure have been disclosed in PCT Publication No. WO 2021/009676 A1 , which is hereby incorporated for all purposes.
Embodiments of the present invention comprise a TLR7/8 agonist of Formula I:
Figure imgf000021_0001
or a pharmaceutically acceptable salt thereof, wherein
R1 and R2 are independently Ci 3 alkyl; or
R1 and R2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
R3 is
Figure imgf000022_0001
R4 is Ci -6 alkyl, or (CH2)nO(CH2)mCH3, wherein said Ci -6 alkyl or any carbon of the (CH2)nO(CH2)mCH3 group is substituted with 0 to 3 F as valency allows;
R5 is Ci-3 alkyl, or OC1 -3 alkyl, wherein the C1-3 alkyl is substituted by 0 to 3 F;
R6 is H, or C1-3 alkyl, wherein the C1-3 alkyl is substituted with 0 to 3 F; m is 0 to 2; and n is 1 to 3.
In some preferred embodiments, the TLR7/8 agonist is of Formula I, wherein:
R1 and R2 are independently Ci 2 alkyl; or
R1 and R2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
Figure imgf000022_0002
R4 is C3-5 alkyl, or (CH2)nO(CH2)mCH3;
R5 is C1-2 alkyl;
R6 is H; m is 1 ; and n is 1 .
In some preferred embodiments of the methods as described herein, the TLR7/8 agonist may be selected from:
2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)methyl)- 2-m ethylpropane- 1 ,3-diol,
2-((4-amino-2-(ethoxymethyl)-1 H-imidazo[4,5-c]qu inolin- 1 -yl)methyl)-2- methylpropane-1 ,3-diol,
2-((4-amino-2-(ethoxymethyl)-7,8-dihydrocyclopenta[b]imidazo[4,5-d]pyridin-
1 (6H)-yl)methyl)-2-methylpropane-1 ,3-diol, 2-((4-amino-2-(ethoxymethyl)-6,7,8,9-tetrahydro-1 H-imidazo[4,5-c]quinolin-1 - yl)methyl)-2-methylpropane-1 ,3-diol,
3-(4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)-2-
(methoxymethyl)-2-methylpropan-1 -ol,
(R)-3-(4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1-yl)-2-
(methoxymethyl)-2-methylpropan-1 -ol,
(S)-3-(4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)-2- (methoxymethyl)-2-methylpropan-1 -ol,
2-((4-amino-6,7-dimethyl-2-(2,2,2-trifluoroethyl)-1 H-imidazo[4,5-c]pyridin- 1 - yl)methyl)-2-methylpropane-1 ,3-diol,
2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)methyl)- 2-ethylpropane-1 ,3-diol,
2-((4-amino-2-butyl-1 H-imidazo[4,5-c]quinolin-1-yl)methyl)-2-methylpropane-1 ,3- diol,
2-((4-amino-2-butyl-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1-yl)methyl)-2- methylpropane-1 ,3-diol, and
2-((4-amino-2-pentyl-1 H-imidazo[4,5-c]quinolin-1 -yl)methyl)-2-methylpropane- 1 ,3-diol; or any pharmaceutically acceptable salt thereof.
In some preferred embodiments of the methods as described herein, the preferred TLR7/8 agonist is 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5- c]pyridin-1-yl)methyl)-2-methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof. The preferred TLR7/8 agonist is also referred as PF- 07225570 in this disclosure (or PF-5570 as referred in FIG.1-FIG.3) and has the following structure A:
Figure imgf000023_0001
In some preferred embodiments, the TLR7/8 agonist, especially the preferred PF- 07225570 or a pharmaceutically acceptable salt thereof, may be administered with an interval of at least 2 days, at least 3 days, at least 4 days, at least 5 days, at least 6 days, at least 7 days, at least 10 days, at least 14 days, or at least 21 days. In some aspects, the dose interval for the TLR7/8 agonist, especially the preferred PF- 07225570 or a pharmaceutically acceptable salt thereof, may be about 2-21 days, 2-14 days, 2-10 days, 2-7 days, 3-21 days, 3-14 days, 3-10 days, 3-7 days, 5-21 days, 5-14 days, 5-10 days, or 5-7 days. In one aspect, the dose interval for the TLR7/8 agonist, especially the preferred PF- 07225570 or a pharmaceutically acceptable salt thereof, is at least about 7 days (7 ± 2 days), at least about 14 days (14 ± 2 days), or at least about 21 days (21 ± 2 days).
III. PD-1 Axis Binding Antagonists
Embodiments of the present disclosure comprise a PD-1 axis binding antagonist. As used herein, the term “PD-1 axis binding antagonist” or “PD-1 axis antagonist” refers to a molecule that inhibits the interaction of a PD-1 axis binding partner (e.g., PD-1 , PD-L1 , PD-L2) with either one or more of its binding partners, for example so as to overcome or partially overcome T-cell dysfunction resulting from signaling on the PD-1 signaling axis — with a result being to restore, partially restore or enhance T-cell function (e.g., proliferation, cytokine production, target cell killing, survival). As used herein, a PD-1 axis binding antagonist includes one or more of (i) a PD-1 antagonist, (ii) a PD-L1 antagonist, and/or (iii) a PD-L2 antagonist. i) PD-1 antagonist
The term “PD-1 antagonist,” as used herein, refers to a molecule that decreases, blocks, inhibits, abrogates or interferes with signal transduction resulting from the interaction of PD-1 with one or more of its binding partners, such as PD-L1 , PD-L2. In some embodiments, the PD-1 antagonist is a molecule that inhibits the binding of PD-1 to its binding partners. In a specific aspect, the PD-1 antagonist inhibits the binding of PD-1 to PD-L1 and/or PD-L2. In specific embodiments, the PD-1 antagonist inhibits the binding of PD-1 to PD-LI. In another embodiment, the PD-1 antagonist inhibits the binding of PD-1 to PD-L2. In a further embodiment, the PD-1 antagonist inhibits the binding of PD-1 to both PD-L1 and PD-L2. For example, PD-1 antagonists include anti- PD-1 antibodies, antigen binding fragments thereof, immunoadhesins, fusion proteins, and other molecules that decrease, block, inhibit, abrogate or interfere with signal transduction resulting from the interaction of PD-1 with PD-L1 and/or PD-L2. In some embodiments, a PD-1 antagonist reduces the negative co-stimulatory signal mediated by or through cell surface proteins expressed on T lymphocytes mediated signaling through PD-1 so as to render a dysfunctional T-cell less non-dysfunctional. In some embodiments, the PD-1 antagonist is an anti-PD-1 antibody (aPD-1 ). In some embodiments, the PD-1 antagonist blocks binding of PD-L1 expressed on a cancer cell to PD-1 expressed on an immune cell (T cell, B cell or NKT cell) and preferably also blocks binding of PD-L2 expressed on a cancer cell to the immune-cell expressed PD-1. Alternative names or synonyms for PD-1 and its ligands include: PDCD1 , PD1 , CD279 and SLEB2 for PD-1 ; PDCD1 L1 , PDL1 , B7H1 , B7-4, CD274 and B7-H for PD-L1 ; and PDCD1 L2, PDL2, B7-DC, Btdc and CD273 for PD-L2. In any of the treatment method, medicaments and uses of the present invention in which a human individual is being treated, the PD-1 antagonist may block binding of human PD- L1 to human PD-1 , and block binding of both human PD-L1 and PD-L2 to human PD-1.
PD-1 antagonists useful in any of the treatment methods, medicaments and uses of the present disclosure include a monoclonal antibody (mAb), or antigen binding fragment thereof, which specifically binds to PD-1 or PD-L1 , and preferably specifically binds to human PD-1 or human PD-L1. The mAb may be a human antibody, a humanized antibody or a chimeric antibody, and may include a human constant region. In some embodiments, the human constant region is selected from the group consisting of lgG1 , lgG2, lgG3 and lgG4 constant regions, and in some embodiments, the human constant region is an IgG 1 or lgG4 constant region. In some embodiments, the antigen binding fragment is selected from the group consisting of Fab, Fab'-SH, F(ab')2, scFv and Fv fragments.
Examples of mAbs that bind to human PD-1 , and useful in the treatment method, medicaments and uses of the present disclosure, are described in U.S. Patent Nos. 7,488,802, 7,521 ,051 , 8,008,449, 8,354,509, 8,168,757, PCT Patent Publication Nos. WO 2004/004771 , WO 2004/072286, WO 2004/056875, and US Patent Publication No. 2011/0271358. Specific anti-human PD-1 mAbs useful as the PD-1 antagonist in the treatment method, medicaments and uses of the present disclosure include: sasanlimab (RN888, PF-06801591 , Pfizer Inc.), nivolumab (OPDIVO®, ONO-4538, BMS-936558, MDX1106, Bristol-Myers Squibb Company), pembrolizumab (KEYTRUDA®, MK-3475, lambrolizumab, Merck & Co., Inc.), pidilizumab (CT-011), cemiplimab (LIBTAYO®, REGN2810, Regeneron Pharmaceuticals, Inc.), tislelizumab (BGB-A317, BeiGene Ltd./Celgene Corporation), spartalizumab (PDR001 , Novartis AG), sintilimab (IBI308, Innovent Biologies, Inc.), MEDI-0680 (AMP-514, AstraZeneca PLC, CAS No.: 1642374-69-3, PCT Publication No. WO2014204856A1), BGB-108 (CAS No.: 1859072-61-9, PCT Publication No. W02016007235A1 ), or AGEN2034 (Agenus Inc. CAS No.: 2230167-06-1 , PCT Publication No. WO2018106864A1 ), genolimzumab (CBT Pharmaceuticals, CAS No. 2135810-45-4, PCT Publication No. WO2017176965 A1 ), CBT-502 (CBT Pharmaceuticals, CAS No.: 2303884-58-2, PCT Publication No. WO2019176965 A1), camrelizumab (SHR-1210, Incyte Corporation), or any combination thereof.
In certain embodiments, the PD-1 antagonist is an anti-PD-1 antibody.
In some embodiments, the anti-PD-1 antibody is nivolumab. In some embodiments, the anti-PD-1 antibody is pembrolizumab. In some embodiments, the anti-PD-1 antibody is sasanlimab. Sasanlimab is a humanized, immunoglobulin G4 (lgG4) monoclonal antibody (mAb) that binds to the PD-1 receptor. By blocking its interaction with PD-L1 and PD-L2, PD-1 pathway-mediated inhibition of the immune response is released, leading to an anti-tumor immune response. Clinical anti-tumor activity with sasanlimab has been seen in a panel of anti-PD1 sensitive solid tumor types including non-small cell lung cancer and urothelial carcinoma. Sasanlimab is described, for example, in US Patent No. US 10,155,037, which is hereby incorporated for all purposes.
Additional exemplary PD-1 antagonists include those described in U.S. Patent Application Publication 20130280265, U.S. Patent Application Publication 20130237580, U.S. Patent Application Publication 20130230514, U.S. Patent Application Publication 20130109843, U.S. Patent Application Publication 20130108651 , U.S. Patent Application Publication 20130017199, U.S. Patent Application Publication 20120251537, U.S. Patent Application Publication 20110271358, European Patent EP2170959B1 , in PCT Publication No. WO 2011/066342, PCT Publication No. WO 2015/035606, PCT Publication No. WO
2015/085847, PCT Publication No. WO 2015/112800, PCT Publication No. WO
2015/112900, PCT Publication No. WO 2016/092419, PCT Publication No. WO
2017/017623, PCT Publication No. WO 2017/024465, PCT Publication No. WO
2017/054646, PCT Publication No. WO 2017/071625, PCT Publication No. WO
2017/019846, PCT Publication No. WO 2017/132827, PCT Publication No. WO
2017/214092, PCT Publication No. WO 2018/013017, PCT Publication No. WO
2018/053106, PCT Publication No. WO 2018/055503, PCT Publication No. WO
2018/053709, PCT Publication No. WO 2018/068336, and PCT Publication No. WO
2018/072743, the entire disclosures of which are incorporated herein by reference.
Other exemplary PD-1 antagonists are described in Curran et al., PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors, PNAS, 2010, 107, 4275; Topalian etal., Safety, activity, and immune correlates of anti-PD-1 antibody in cancer, New Engl. J. Med. 2012, 366, 2443; Brahmer et al., Safety and activity of anti-PD-L1 antibody in patients with advanced cancer, New Engl. J. Med. 2012, 366, 2455; Dolan et al., PD-1 pathway inhibitors: changing the landscape of cancer immunotherapy, Cancer Control 2014, 21 , 3; and Sunshine et al., Pd-1/Pd-L1 Inhibitors, Curr. Opin. in Pharmacol. 2015, 23. ii) PD-L1 antagonist
The term “PD-L1 antagonist,” as used herein, refers to a molecule that decreases, blocks, inhibits, abrogates or interferes with signal transduction resulting from the interaction of PD-L1 with either one or more of its binding partners, such as PD-1 , B7-1. In some embodiments, the PD-1 axis antagonist comprises a PD-L1 antagonist. In some embodiments, the PD-L1 antagonist inhibits the binding of PD-L1 to its binding partners. In a specific aspect, the PD-L1 antagonist inhibits the binding of PD-L1 to PD-1 . In another specific aspect, the PD-L1 antagonist inhibits the binding of PD-L1 to PD-1 and/or B7-1. In another specific aspect, the PD-L1 antagonist inhibits the binding of PD-L1 to both PD-1 and B7-1.
In some embodiments, the PD-L1 antagonists include anti-PD-L1 antibodies, antigen binding fragments thereof, immunoadhesins, fusion proteins, and other molecules that decrease, block, inhibit, abrogate or interfere with signal transduction resulting from the interaction of PD-L1 with one or more of its binding partners, such as PD-1 , and/or B7-1. In some embodiments, a PD-L1 antagonist reduces the negative costimulatory signal mediated by or through cell surface proteins expressed on T lymphocytes mediated signaling through PD-L1 so as render a dysfunctional T-cell less non-dysfunctional. In some embodiments, a PD-L1 antagonist is an anti-PD-L1 antibody (OPD-L1).
In some embodiments, the PD-1 antagonist is an anti-PD-1 antibody.
In some embodiments, the anti-PD-1 antibody useful for this disclosure include BMS-936559 (Bristol-Myers Squibb Company, CAS No. 1422185-22-5, PCT Publication No. WO2013019906 A1), KN035 (envafolimab, Jiangsu Alphamab biopharmaceuticals Co., Ltd., CAS No. 2102192-68-5, Zhang F, Wei H, Wang X, Bai Y, Wang P, Wu J, et al. Structural basis of a novel PD-L1 nanobody for immune checkpoint blockade. Cell Discov. 2017;3:1700), atezolizumab (TECENTRIQ®, MPDL3280A, Roche Holding AG), durvalumab (IMFINZI®, AstraZeneca PLC), and/or avelumab (BAVENCIO®, Merck & Co., Inc. and Pfizer, Inc.).
Additional exemplary PD-L1 antagonists include those described in U.S. Patent Application Publication 20090055944, U.S. Patent Application Publication 20100203056, U.S. Patent Application Publication 20120039906, U.S. Patent Application Publication 20130045202, U.S. Patent Application Publication
20130309250, U.S. Patent Application Publication US20130034559, U.S. Patent
Application Publication US20150282460, U.S. Patent Application Publication
20160108123, PCT Publication No. WO 2011/066389, PCT Publication No. WO
2016/000619, PCT Publication No. WO 2016/094273, PCT Publication No. WO
2016/061142, PCT Publication No. WO 2016/149201 , PCT Publication No. WO
2016/149350, PCT Publication No. WO 2016/179576, PCT Publication No. WO
2017/020801 , PCT Publication No. WO 2017/103147, PCT Publication No. WO
2017/112741 , PCT Publication No. WO 2017/205213, PCT Publication No. WO
2017/054646, PCT Publication No. WO 2017/084495, PCT Publication No. WO
2017/161976, PCT Publication No. WO 2018/005682, PCT Publication No. WO
2018/053106, PCT Publication No. WO 2018/085469, PCT Publication No. WO
2018/111890, and PCT Publication No. WO 2018/106529, the entire disclosures of which are incorporated herein by reference. Other exemplary PD-L1 binding antagonists are described in Sunshine etal., 2015. iii) PD-L2 antagonist
The term “PD-L2 antagonist,” as used herein, refers to a molecule that decreases, blocks, inhibits, abrogates or interferes with signal transduction resulting from the interaction of PD-L2 with either one or more of its binding partners, such as PD-1. In some embodiments, a PD-L2 antagonist is a molecule that inhibits the binding of PD-L2 to its binding partners. In a specific aspect, the PD-L2 antagonist inhibits binding of PD-L2 to PD-1. In some embodiments, the PD-L2 antagonists include anti- PD-L2 antibodies, antigen binding fragments thereof, immunoadhesins, fusion proteins, and other molecules that decrease, block, inhibit, abrogate or interfere with signal transduction resulting from the interaction of PD-L2 with either one or more of its binding partners, such as PD-1. In some embodiments, a PD-L2 antagonist reduces the negative co-stimulatory signal mediated by or through cell surface proteins expressed on T lymphocytes mediated signaling through PD-L2 so as render a dysfunctional T-cell less non-dysfunctional. In some embodiments, a PD-L2 antagonist is a PD-L2 immunoadhesin.
IV. METHODS, USES AND MEDICAMENTS
General Methods Standard methods in molecular biology are described in Sambrook, Fritsch and Maniatis (1982 & 1989 2nd Edition, 2001 3rd Edition) Molecular Cloning, A Laboratory Manual; Sambrook and Russell Molecular Cloning, 3rd ed., 2001 ; Wu, Recombinant DNA, Vol. 217. Standard methods also appear in Ausbel, et al., Current Protocols in Molecular Biology, Vols.1-4, 2001 , which describes cloning in bacterial cells and DNA mutagenesis (Vol. 1), cloning in mammalian cells and yeast (Vol. 2), glycoconjugates and protein expression (Vol. 3), and bioinformatics (Vol. 4).
Methods for protein purification including immunoprecipitation, chromatography, electrophoresis, centrifugation, and crystallization are described (Coligan, et al., Current Protocols in Protein Science, Vol. 1 , 2000, John Wiley and Sons, Inc., New York). Chemical analysis, chemical modification, post-translational modification, production of fusion proteins, and glycosylation of proteins are described (e.g., Coligan, et al., Current Protocols in Protein Science, Vol. 2, 2000; Ausubel, et al., Current Protocols in Molecular Biology, Vol. 3, 2001 , pp. 16.0.5-16.22.17; Sigma-Aldrich, Co. Products for Life Science Research, 2001 , pp. 45-89; Amersham Pharmacia Biotech (2001) BioDirectory, pp. 384-391). Production, purification, and fragmentation of polyclonal and monoclonal antibodies are described (Coligan, etal., Current Protocols in Immunology, Vol. 1 , 2001 ; Harlow and Lane, Using Antibodies, 1999). Standard techniques for characterizing ligand/receptor interactions are available (e.g., Coligan, etal., Current Protocols in Immunology, Vol. 4, 2001).
Monoclonal, polyclonal, and humanized antibodies can be prepared (e.g., Sheperd and Dean (eds.) Monoclonal Antibodies, 2000; Kontermann and Dubel (eds.) Antibody Engineering, 2001 ; Harlow and Lane, Antibodies A Laboratory Manual, 1988, pp. 139-243; Carpenter, et al., Non-Fc receptor-binding humanized anti-CD3 antibodies induce apoptosis of activated human T cells, J. Immunol. 2000, 165:6205; He, et al., Humanization and pharmacokinetics of a monoclonal antibody with specificity for both E-and P-selectin,J. Immunol. 1998, 160:1029; Tang et al., Use of a peptide mimotope to guide the humanization of MRK-16, an anti-P-glycoprotein monoclonal antibody , J. Biol. Chem. 1999, 274:27371-27378; Baca et al., Antibody humanization using monovalent phage display, J. Biol. Chem. 1997, 272:10678-10684; Chothia et al., Conformations of immunoglobulin hypervariable regions, Nature 1989, 342:877-883; Foote and Winter Antibody framework residues affecting the conformation of the hypervariable loops, J. Mol. Biol. 1992, 224:487-499; U.S. Patent No. 6,329,511 ).
An alternative to humanization is to use human antibody libraries displayed on phage or human antibody libraries in transgenic mice (Vaughan et al., Human antibodies with sub-nanomolar affinities isolated from a large non-immunized phage display library, Nature Biotechnol. 1996, 14:309-314; Barbas , Synthetic human antibodies, Nature Medicine 1995, 1 :837-839; Mendez et al., Functional transplant of megabase human immunoglobulin loci recapitulates human antibody response in mice, Nature Genetics 1997, 15:146-156; Hoogenboom and Chames, Natural and designer binding sites made by phage display technology, Immunol. Today 2000, 21 :371-377; Barbas etal., Phage Display: A Laboratory Manual, 2001 ; Kay et al., Phage Display of Peptides and Proteins: A Laboratory Manual, 1996; de Bruin et al., Selection of high- affinity phage antibodies from phage display libraries, Nature Biotechnol. 1999, 17:397- 399).
Purification of antigen is not necessary for the generation of antibodies. Animals can be immunized with cells bearing the antigen of interest. Splenocytes can then be isolated from the immunized animals, and the splenocytes can be fused with a myeloma cell line to produce a hybridoma (e.g., Meyaard, L., et. al., LAIR-1 , a novel inhibitory receptor expressed on human mononuclear leukocytes, Immunity 1997, 7:283-290; Wright etal., Inhibition of chicken adipocyte differentiation by in vitro exposure to monoclonal antibodies against embryonic chicken adipocyte plasma membranes, Immunity 2000, 13:233-242; Preston, et al., The leukocyte/neuron cell surface antigen OX2 binds to a ligand on macrophages,) Ear. J. Immunol. 1997, 27:1911 -1918, Kaithamana et al., Induction of experimental autoimmune Graves' disease in BALB/c mice, J. Immunol. 1999, 163:5157-5164).
Antibodies can be conjugated, e.g., to small drug molecules, enzymes, liposomes, polyethylene glycol (PEG). Antibodies are useful for therapeutic, diagnostic, kit or other purposes, and include antibodies coupled, e.g., to dyes, radioisotopes, enzymes, or metals, e.g., colloidal gold (e.g., Le Doussal et al., Enhanced in vivo targeting of an asymmetric bivalent hapten to double-antigen-positive mouse B cells with monoclonal antibody conjugate cocktails, J. Immunol. 1991 , 146:169-175; Gibellini et al., Extracellular HIV-1 Tat protein induces the rapid Ser133 phosphorylation and activation of CREB transcription factor in both Jurkat lymphoblastoid T cells and primary peripheral blood mononuclear cells, J. Immunol. 1998160:3891-3898; Hsing and Bishop, Requirement for nuclear factor-KB activation by a distinct subset of CD40- mediated effector functions in B lymphocytes, J. Immunol. 1999, 162:2804-2811 ; Everts et al., Selective intracellular delivery of dexamethasone into activated endothelial cells using an E-selectin-directed immunoconjugate, J. Immunol. 2002, 168:883-889). Methods for flow cytometry, including fluorescence activated cell sorting (FACS), are available (e.g., Owens, etal., Flow Cytometry Principles for Clinical Laboratory Practice, 1994; Givan Flow Cytometry, 2nd ed.; 2001 ; Shapiro, Practical Flow Cytometry, 2003). Fluorescent reagents suitable for modifying nucleic acids, including nucleic acid primers and probes, polypeptides, and antibodies, for use, e.g., as diagnostic reagents, are available (Molecular Probes, Catalogue, 2003; Sigma-Aldrich, Catalogue, 2003.
Standard methods of histology of the immune system are described (e.g., Muller- Harmelink (ed.), Human Thymus: Histopathology and Pathology, 1986; Hiatt, etal., Color Atlas of Histology, 2000; Louis, et al., Basic Histology: Text and Atlas, 2002.
Software packages and databases for determining, e.g., antigenic fragments, leader sequences, protein folding, functional domains, glycosylation sites, and sequence alignments, are available (e.g., GenBank, Vector NTI® Suite (Informax, Inc, Bethesda, MD); GCG Wisconsin Package (Accelrys, Inc., San Diego, CA); DeCypher® (TimeLogic Corp., Crystal Bay, Nevada); Menne, etal., A comparison of signal sequence prediction methods using a test set of signal peptides, Bioinformatics 2000, 16: 741-742; Menne, K.M.L., et. al. A comparisonof signal sequence prediction methods using a test set of signal peptides, Bioinformatics 2000, 16, 741-742; Wren, et al., SIGNAL-sequence information and GeNomic AnaLy sis Comput. Methods Programs Biomed. 2002, 68:177-181 ; von Heijne, Patterns of amino acids near signal-sequence cleavage sites, Ear. J. Biochem. 1983, 133:17-21 ; von Heijne, A new method for predicting signal sequence cleavage sites, Nucleic Acids Res. 1986, 14:4683-4690).
Therapeutic Methods and Uses
The disclosure provides therapeutic methods and uses comprising administering to the subject the combinations as described herein, optionally in further combination with other therapeutic agents.
In one aspect, the disclosure provides a method for treating a cancer in a subject comprising administering to the subject a combination therapy of the disclosure. In one aspect, the disclosure provides a method for treating a cancer comprising administering to a subject in need thereof an amount of a PD-1 axis binding antagonist and an amount of a TLR7/8 agonist, wherein the amounts together are effective in treating cancer, and wherein the TLR7/8 agonist is a compound of Formula I, or a pharmaceutically acceptable salt thereof, disclosed herein. In some such embodiments, the subject is a human. In some embodiments, the method involves the use of an anti-PD-L1 antibody in combination with a TLR7/8 agonist described herein.
In some preferred embodiments, the method involves the use of an anti-PD-1 antibody selected from sasanlimab, nivolumab, pembrolizumab, pidilizumab, cemiplimab, tislelizumab, spartalizumab, sintilimab, MEDI-0680, BGB-108, AGEN2034 genolimzumab, CBT-502, camrelizumab, or any combination thereof, in combination with 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1-yl)methyl)-2- methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof.
In some more preferred embodiments, the method involves the use of sasanlimab, in combination with 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H- imidazo[4,5-c]pyridin-1 -yl)methyl)-2-methylpropane- 1 ,3-diol, or a pharmaceutically acceptable salt thereof.
In some embodiments, the treatment results in sustained response in the individual after cessation of the treatment. The methods of this disclosure may find use in treating conditions where enhanced immunogenicity is desired such as increasing tumor immunogenicity for the treatment of cancer. As such, a variety of cancers may be treated, or their progression may be delayed.
In some embodiments, the individual has cancer that is resistant (has been demonstrated to be resistant) to one or more PD-1 axis binding antagonists. In some embodiments, resistance to PD-1 axis binding antagonist includes recurrence of cancer or refractory cancer. Recurrence may refer to the reappearance of cancer, in the original site or a new site, after treatment. In some embodiments, resistance to PD-1 axis binding antagonist includes progression of the cancer during treatment with the PD-1 axis binding antagonist. In some embodiments, resistance to PD-1 axis binding antagonist includes cancer that does not response to treatment. The cancer may be resistant at the beginning of treatment or it may become resistant during treatment. In some embodiments, the cancer is at early stage or at late stage.
In certain embodiments, the subject has received one, two, three, four, five or more prior cancer treatments. In other embodiments, the subject is treatment- naive. In some embodiments, the subject has progressed on other cancer treatments. In certain embodiments, the prior cancer treatment comprised an immunotherapy. In other embodiments, the prior cancer treatment comprised a chemotherapy. In some embodiments, the tumor has reoccurred. In some embodiments, the tumor is metastatic. In other embodiments, the tumor is not metastatic. In some embodiments, the subject has received a prior therapy to treat the tumor and the tumor is relapsed or refractory. In some embodiments, the subject has received a prior immuno-oncology therapy to treat the tumor and the tumor is relapsed or refractory. In some embodiments, the subject has received more than one prior therapy to treat the tumor and the subject is relapsed or refractory.
In some embodiments of the methods as described herein, the cancer is a solid tumor.
In a further embodiment, the disclosure is related to a method for treating cancer, wherein the cancer may be brain cancer, head/neck cancer (including squamous cell carcinoma of the head and neck (SCCHN)), prostate cancer, bladder cancer (including non-muscle invasive bladder cancer (NMIBC), or urothelial carcinoma, also known as transitional cell carcinoma (TCC)), lung cancer (including squamous cell carcinoma, small cell lung cancer (SCLC), and non-small cell lung cancer (NSCLC)), breast cancer, ovarian cancer, bone cancer, colorectal cancer, kidney cancer, liver cancer (including hepatocellular carcinoma (HCC)), pancreatic cancer, esophageal cancer (including sauamous cell carcinoma (SCCGC)), gastric cancer, gastroesophageal junction cancer, thyroid cancer, cervical cancer, uterine cancer, and/or renal cancer. Preferably, the cancer is bladder cancer. In a more preferred aspect, the method of the present disclosure is for treating non-muscle invasive bladder cancer (NMIBC).
In some embodiments, the combination therapy of the disclosure comprises administration of a PD-1 axis binding antagonist in combination with a TLR7/8 agonist. In the methods provided herein, each of the PD-1 axis binding antagonist and the TLR7/8 agonist, may be administered in any suitable manner known in the art. In one embodiment, the PD-1 axis binding antagonist and the TLR7/8 agonist are administered simultaneously or sequentially in either order.
In some embodiments, the disclosure provides a combination comprising: (i) a PD-1 axis binding antagonist described herein; and (ii) a TLR7/8 agonist of Formula I, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer in a subject. In some embodiments, the PD-1 axis binding antagonist is a PD-1 antagonist. In some embodiments, the PD-1 axis binding antagonist is a PD-L1 antagonist.
In some embodiments, the disclosure provides a combination comprising: (i) an anti-PD-1 antibody; and (ii) 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5- c]pyridin-1-yl)methyl)-2-methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer in a subject. In one preferred embodiment, the disclosure provides a combination comprising: (i) sasanlimab; and (ii) 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5- c]pyridin-1-yl)methyl)-2-methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof. In one aspect, the combination is for use in the treatment of cancer in a subject.
V. Dosage Forms and Regimens
Administration of the compounds of the disclosure may be affected by any method that enables delivery of the compounds to the site of action. These methods include oral routes, intraduodenal routes, parenteral injection (including intravenous, subcutaneous (sc), intramuscular, intravesical delivery such as intravesical instillation, intravascular or infusion), topical, and rectal administration.
Those skilled in the art will be able to determine the appropriate amount, dose or dosage of each compound, as used in the combination of the present disclosure, to administer to a patient, taking into account variety of factors, including, though not limited to, the degree of advancement of the disease, age, weight, general health, gender, diet, the compound administered, the time and route of administration, the nature and advancement of cancer, requiring treatment, and other medications the individual is taking.
In some embodiments, the methods of administration of the agents and combinations herein may include oral, intravenous, intramuscular subcutaneous, topical, transdermal, intraperitoneal, by implantation, by inhalation, intrathecal, intraventricular, intravesical delivery such as intravesical instillation, or intranasal administration. In a preferred embodiment, the method of administration for the TLR7/8 agonist is by intravesical instillation.
Dosage regimens may be adjusted to provide the optimum desired response. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form, as used herein, refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the disclosure is dictated by and directly dependent on (a) the unique characteristics of the chemotherapeutic agent and the particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals.
Thus, the skilled artisan would appreciate, based upon the disclosure provided herein, that the dose and dosing regimen is adjusted in accordance with methods well- known in the therapeutic arts. That is, the maximum tolerable dose can be readily established, and the effective amount providing a detectable therapeutic benefit to a patient may also be determined, as can the temporal requirements for administering each agent to provide a detectable therapeutic benefit to the patient. Accordingly, while certain dose and administration regimens are exemplified herein, these examples in no way limit the dose and administration regimen that may be provided to a patient in practicing the present disclosure.
For combination therapies as described herein, the agents may be administered at their approved dosages. Treatment is continued as long as clinical benefit is observed or until unacceptable toxicity or disease progression occurs. Nevertheless, in certain embodiments, the combination therapies of the present disclosure may advantageously utilize lower dosages of the administered therapeutic agents, thus avoiding possible toxicities or complications associated with the various monotherapies. For example, the dosages of the agents administered are significantly lower than the approved dosage, e.g., a subtherapeutic dosage of the TLR7/8 agonist is administered in combination with a subtherapeutic dosage of a PD-1 axis binding antagonist. It will be appreciated by the skilled practitioner that when the agents of the disclosure are used as part of a combination therapy, a lower dosage of the agent may be desirable than when the agent alone is administered to a subject, a synergistic therapeutic effect may be achieved through the use of combination therapy which, in turn, permits use of a lower dose of the agent to achieve the desired therapeutic effect.
In one embodiment, the dosages may be lower and may also be applied less frequently, which may diminish the incidence or severity of side-effects. This is in accordance with the desires and requirements of the subjects to be treated.
The amount of the agent of the disclosure administered will be dependent on the subject being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician. An effective amount of the PD-1 axis binding antagonist and the TLR7/8 agonist may be administered for prevention or treatment of disease. The appropriate dosage of the TLR7/8 agonist, and PD-1 axis binding antagonist, may be determined based on the type of disease to be treated, the TLR7/8 agonist, the PD-1 axis binding antagonist, the severity and course of the disease, the clinical condition of the subject, the subject's clinical history and response to the treatment, and the discretion of the attending physician. In some embodiments, combination treatment with TLR7/8 agonist, and PD- 1 axis binding antagonist (e.g., anti- PD-1 antibody or anti-PD-L1 antibody), are synergistic, whereby an efficacious dose of the TLR7/8 agonist, PD-1 axis binding antagonist, in the combination is reduced relative to efficacious dose of the each of the TLR7/8 agonist, PD-1 axis binding antagonist, as a single agent.
In some preferred embodiments, the TLR7/8 agonist such as 2-((4-amino-2- (ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1-yl)methyl)-2-methylpropane- 1 ,3-diol, or a pharmaceutically acceptable salt thereof, can be administered with an interval of about at least 7 days (7 ± 2 days), at least 14 days (14± 2 days), or at least 21 days (21 ± 2 days). The efficacy observed (see Table 1 to Table 5, and FIG. 1 to FIG. 3) for a single-dose treatment or with a weekly dose interval for a small molecule compound is very surprising. Such weekly dose interval, or longer interval, may provide more convenience and less toxicity to a patient being treated.
VI. Kits
In some embodiments, the disclosure provides a kit comprising: (i) a pharmaceutical composition comprising a PD-1 axis binding antagonist described herein; and (ii) a pharmaceutical composition comprising a TLR7/8 agonist described herein and a pharmaceutically acceptable carrier.
In some embodiments, the kit further comprises package insert comprising instructions for using the TLR7/8 agonist in conjunction with PD-1 axis binding antagonist (e.g., anti-PD-1 or anti-PD-L1 antibody), treat or delay progression of cancer in a subject or to enhance immune function of a subject having cancer. In further embodiment, any of the PD-1 axis binding antagonists and the TLR7/8 agonists described herein may be included in the kits. In one preferred embodiment, the PD-1 binding antagonist is sasanlimab, and the TLR7/8 agonist is 2-((4-amino-2- (ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1-yl)methyl)-2-methylpropane- 1 ,3-diol, or a pharmaceutically acceptable salt thereof. In some embodiments, the PD-1 axis binding antagonist and the TLR7/8 agonist and are stored in the same container or in separate containers. Suitable containers include, for example, bottles, vials, bags and syringes. The container may be formed from a variety of materials such as glass, plastic (such as polyvinyl chloride or polyolefin), or metal alloy. In some embodiments, the container holds the formulation and the label on, or associated with, the container may indicate directions for use. The kit may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use. In some embodiments, the kit further includes one or more of another agent (e.g., a chemotherapeutic agent, and anti-neoplastic agent). Suitable containers for the one or more agent include, for example, bottles, vials, bags and syringes.
The specification is sufficient to enable one skilled in the art to practice the disclosure. Various modifications of the disclosure in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and fall within the scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference in their entirety for all purposes.
VII. Embodiments (EBs)
EB1 : A method for treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of a Programmed Death 1 protein (PD-1) axis binding antagonist in combination with a compound of Formula I:
Figure imgf000037_0001
or a pharmaceutically acceptable salt thereof, wherein
R1 and R2 are independently Ci -3 alkyl; or
R1 and R2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
R3 is
Figure imgf000038_0001
R4 is C1-6 alkyl, or (CH2)nO(CH2)mCH3, wherein said C1-6 alkyl or any carbon of the (CH2)nO(CH2)mCH3 group is substituted with 0 to 3 F as valency allows;
R5 is C1-3 alkyl, or OC1 -3 alkyl, wherein the C1-3 alkyl is substituted by 0 to 3 F;
R6 is H, or C1-3 alkyl, wherein the C1-3 alkyl is substituted with 0 to 3 F; m is 0 to 2; and n is 1 to 3.
EB2: The method of EB1 , wherein:
R1 and R2 are independently Ci 2 alkyl; or
R1 and R2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
Figure imgf000038_0002
R4 is C3-5 alkyl, or (CH2)nO(CH2)mCH3;
R5 is C1-2 alkyl;
R6 is H; m is 1 ; and n is 1 .
EB3: The method of EB1 , wherein the compound of Formula I is:
2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)methyl)-
2-m ethylpropane- 1 ,3-diol,
2-((4-amino-2-(ethoxymethyl)-1 H-imidazo[4,5-c]qu inolin- 1-yl)methyl)-2- methylpropane-1 ,3-diol,
2-((4-amino-2-(ethoxymethyl)-7,8-dihydrocyclopenta[b]imidazo[4,5-d]pyridin-
1 (6H)-yl)methyl)-2-methylpropane-1 ,3-diol,
2-((4-amino-2-(ethoxymethyl)-6,7,8,9-tetrahydro-1 H-imidazo[4,5-c]quinolin-1 - yl)methyl)-2-methylpropane-1 ,3-diol,
3-(4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)-2-
(methoxymethyl)-2-methylpropan-1 -ol, (R)-3-(4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1-yl)-2- (methoxymethyl)-2-methylpropan-1 -ol,
(S)-3-(4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)-2- (methoxymethyl)-2-methylpropan-1 -ol,
2-((4-amino-6,7-dimethyl-2-(2,2,2-trifluoroethyl)-1 H-imidazo[4,5-c]pyridin- 1 - yl)methyl)-2-methylpropane-1 ,3-diol,
2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)methyl)- 2-ethylpropane-1 ,3-diol,
2-((4-amino-2-butyl-1 H-imidazo[4,5-c]quinolin-1-yl)methyl)-2-methylpropane-1 ,3- diol,
2-((4-amino-2-butyl-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1-yl)methyl)-2- methylpropane-1 ,3-diol, and
2-((4-amino-2-pentyl-1 H-imidazo[4, 5-c]quinolin-1 -yl)methyl)-2-methylpropane- 1 ,3-diol; or a pharmaceutically acceptable salt thereof.
EB4: The method of any one of EB1 to EB3, wherein the compound of Formula I is 2- ((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1-yl)methyl)-2- methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof.
EB5: The method of any one of EB1 to EB4, wherein the PD-1 axis binding antagonist is an anti-PD-1 antibody selected from the group consisting of sasanlimab, nivolumab, pembrolizumab, pidilizumab, cemiplimab, tislelizumab, spartalizumab, sintilimab, MEDI- 0680, BGB-108, AGEN2034, genolimzumab, CBT-502, camrelizumab, and any combination thereof.
EB6: The method of EB5, wherein the anti-PD-1 antibody is sasanlimab.
EB7: The method of any one of EB1 to EB4, wherein the PD-1 axis binding antagonist is an anti-PD-L1 antibody selected from the group consisting of BMS-936559, KN035, atezolizumab, durvalumab, avelumab, and any combination thereof.
E8: The method of any one of EB1 to EB7, wherein the cancer is bladder cancer.
E9. The method of EB8, wherein the bladder cancer is non-muscle invasive bladder cancer (NMIBC).
E10: The method of EB9, wherein the NMIBC is Bacillus Calmette-Guerin (BCG) unresponsive NMIBC.
E11 : The method of any one of EB1 to EB10, wherein the PD-1 axis binding antagonist is administered by intravenous infusion or subcutaneous delivery, and the compound of Formula I, or a pharmaceutically acceptable salt thereof, is administered by intravesical delivery to bladder epithelium of the subject.
E12: The method of any one of EB1 to EB11 , wherein a therapeutically effective amount of BCG immunotherapy is further administered.
EB13: The method of any one of EB4 to EB12, wherein 2-((4-amino-2-(ethoxymethyl)- 6,7-dimethyl-1 H-imidazo[4,5-c]pyridin- 1 -yl)methyl)-2-methylpropane- 1 ,3-diol, or a pharmaceutically acceptable salt thereof, is administered with a dosing interval of about at least 7 days (7± 2 days).
EB14: A combination comprising a Programmed Death 1 protein (PD-1 ) axis binding antagonist and a compound of Formula I , or a pharmaceutically acceptable salt thereof, wherein the PD-1 axis binding antagonist comprises an anti-PD-1 antibody selected from the group consisting of sasanlimab, nivolumab, pembrolizumab, pidilizumab, cemiplimab, tislelizumab, spartalizumab, sintilimab, MEDI-0680, BGB-108, AGEN2034, genolimzumab, CBT-502, camrelizumab, and any combination thereof, or an anti-PD-L1 antibody selected from the group consisting of BMS-936559, KN035, atezolizumab, durvalumab, avelumab, and any combination thereof ; and wherein the compound of Formula I is:
Figure imgf000040_0001
or a pharmaceutically acceptable salt thereof, wherein
R1 and R2 are independently Ci -3 alkyl; or
R1 and R2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
Figure imgf000040_0002
R4 is Ci -6 alkyl, or (CH2)nO(CH2)mCH3, wherein said Ci -6 alkyl or any carbon of the (CH2)nO(CH2)mCH3 group is substituted with 0 to 3 F as valency allows;
R5 is C1 -3 alkyl, or OC1 -3 alkyl, wherein the C1 -3 alkyl is substituted by 0 to 3 F; R6 is H, or C1-3 alkyl, wherein the C1-3 alkyl is substituted with 0 to 3 F; m is 0 to 2; and n is 1 to 3.
EB15: The combination of EB14, wherein:
R1 and R2 are independently Ci 2 alkyl; or
R1 and R2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
R3 is
Figure imgf000041_0001
R4 is C3-5 alkyl, or (CH2)nO(CH2)mCH3;
R5 is C1-2 alkyl;
R6 is H; m is 1 ; and n is 1 .
EB16: The combination of EB14 or EB15, wherein the PD-1 axis binding antagonist is sasanlimab, and the compound of Formula I is 2-((4-amino-2-(ethoxymethyl)-6,7- dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)methyl)-2-methylpropane- 1 ,3-diol, or a pharmaceutically acceptable salt thereof.
EB17: The combination of any one of EB14 to EB16, for use in the treatment of cancer.
EB18: The combination for use in the treatment of cancer of EB 17, wherein said cancer is bladder cancer
EB19: The combination for use in the treatment of cancer of EB18, wherein the bladder cancer is NMIBC.
EB20: The combination for use in the treatment of cancer of EB19, wherein the NMIBC is BCG un-responsive NMIBC.
EB21 : The combination for use in the treatment of cancer of any one of EB18 to EB20, wherein 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1- yl)methyl)-2-methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof, is administered with a dosing interval of about at least 7 days).
EB22. The method of any of EB4 to EB13, wherein the compound 2-((4-amino-2- (ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1-yl)methyl)-2-methylpropane- 1 ,3-diol, or a pharmaceutically acceptable salt thereof, is administered with a dosing interval of at least about 14 days.
EB23. The method of any of EB1 to EB13, wherein the compound 2-((4-amino-2- (ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1-yl)methyl)-2-methylpropane- 1 ,3-diol, or a pharmaceutically acceptable salt thereof, is administered with a dosing interval of at least about 21 days.
EB24. The combination for use in the treatment of cancer of any one of EB18 to EB20, wherein the compound 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5- c]pyridin-1-yl)methyl)-2-methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof, is administered with a dosing interval of at least about 14 days.
EB25. The combination for use in the treatment of cancer of any one of EB18 to EB20, and EB24, wherein the compound 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H- imidazo[4,5-c]pyridin-1 -yl)methyl)-2-methylpropane- 1 ,3-diol, or a pharmaceutically acceptable salt thereof, is administered with a dosing interval of at least about 21 days.
VIII. Examples
In order that this invention may be better understood, the following examples are set forth. These examples are for purposes of illustration only and are not to be construed as limiting the scope of the invention in any manner.
Example 1 : In vivo efficacy induced by combinatorial therapy of PF-07225570 and anti-PD-1 in MB49 syngeneic tumor model
The purpose of this study was to evaluate the in vivo efficacy of the combinatorial therapy of PF-07225570 and murine surrogate anti-PD-1 (PF-06937004) in a syngeneic mouse bladder cancer model. MB49 cells were implanted subcutaneously and PF-07225570 was tested through intra-tumor (IT) delivery to mimic local administration while anti-PD-1 was dosed Intra-peritoneally (IP) for systemic exposure. This study addressed two main questions: 1) does combinatorial therapy of PF-07225570 and anti-PD-1 treatment provide significant benefit over single agent therapy in MB-49 tumor bearing mice, and 2) does reducing number of doses or increasing time between doses of PF-07225570 in combination with anti-PD-1 still provide significant benefit in MB-49 tumor bearing mice? Efficacy was assessed comparing combination therapy to control or single agent therapy.
Materials and Methods-Table A
Figure imgf000043_0001
RPMI: Roswell Park Memorial Institute; DMEM: Dulbecco’s Modified Eagle Medium; FBS: Fetal bovine serum; DPBS: Dulbecco’s Phosphate buffer solution; EDTA: Ethylenediaminetetraacetic. Compounds Tested-Table B:
Figure imgf000043_0002
*PF-06937004 is a chimeric mAb (human anti-PD-1 CDR) on mouse lgG1 (mlgG1) frame. It binds to mouse PD-1 with an affinity of about 685 nM. PF-06937004 is an anti-mouse PD-1 mlgG1 having the same heavy chain variable region (VH) and light chain variable region (VL) sequence of the PD1 -F2 clone described in PCT Publication No. WO 2004/056875 A1 , which is hereby incorporated for all purposes. No substantial mutation has been introduced into the mouse fragment crystallizable (Fc) regions.
Animals-Table C
Organ system evaluated: Measurement of subcutaneous tumor growth and mouse body weight
Species/Strain: Mus musculus/C57BI/6J mice
Doses: 14mg/kg of PF-07225570; 10mg/kg of Anti-PD-1
Gender and No. per group/replicates: Female 10 mice/group
Age: 7-8 weeks old
Source: The Jackson Laboratory, Sacramento, CA
Figure imgf000043_0003
Q1 Dx1 : Single dose; Q3Dx2: Dose every 3 days for a total of 2 doses; Q3Dx3: Dose every 3 days for atotal of 3 doses; QWx2: Dose every week foratotal of 2 doses.
Equipment Fowler calipers Ultra Cal VI, Fowler High Precision, Newton Massachusetts, order number: NC1677319, Model number: 54-100-444-1.
Ohaus balance SPX422, Ohaus Corporation, Parsippany, New Jersey, order number: 30253020, model number: (01-922-401).
Software
Studylog 4.0 was used to acquire mouse body weights and tumor volumes. GraphPad Prism 9.0.0 were used for analysis of mouse tumor volumes, body weights and survival. Statistics
The statistical significance of differences between experimental groups was assessed by ANCOVA using Pfizer-internal TGI analyzer software. Data is depicted as mean ±SEM. *p<0.05 **p <0.01 , ***p <0.001 , ****p <0.0001. Statistical significance in survival determination using Logrank Mantel-cox test compared to Vehicle + Isotype using GraphPad Prism *p <0.05 **p <0.01 , ***p<0.001 , ****p<0.0001. Fisher’s exact test for contingency to determine statistically significant differences between proportion of responders vs non- responders using GraphPad Prism.
Methods
MB-49 cells were cultured in DM EM supplemented with 10% FBS. The cells were expanded for SC implantation in mice. Cells were harvested by 5-minute digestion with 0.25% trypsin and resuspended in DPBS. Mice were anaesthetized with 3% vaporized Isoflurane. The lower right flank of 7 to 8-week old female C57BI6/J mice were shaved and implanted with 200,000 MB-49 cells using 27.5-gauge syringe. At day 7 post implantation, tumor volumes for individual mice were measured and assigned to groups by mean tumor volumes at n=10 per group. Length (L) and width (W) of tumors were recorded and tumor volume was calculated using the formula: Volume = 0.5*L*WA2. Mice were treated with isotype or anti-PD-1 (IP) or with vehicle or PF- 07225570 (IT). In brief, anti-PD-1 was dosed on a Q3Dx3 schedule, and PF-07225570 treatment was altered testing Q3Dx3, Q3Dx2, Q1 Dx1 , or QWx2 dose strategy. Body weight and tumor volume were measured twice a week throughout the study. TGI was calculated with the following formula: 1 -(Tend-Tbegin/Cend-Cbegin) where Tend is final average tumor volume of treated group, Tbegin is average starting volume of treated group, Cend is final average volume of control group, and Cbegin is average starting volume of control group. TGI was calculated on the first day an animal was lost in any group due to excessive tumor volume (for Vehicle + Isotype controls, TGI was assessed at day 20 and for single agent controls, TGI was assessed at day 24). In Vivo Efficacy Study Antitumor efficacy was evaluated following different dose regimen strategies of combination therapy with PF-07225570 and anti-PD-1 in the MB49 syngeneic mouse model. Anti-PD-1 treatment remained consistent with Q3Dx3 IP dosing and PF- 07225570 IT treatment was altered to explore how reducing frequency (Q3Dx3, Q3Dx2, Q1 Dx1) or increasing length of time (QWx2) between dosing would affect TGI, number of responders or survival compared to isotype + vehicle controls or single agent treatments.
TGI, calculated at day 20 post tumor cell implant, and p-values are summarized in Table 1 and FIG. 1. No significant body weight loss was observed in any group. Statistically significant TGI was measured in all treatment groups when compared to vehicle + isotype controls. While single agent PF-07225570 (TGI = 87%; p <0.0001 ) and anti-PD-1 (TGI = 75%; p=0.00379) resulted in significant anti-tumor efficacy, the greatest efficacy was achieved with combination therapy (>93% TGI for all combination conditions tested compared to vehicle + isotype controls (p <0.0001 ). PF-07225570 administered Q3Dx3 in the combination regimen also resulted in statistically significant TGI compared to anti-PD-1 single agent therapy (p = 0.0091) but did not reach significance compared to PF-07225570 single agent therapy (p = 0.1676) at day 20 post implant. Significant single agent efficacy of anti-PD-1 and PF-07225570 was observed in this model when using vehicle + isotype as the control using TGI calculated at day 20 post implant. Using single agents as the control group in the TGI formula, we were able to calculate TGI and compare combination groups to single agents at day 24 post implant. TGI and p-values are summarized in Table 2 (anti-PD-1 single agent as the control), Table 3 (PF-07225570 single agent as the control), and FIG. 2. PF- 07225570 Q3Dx3 combination therapy resulted in significant TGI compared to both anti-PD-1 and PF-07225570 single agent controls at day 24 post implant (TGI = 94.1%, p = 0.0382 and TGI = 93.84%, p = 0.0287, respectively).
Table 1 : In Vivo Efficacy Summary for Combinatorial Study of PF-07225570 and
Anti-PD-1 in MB-49 Syngeneic Model Compared to Vehicle + Isotype Control
Figure imgf000045_0001
Figure imgf000046_0001
p-values determined by ANCOVA and TGI were determined on day 20 with Vehicle + Isotype as control using Pfizer-internal TGI analyzer software. Number of animals per group = 10.
Table 2: In Vivo Efficacy Summary for Combinatorial Study of PF-07225570 and
Anti-PD-1 in MB-49 Syngeneic Model Compared to Single Agent Anti-PD-1 Control
Figure imgf000046_0002
p-values determined by ANCOVA and TGI were determined on day 24 with Vehicle + anti- PD-1 as control using Pfizer-internal TGI analyzer software. Number of animals per group = 10
Table 3: In Vivo Efficacy Summary for Combinatorial Study of PF-07225570 and
Anti-PD-1 in MB-49 Syngeneic Model Compared to Single Agent PF-07225570 Control
Figure imgf000046_0003
Responders were defined as having two consecutive tumor measurements that were lower than previous measurements after randomization and dosing began. The number of responders and p-values are summarized in Table 4. Single agent therapy did not produce a statistically significant number of responders compared to vehicle + isotype control group, whereas combination therapy induced statistically significant increased numbers of responders in all combination regimens tested compared to vehicle + isotype control (Q3Dx3, p = 0.0001 ; Q3Dx2, p = 0.0007; Q1 Dx1 , p = 0.0031 ; QWx2, p = 0.0031). PF-07225570 Q3Dx3 combination therapy also resulted in statistically significant increased number of responders compared to anti-PD-1 single agent therapy (p = 0.0055) and trended towards significance compared to PF- 07225570 single agent therapy (p = 0.0573). Table 4: Number of Responders in Combinatorial Study of PF-07225570 and
Anti-PD-1 in MB-49 Syngeneic Model
Figure imgf000047_0001
Survival and p-values are summarized in Table 5 and FIG. 3. Single agent therapy resulted in significant survival benefit compared to vehicle + isotype treated control (anti-PD-1 alone, p=0.006; PF-07225570 alone, p=0.0021 ) and all combination therapies tested resulted in significant survival benefit (p <0.0001 ) compared to vehicle + isotype treated controls. Additionally, combination group PF-07225570 dosed Q3Dx3 also showed significant survival benefit over single agent anti-PD-1 (p=0.0271 ) and single agent PF-07225570 (p=0.0319). PF-07225570 Q1 Dx1 combination therapy also resulted in statistically significant
TGI at day 20 compared to anti-PD-1 single agent therapy (p = 0.009, Table 1) and at day 24 compared to PF-07225570 single agent therapy (TGI = 67.07%, p = 0.046, Table 3). Q3Dx2 combination therapy resulted in statistically significant increased number of responders compared to anti-PD-1 single agent therapy (p = 0.023, Table 4). Lastly, Q1 Dx1 combination therapy showed significant survival benefit over single agent anti-PD-1 (p = 0.0361 , Table 5) and single agent PF-07225570 (p = 0.0392, Table 5) treatment.
Table 5: Survival Summary for Combinatorial Study of PF-07225570 and Anti-
PD-1 in MB-49 Syngeneic Model
Figure imgf000048_0001
group=10
In summary, single agent PF-07225570 and single agent anti-PD-1 provided statistically significant TGI and survival benefit compare to vehicle + isotype controls. While the number of responders was not significantly different comparing single agent therapy to vehicle + isotype controls, all combination therapies, regardless of PF- 07225570 dose regimen tested, resulted in an increased number of responders, significant TGI at day 20 post implant, and significant survival advantage compared to vehicle + isotype treated controls. The combination of anti-PD-1 and PF-07225570 dosed Q3Dx3 provided significant survival and TGI at day 24 post implant over anti-PD- 1 and PF-07225570 single agent therapy. In addition, combination of anti-PD-1 and PF- 07225570 dosed Q3Dx3 provided significant responder benefit over anti-PD-1 and near significant (p = 0.0573) responder benefit over PF-07225570 single agent controls. Reducing PF-07225570 dosing (Q3Dx2 or Q1 Dx1 ) in combination with anti-PD-1 did provided some efficacy over anti-PD-1 and single agent PF-07225570 therapy but did not reach significance with every metric. Taken together, this data supports the therapeutic benefit of PF-07225570 in combination with anti-PD-1 in the MB-49 bladder cancer model.
All references cited herein, including patent applications, patent publications cited in the specification are herein incorporated by reference in their entirety. Although the foregoing disclosure has been described in some detail by way of illustration and example, it will be readily apparent to those of ordinary skill in the art in light of the teachings of this disclosure that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims.
The foregoing description and Examples detail certain specific embodiments of the disclosure and describes the best mode contemplated by the inventors. It will be appreciated, however, that no matter how detailed the foregoing may appear in text, the disclosure may be practiced in many ways and the disclosure should be construed in accordance with the appended claims and any equivalents thereof.

Claims

49 CLAIMS What is claimed:
1. A method for treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of a Programmed Death 1 protein (PD-1 ) axis binding antagonist in combination with a compound of Formula I:
Figure imgf000050_0001
or a pharmaceutically acceptable salt thereof, wherein
R1 and R2 are independently Ci 3 alkyl; or
R1 and R2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
R3 is
Figure imgf000050_0002
R4 is Ci -6 alkyl, or (CH2)nO(CH2)mCH3, wherein said Ci -6 alkyl or any carbon of the (CH2)nO(CH2)mCH3 group is substituted with 0 to 3 F as valency allows;
R5 is Ci-3 alkyl, or OC1 -3 alkyl, wherein the C1-3 alkyl is substituted by 0 to 3 F;
R6 is H, or C1-3 alkyl, wherein the C1-3 alkyl is substituted with 0 to 3 F; m is 0 to 2; and n is 1 to 3.
2. The method of claim 1 , wherein:
R1 and R2 are independently Ci 2 alkyl; or
R1 and R2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
R3 is 50
Figure imgf000051_0001
R4 is C3-5 alkyl, or (CH2)nO(CH2)mCH3;
R5 is C1-2 alkyl;
R6 is H; m is 1 ; and n is 1 .
3. The method of claim 1 , wherein the compound of Formula I is:
2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)methyl)- 2-methylpropane-1 ,3-diol,
2-((4-amino-2-(ethoxymethyl)-1 H-imidazo[4,5-c]qu inolin- 1 -yl)methyl)-2- methylpropane-1 ,3-diol,
2-((4-amino-2-(ethoxymethyl)-7,8-dihydrocyclopenta[b]imidazo[4,5-d]pyridin-
1 (6H)-yl)methyl)-2-methylpropane-1 ,3-diol,
2-((4-amino-2-(ethoxymethyl)-6,7,8,9-tetrahydro-1 H-imidazo[4,5-c]quinolin-1 - yl)methyl)-2-methylpropane-1 ,3-diol,
3-(4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)-2-
(methoxymethyl)-2-methylpropan-1 -ol,
(R)-3-(4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin- 1-yl)-2-
(methoxymethyl)-2-methylpropan-1 -ol,
(S)-3-(4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)-2- (methoxymethyl)-2-methylpropan-1 -ol,
2-((4-amino-6,7-dimethyl-2-(2,2,2-trifluoroethyl)-1 H-imidazo[4,5-c]pyridin- 1 - yl)methyl)-2-methylpropane-1 ,3-diol,
2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)methyl)-
2-ethylpropane-1 ,3-diol,
2-((4-amino-2-butyl-1 H-imidazo[4,5-c]quinolin-1 -yl)methyl)-2-methylpropane-1 ,3- diol,
2-((4-amino-2-butyl-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)methyl)-2- methylpropane-1 ,3-diol, and
2-((4-amino-2-pentyl-1 H-imidazo[4, 5-c]quinolin-1 -yl)methyl)-2-methylpropane-
1 ,3-diol; or a pharmaceutically acceptable salt thereof. 51
4. The method of any one of claims 1 to 3, wherein the compound of Formula I is 2-((4- amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)methyl)-2- methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof.
5. The method of any one of claims 1 to 4, wherein the PD-1 axis binding antagonist is an anti-PD-1 antibody selected from the group consisting of sasanlimab, nivolumab, pembrolizumab, pidilizumab, cemiplimab, tislelizumab, spartalizumab, sintilimab, MEDI- 0680, BGB-108, AGEN2034, genolimzumab, CBT-502, camrelizumab, and any combination thereof.
6. The method of claim 5, wherein the anti-PD-1 antibody is sasanlimab.
7. The method of any one of claims 1 to 4, wherein the PD-1 axis binding antagonist is an anti-PD-L1 antibody selected from the group consisting of BMS-936559, KN035, atezolizumab, durvalumab, avelumab, and any combination thereof.
8. The method of any one of claims 1-7, wherein the cancer is bladder cancer.
9. The method of claim 8, wherein the bladder cancer is non-muscle invasive bladder cancer (NMIBC).
10. The method of claim 9, wherein the NMIBC is Bacillus Calmette-Guerin (BCG) unresponsive NMIBC.
11. The method of any one of claims 1-10, wherein the PD-1 axis binding antagonist is administered by intravenous infusion or subcutaneous delivery, and the compound of Formula I, or a pharmaceutically acceptable salt thereof, is administered by intravesical delivery to bladder epithelium of the subject.
12. The method of any one of claims 1 -11 , wherein a therapeutically effective amount of BCG immunotherapy is further administered.
13. The method of any one of claims 4-12, wherein the compound of Formula I is 2-((4- amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)methyl)-2- methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof, is administered with a dosing interval of at least about 7 days.
14. A combination comprising a Programmed Death 1 protein (PD-1) axis binding antagonist and a compound of Formula I , or a pharmaceutically acceptable salt thereof, wherein the PD-1 axis binding antagonist comprises an anti-PD-1 antibody selected from the group consisting of sasanlimab, nivolumab, pembrolizumab, pidilizumab, cemiplimab, tislelizumab, spartalizumab, sintilimab, MEDI-0680, BGB-108, AGEN2034, genolimzumab, CBT-502, camrelizumab, and any combination thereof, or an anti-PD-L1 antibody selected from the group consisting of BMS-936559, KN035, atezolizumab, durvalumab, avelumab, and any combination thereof; and wherein the compound of Formula I is:
Figure imgf000053_0001
or a pharmaceutically acceptable salt thereof, wherein
R1 and R2 are independently Ci 3 alkyl; or
R1 and R2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
R3 is
Figure imgf000053_0002
R4 is Ci -6 alkyl, or (CH2)nO(CH2)mCH3, wherein said Ci -6 alkyl or any carbon of the (CH2)nO(CH2)mCH3 group is substituted with 0 to 3 F as valency allows;
R5 is C1-3 alkyl, or OC1 -3 alkyl, wherein the C1-3 alkyl is substituted by 0 to 3 F;
R6 is H, or C1-3 alkyl, wherein the C1-3 alkyl is substituted with 0 to 3 F; m is 0 to 2; and n is 1 to 3.
15. The combination of claim 14, wherein: R1 and R2 are independently Ci 2 alkyl; or
R1 and R2 are joined to form a 5- to 7-membered carbocyclic ring, wherein said carbocyclic ring may be saturated or unsaturated;
R3 is
Figure imgf000054_0001
R4 is Cs s alkyl, or (CH2)nO(CH2)mCH3;
R5 is C1-2 alkyl;
R6 is H; m is 1 ; and n is 1 .
16. The combination of claim 14 or 15, wherein the PD-1 axis binding antagonist is sasanlimab, and the compound of Formula I is 2-((4-amino-2-(ethoxymethyl)-6,7- dimethyl-1 H-imidazo[4,5-c]pyridin-1 -yl)methyl)-2-methylpropane- 1 ,3-diol, or a pharmaceutically acceptable salt thereof.
17. The combination of any one of claims 14 to 16 for use in the treatment of cancer.
18. The combination for use in the treatment of cancer of claim 17, wherein said cancer is bladder cancer.
19. The combination for use in the treatment of cancer of claim 18, wherein the bladder cancer is NMIBC.
20. The combination for use in the treatment of cancer of claim 19, wherein the NMIBC is BCG un-responsive NMIBC.
21 . The combination for use in the treatment of cancer of any one of claims 18-20, wherein 2-((4-amino-2-(ethoxymethyl)-6,7-dimethyl-1 H-imidazo[4,5-c]pyridin-1 - yl)methyl)-2-methylpropane-1 ,3-diol, or a pharmaceutically acceptable salt thereof, is administered with a dosing interval of at least about 7 days.
PCT/IB2022/060461 2021-11-03 2022-10-31 Combination therapies using tlr7/8 agonist WO2023079428A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163263470P 2021-11-03 2021-11-03
US63/263,470 2021-11-03

Publications (1)

Publication Number Publication Date
WO2023079428A1 true WO2023079428A1 (en) 2023-05-11

Family

ID=84238095

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2022/060461 WO2023079428A1 (en) 2021-11-03 2022-10-31 Combination therapies using tlr7/8 agonist

Country Status (1)

Country Link
WO (1) WO2023079428A1 (en)

Citations (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5428130A (en) 1989-02-23 1995-06-27 Genentech, Inc. Hybrid immunoglobulins
US6329511B1 (en) 1998-12-01 2001-12-11 Protein Design Labs, Inc. Humanized antibodies to γ-interferon
WO2004004771A1 (en) 2002-07-03 2004-01-15 Ono Pharmaceutical Co., Ltd. Immunopotentiating compositions
WO2004056875A1 (en) 2002-12-23 2004-07-08 Wyeth Antibodies against pd-1 and uses therefor
WO2004072286A1 (en) 2003-01-23 2004-08-26 Ono Pharmaceutical Co., Ltd. Substance specific to human pd-1
US20090055944A1 (en) 2005-07-01 2009-02-26 Medarex, Inc. Human monoclonal antibodies to be programmed death ligand 1 (pd-l1)
US20100203056A1 (en) 2008-12-09 2010-08-12 Genentech, Inc. Anti-pd-l1 antibodies and their use to enhance t-cell function
WO2011066389A1 (en) 2009-11-24 2011-06-03 Medimmmune, Limited Targeted binding agents against b7-h1
WO2011066342A2 (en) 2009-11-24 2011-06-03 Amplimmune, Inc. Simultaneous inhibition of pd-l1/pd-l2
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
US20110271358A1 (en) 2008-09-26 2011-11-03 Dana-Farber Cancer Institute, Inc. Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor
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
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
US20120251537A1 (en) 2006-12-27 2012-10-04 Emory University Compositions and methods for the treatment of infections and tumors
US8354509B2 (en) 2007-06-18 2013-01-15 Msd Oss B.V. Antibodies to human programmed death receptor PD-1
WO2013019906A1 (en) 2011-08-01 2013-02-07 Genentech, Inc. Methods of treating cancer using pd-1 axis binding antagonists and mek inhibitors
US20130230514A1 (en) 2008-08-25 2013-09-05 Amplimmune, Inc. Compositions of pd-1 antagonists and methods of use
US20130237580A1 (en) 2012-03-07 2013-09-12 Aurigene Discovery Technologies Limited Peptidomimetic compounds as immunomodulators
US20130280265A1 (en) 2011-09-20 2013-10-24 Vical Incorporated Synergistic anti-tumor efficacy using alloantigen combination immunotherapy
US20130309250A1 (en) 2012-05-15 2013-11-21 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting pd-1/pd-l1 signaling
WO2014204856A1 (en) 2013-06-17 2014-12-24 Catabasis Pharmaceuticals, Inc. Fatty acid anticancer derivatives and their uses
WO2015035606A1 (en) 2013-09-13 2015-03-19 Beigene, Ltd. Anti-pd1 antibodies and their use as therapeutics and diagnostics
WO2015085847A1 (en) 2013-12-12 2015-06-18 上海恒瑞医药有限公司 Pd-1 antibody, antigen-binding fragment thereof, and medical application thereof
WO2015112800A1 (en) 2014-01-23 2015-07-30 Regeneron Pharmaceuticals, Inc. Human antibodies to pd-1
WO2015112900A1 (en) 2014-01-24 2015-07-30 Dana-Farber Cancer Institue, Inc. Antibody molecules to pd-1 and uses thereof
US20150282460A1 (en) 2014-04-04 2015-10-08 Crown Bioscience Inc. (Taicang) Animal model with human immune system
WO2016000619A1 (en) 2014-07-03 2016-01-07 Beigene, Ltd. Anti-pd-l1 antibodies and their use as therapeutics and diagnostics
WO2016007235A1 (en) 2014-07-11 2016-01-14 Genentech, Inc. Anti-pd-l1 antibodies and diagnostic uses thereof
WO2016061142A1 (en) 2014-10-14 2016-04-21 Novartis Ag Antibody molecules to pd-l1 and uses thereof
WO2016092419A1 (en) 2014-12-09 2016-06-16 Rinat Neuroscience Corp. Anti-pd-1 antibodies and methods of use thereof
WO2016094273A1 (en) 2014-12-08 2016-06-16 Dana-Farber Cancer Institute, Inc. Methods for upregulating immune responses using combinations of anti-rgmb and anti-pd-1 agents
WO2016149201A2 (en) 2015-03-13 2016-09-22 Cytomx Therapeutics, Inc. Anti-pdl1 antibodies, activatable anti-pdl1 antibodies, and methods of use thereof
WO2016149350A1 (en) 2015-03-17 2016-09-22 Mayo Foundation For Medical Education And Research Methods and materials for assessing and treating cancer
WO2016179576A1 (en) 2015-05-07 2016-11-10 Bioxcel Corporation Novel immunomodulatory therapeutic strategies targeting tumors in cancer
WO2017017623A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combined use of anti pd-1 and anti m-csf antibodies in the treatment of cancer
WO2017019846A1 (en) 2015-07-30 2017-02-02 Macrogenics, Inc. Pd-1-binding molecules and methods use thereof
WO2017020801A1 (en) 2015-07-31 2017-02-09 苏州康宁杰瑞生物科技有限公司 Single domain antibody for programmed death-ligand (pd-l1) and derived protein thereof
WO2017024465A1 (en) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
WO2017054646A1 (en) 2015-09-28 2017-04-06 江苏恒瑞医药股份有限公司 Stable anti-pd-1 antibody pharmaceutical preparation and application thereof in medicine
WO2017071625A1 (en) 2015-10-30 2017-05-04 中山康方生物医药有限公司 Anti-pd-1 monoclonal antibody, and pharmaceutical composition and use thereof
WO2017084495A1 (en) 2015-11-17 2017-05-26 江苏恒瑞医药股份有限公司 Pd-l1 antibody, antigen fragment binding thereof and pharmaceutical use thereof
WO2017103147A1 (en) 2015-12-18 2017-06-22 Institut Gustave Roussy Method for assessing the response to pd-1/pdl-1 targeting drugs
WO2017112741A1 (en) 2015-12-22 2017-06-29 Novartis Ag Mesothelin chimeric antigen receptor (car) and antibody against pd-l1 inhibitor for combined use in anticancer therapy
WO2017132827A1 (en) 2016-02-02 2017-08-10 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
WO2017161976A1 (en) 2016-03-23 2017-09-28 Mabspace Biosciences (Suzhou) Co., Ltd Novel anti-pd-l1 antibodies
WO2017176965A1 (en) 2016-04-07 2017-10-12 Chemocentryx, Inc. Reducing tumor burden by administering ccr1 antagonists in combination with pd-1 inhibitors or pd-l1 inhibitors
WO2017205213A1 (en) 2016-05-23 2017-11-30 Eli Lilly And Company Combination therapy of abemaciclib and immune checkpoint modulators for use in the treatment of cancer
WO2017214092A1 (en) 2016-06-07 2017-12-14 Macrogenics, Inc. Combination therapy
WO2018005682A2 (en) 2016-06-29 2018-01-04 Checkpoint Therapeutics, Inc. Pd-l1-specific antibodies and methods of using the same
WO2018013017A1 (en) 2016-07-13 2018-01-18 Закрытое Акционерное Общество "Биокад" Anti-pd-1 antibodies, method for producing same and method for using same
WO2018053106A1 (en) 2016-09-14 2018-03-22 Abbvie Biotherapeutics Inc. Anti-pd-1(cd279) antibodies
WO2018055503A1 (en) 2016-09-20 2018-03-29 Novartis Ag Combination comprising a pd-1 antagonist and an fgfr4 inhibitor
WO2018053709A1 (en) 2016-09-21 2018-03-29 Wuxi Biologics (Shanghai) Co., Ltd. The novel monoclonal antibodies to programmed death 1 (pd-1)
WO2018068336A1 (en) 2016-10-15 2018-04-19 Innovent Biologics (Suzhou) Co., Ltd Pd-1 antibodies
WO2018072743A1 (en) 2016-10-21 2018-04-26 苏州盛迪亚生物医药有限公司 Use of pd-1 antibody conjugated with ido inhibitor in preparing anti-tumor drug
WO2018085469A2 (en) 2016-11-01 2018-05-11 Anaptysbio, Inc. Antibodies directed against t cell immunoglobulin and mucin protein 3 (tim-3)
WO2018106529A1 (en) 2016-12-08 2018-06-14 Eli Lilly And Company Anti-tim-3 antibodies for combination with anti-pd-l1 antibodies
WO2018106864A1 (en) 2016-12-07 2018-06-14 Agenus Inc. Antibodies and methods of use thereof
WO2018111890A1 (en) 2016-12-12 2018-06-21 Genentech, Inc. Methods of treating cancer using anti-pd-l1 antibodies and antiandrogens
WO2019176965A1 (en) 2018-03-14 2019-09-19 Ricoh Company, Ltd. Composite for forming electrode, method of manufacturing electrode, and method of manufacturing nonaqueous electric storage element
WO2021009676A1 (en) 2019-07-17 2021-01-21 Pfizer Inc. Imidazo[4,5-c]pyridine derivatives as toll-like receptor agonsits

Patent Citations (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5428130A (en) 1989-02-23 1995-06-27 Genentech, Inc. Hybrid immunoglobulins
US6329511B1 (en) 1998-12-01 2001-12-11 Protein Design Labs, Inc. Humanized antibodies to γ-interferon
WO2004004771A1 (en) 2002-07-03 2004-01-15 Ono Pharmaceutical Co., Ltd. Immunopotentiating compositions
US7521051B2 (en) 2002-12-23 2009-04-21 Medimmune Limited Methods of upmodulating adaptive immune response using anti-PD-1 antibodies
US7488802B2 (en) 2002-12-23 2009-02-10 Wyeth Antibodies against PD-1
WO2004056875A1 (en) 2002-12-23 2004-07-08 Wyeth Antibodies against pd-1 and uses therefor
WO2004072286A1 (en) 2003-01-23 2004-08-26 Ono Pharmaceutical Co., Ltd. Substance specific to human pd-1
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
US20090055944A1 (en) 2005-07-01 2009-02-26 Medarex, Inc. Human monoclonal antibodies to be programmed death ligand 1 (pd-l1)
US20120251537A1 (en) 2006-12-27 2012-10-04 Emory University Compositions and methods for the treatment of infections and tumors
EP2170959B1 (en) 2007-06-18 2013-10-02 Merck Sharp & Dohme B.V. Antibodies to human programmed death receptor pd-1
US20130109843A1 (en) 2007-06-18 2013-05-02 Mds Oss B.V. Antibodies to human programmed death receptor pd-1
US20130108651A1 (en) 2007-06-18 2013-05-02 Msd Oss B.V. Antibodies to human programmed death receptor pd-1
US8354509B2 (en) 2007-06-18 2013-01-15 Msd Oss B.V. Antibodies to human programmed death receptor PD-1
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
US20130230514A1 (en) 2008-08-25 2013-09-05 Amplimmune, Inc. Compositions of pd-1 antagonists and methods of use
US20110271358A1 (en) 2008-09-26 2011-11-03 Dana-Farber Cancer Institute, Inc. Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor
US20100203056A1 (en) 2008-12-09 2010-08-12 Genentech, Inc. Anti-pd-l1 antibodies and their use to enhance t-cell function
US20130045202A1 (en) 2008-12-09 2013-02-21 Genentech, Inc. Anti-pd-l1 antibodies and articles of manufacture
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
US20130017199A1 (en) 2009-11-24 2013-01-17 AMPLIMMUNE ,Inc. a corporation Simultaneous inhibition of pd-l1/pd-l2
US20130034559A1 (en) 2009-11-24 2013-02-07 Medlmmune Limited Targeted Binding Agents Against B7-H1
WO2011066342A2 (en) 2009-11-24 2011-06-03 Amplimmune, Inc. Simultaneous inhibition of pd-l1/pd-l2
WO2011066389A1 (en) 2009-11-24 2011-06-03 Medimmmune, Limited Targeted binding agents against b7-h1
WO2013019906A1 (en) 2011-08-01 2013-02-07 Genentech, Inc. Methods of treating cancer using pd-1 axis binding antagonists and mek inhibitors
US20130280265A1 (en) 2011-09-20 2013-10-24 Vical Incorporated Synergistic anti-tumor efficacy using alloantigen combination immunotherapy
US20130237580A1 (en) 2012-03-07 2013-09-12 Aurigene Discovery Technologies Limited Peptidomimetic compounds as immunomodulators
US20130309250A1 (en) 2012-05-15 2013-11-21 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting pd-1/pd-l1 signaling
WO2014204856A1 (en) 2013-06-17 2014-12-24 Catabasis Pharmaceuticals, Inc. Fatty acid anticancer derivatives and their uses
WO2015035606A1 (en) 2013-09-13 2015-03-19 Beigene, Ltd. Anti-pd1 antibodies and their use as therapeutics and diagnostics
WO2015085847A1 (en) 2013-12-12 2015-06-18 上海恒瑞医药有限公司 Pd-1 antibody, antigen-binding fragment thereof, and medical application thereof
WO2015112800A1 (en) 2014-01-23 2015-07-30 Regeneron Pharmaceuticals, Inc. Human antibodies to pd-1
WO2015112900A1 (en) 2014-01-24 2015-07-30 Dana-Farber Cancer Institue, Inc. Antibody molecules to pd-1 and uses thereof
US20150282460A1 (en) 2014-04-04 2015-10-08 Crown Bioscience Inc. (Taicang) Animal model with human immune system
WO2016000619A1 (en) 2014-07-03 2016-01-07 Beigene, Ltd. Anti-pd-l1 antibodies and their use as therapeutics and diagnostics
WO2016007235A1 (en) 2014-07-11 2016-01-14 Genentech, Inc. Anti-pd-l1 antibodies and diagnostic uses thereof
WO2016061142A1 (en) 2014-10-14 2016-04-21 Novartis Ag Antibody molecules to pd-l1 and uses thereof
US20160108123A1 (en) 2014-10-14 2016-04-21 Novartis Ag Antibody molecules to pd-l1 and uses thereof
WO2016094273A1 (en) 2014-12-08 2016-06-16 Dana-Farber Cancer Institute, Inc. Methods for upregulating immune responses using combinations of anti-rgmb and anti-pd-1 agents
WO2016092419A1 (en) 2014-12-09 2016-06-16 Rinat Neuroscience Corp. Anti-pd-1 antibodies and methods of use thereof
US10155037B2 (en) 2014-12-09 2018-12-18 Rinat Neuroscience Corp. Anti-PD-1 antibodies and methods of use thereof
WO2016149201A2 (en) 2015-03-13 2016-09-22 Cytomx Therapeutics, Inc. Anti-pdl1 antibodies, activatable anti-pdl1 antibodies, and methods of use thereof
WO2016149350A1 (en) 2015-03-17 2016-09-22 Mayo Foundation For Medical Education And Research Methods and materials for assessing and treating cancer
WO2016179576A1 (en) 2015-05-07 2016-11-10 Bioxcel Corporation Novel immunomodulatory therapeutic strategies targeting tumors in cancer
WO2017017623A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combined use of anti pd-1 and anti m-csf antibodies in the treatment of cancer
WO2017019846A1 (en) 2015-07-30 2017-02-02 Macrogenics, Inc. Pd-1-binding molecules and methods use thereof
WO2017020801A1 (en) 2015-07-31 2017-02-09 苏州康宁杰瑞生物科技有限公司 Single domain antibody for programmed death-ligand (pd-l1) and derived protein thereof
WO2017024465A1 (en) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
WO2017054646A1 (en) 2015-09-28 2017-04-06 江苏恒瑞医药股份有限公司 Stable anti-pd-1 antibody pharmaceutical preparation and application thereof in medicine
WO2017071625A1 (en) 2015-10-30 2017-05-04 中山康方生物医药有限公司 Anti-pd-1 monoclonal antibody, and pharmaceutical composition and use thereof
WO2017084495A1 (en) 2015-11-17 2017-05-26 江苏恒瑞医药股份有限公司 Pd-l1 antibody, antigen fragment binding thereof and pharmaceutical use thereof
WO2017103147A1 (en) 2015-12-18 2017-06-22 Institut Gustave Roussy Method for assessing the response to pd-1/pdl-1 targeting drugs
WO2017112741A1 (en) 2015-12-22 2017-06-29 Novartis Ag Mesothelin chimeric antigen receptor (car) and antibody against pd-l1 inhibitor for combined use in anticancer therapy
WO2017132827A1 (en) 2016-02-02 2017-08-10 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
WO2017161976A1 (en) 2016-03-23 2017-09-28 Mabspace Biosciences (Suzhou) Co., Ltd Novel anti-pd-l1 antibodies
WO2017176965A1 (en) 2016-04-07 2017-10-12 Chemocentryx, Inc. Reducing tumor burden by administering ccr1 antagonists in combination with pd-1 inhibitors or pd-l1 inhibitors
WO2017205213A1 (en) 2016-05-23 2017-11-30 Eli Lilly And Company Combination therapy of abemaciclib and immune checkpoint modulators for use in the treatment of cancer
WO2017214092A1 (en) 2016-06-07 2017-12-14 Macrogenics, Inc. Combination therapy
WO2018005682A2 (en) 2016-06-29 2018-01-04 Checkpoint Therapeutics, Inc. Pd-l1-specific antibodies and methods of using the same
WO2018013017A1 (en) 2016-07-13 2018-01-18 Закрытое Акционерное Общество "Биокад" Anti-pd-1 antibodies, method for producing same and method for using same
WO2018053106A1 (en) 2016-09-14 2018-03-22 Abbvie Biotherapeutics Inc. Anti-pd-1(cd279) antibodies
WO2018055503A1 (en) 2016-09-20 2018-03-29 Novartis Ag Combination comprising a pd-1 antagonist and an fgfr4 inhibitor
WO2018053709A1 (en) 2016-09-21 2018-03-29 Wuxi Biologics (Shanghai) Co., Ltd. The novel monoclonal antibodies to programmed death 1 (pd-1)
WO2018068336A1 (en) 2016-10-15 2018-04-19 Innovent Biologics (Suzhou) Co., Ltd Pd-1 antibodies
WO2018072743A1 (en) 2016-10-21 2018-04-26 苏州盛迪亚生物医药有限公司 Use of pd-1 antibody conjugated with ido inhibitor in preparing anti-tumor drug
WO2018085469A2 (en) 2016-11-01 2018-05-11 Anaptysbio, Inc. Antibodies directed against t cell immunoglobulin and mucin protein 3 (tim-3)
WO2018106864A1 (en) 2016-12-07 2018-06-14 Agenus Inc. Antibodies and methods of use thereof
WO2018106529A1 (en) 2016-12-08 2018-06-14 Eli Lilly And Company Anti-tim-3 antibodies for combination with anti-pd-l1 antibodies
WO2018111890A1 (en) 2016-12-12 2018-06-21 Genentech, Inc. Methods of treating cancer using anti-pd-l1 antibodies and antiandrogens
WO2019176965A1 (en) 2018-03-14 2019-09-19 Ricoh Company, Ltd. Composite for forming electrode, method of manufacturing electrode, and method of manufacturing nonaqueous electric storage element
WO2021009676A1 (en) 2019-07-17 2021-01-21 Pfizer Inc. Imidazo[4,5-c]pyridine derivatives as toll-like receptor agonsits

Non-Patent Citations (55)

* Cited by examiner, † Cited by third party
Title
"Foote and Winter Antibody framework residues affecting the conformation of the hypervariable loops", J. MOL. BIOL., vol. 224, 1992, pages 487 - 499
"Givan Flow Cytometry", 2001
AMERSHAM: "Pharmacia Biotech", BIODIRECTORY, 2001, pages 384 - 391
ANONYMOUS: "PF-07225570 Alone or in Combination With an Anti-PD-1 Antibody in Recurrent Non-muscle Invasive Bladder Cancer (NMIBC)", 24 October 2022 (2022-10-24), XP093014821, Retrieved from the Internet <URL:https://clinicaltrials.gov/ct2/show/record/NCT05259397> [retrieved on 20230117] *
ASHISHM KAMAT ET AL: "Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of bladder carcinoma", JOURNAL FOR IMMUNOTHERAPY OF CANCER, BIOMED CENTRAL LTD, LONDON, UK, vol. 5, no. 1, 15 August 2017 (2017-08-15), pages 1 - 16, XP021247832, DOI: 10.1186/S40425-017-0271-0 *
AUSUBEL ET AL., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, vol. 3, 2001
BACA ET AL.: "Antibody humanization using monovalent phage display", J. BIOL. CHEM., vol. 272, 1997, pages 10678 - 10684, XP002308888, DOI: 10.1074/jbc.272.16.10678
BARBAS ET AL., PHAGE DISPLAY: A LABORATORY MANUAL, 2001
BARBAS: "Synthetic human antibodies", NATURE MEDICINE, vol. 1, 1995, pages 837 - 839, XP001194649, DOI: 10.1038/nm0895-837
BRAHMER ET AL.: "Safety and activity of anti-PD-L1 antibody in patients with advanced cancer", NEW ENGL. J. MED., vol. 366, 2012, pages 2455, XP002685330, DOI: 10.1056/NEJMoa1200694
CARPENTER ET AL.: "Non-Fc receptor-binding humanized anti-CD3 antibodies induce apoptosis of activated human T cells", J. IMMUNOL., vol. 165, 2000, pages 6205, XP002429073
CAS , no. 2102192-68-5
CAS, no. 1859072-61-9
CHOTHIA ET AL.: "Conformations of immunoglobulin hypervariable regions", NATURE, vol. 342, 1989, pages 877 - 883, XP002030586, DOI: 10.1038/342877a0
CLACKSON ET AL.: "Making antibody fragments using phage display libraries", NATURE, vol. 352, 1991, pages 624 - 628, XP002101159, DOI: 10.1038/352624a0
COLIGAN ET AL., CURRENT PROTOCOLS IN IMMUNOLOGY, vol. 4, 2001
COLIGAN ET AL., CURRENT PROTOCOLS IN PROTEIN SCIENCE, vol. 2, 2000
CONSTANTINIDOU ANASTASIA ET AL: "Targeting Programmed Cell Death -1 (PD-1) and Ligand (PD-L1): A new era in cancer active immunotherapy", PHARMACOLOGY & THERAPEUTICS, ELSEVIER, GB, vol. 194, 28 September 2018 (2018-09-28), pages 84 - 106, XP085583247, ISSN: 0163-7258, DOI: 10.1016/J.PHARMTHERA.2018.09.008 *
CURRAN ET AL.: "PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors", PNAS, vol. 107, 2010, pages 4275, XP055611670, DOI: 10.1073/pnas.0915174107
DE BRUIN ET AL.: "Selection of high-affinity phage antibodies from phage display libraries", NATURE BIOTECHNOL, vol. 17, 1999, pages 397 - 399, XP002988308, DOI: 10.1038/7959
DOLAN ET AL.: "PD-1 pathway inhibitors: changing the landscape of cancer immunotherapy", CANCER CONTROL, vol. 21, 2014, pages 3, XP055428249
EVERTS ET AL.: "Selective intracellular delivery of dexamethasone into activated endothelial cells using an E-selectin-directed immunoconjugate", J. IMMUNOL., vol. 168, 2002, pages 883 - 889
GIBELLINI ET AL.: "Extracellular HIV-1 Tat protein induces the rapid Ser133 phosphorylation and activation of CREB transcription factor in both Jurkat lymphoblastoid T cells and primary peripheral blood mononuclear cells", J. IMMUNOL., vol. 1998160, pages 3891 - 3898
HARLOWLANE, ANTIBODIES A LABORATORY MANUAL, 1988, pages 139 - 243
HARLOWLANE, USING ANTIBODIES, 1999
HE ET AL.: "Humanization and pharmacokinetics of a monoclonal antibody with specificity for both E-and P-selectin", J. IMMUNOL., vol. 160, 1998, pages 1029, XP002959848
HOOGENBOOMCHAMES: "Natural and designer binding sites made by phage display technology", IMMUNOL. TODAY, vol. 21, 2000, pages 371 - 377
HSINGBISHOP: "Requirement for nuclear factor- B activation by a distinct subset of CD40-mediated effector functions in B lymphocytes", J. IMMUNOL., vol. 162, 1999, pages 2804 - 2811
JOHNSON MELISSA L. ET AL: "Assessment of Subcutaneous vs Intravenous Administration of Anti-PD-1 Antibody PF-06801591 in Patients With Advanced Solid Tumors : A Phase 1 Dose-Escalation Trial", JAMA ONCOLOGY, vol. 5, no. 7, 1 July 2019 (2019-07-01), US, pages 999, XP055905849, ISSN: 2374-2437, Retrieved from the Internet <URL:http://dx.doi.org/10.1001/jamaoncol.2019.0836> DOI: 10.1001/jamaoncol.2019.0836 *
KAITHAMANA ET AL.: "Induction of experimental autoimmune Graves' disease in BALB/c mice", J. IMMUNOL., vol. 163, 1999, pages 5157 - 5164
KAY ET AL., PHAGE DISPLAY OF PEPTIDES AND PROTEINS: A LABORATORY MANUAL, 1996
KOHLER ET AL.: "Continuous cultures of fused cells secreting antibody of predefined specificity", NATURE, vol. 256, 1975, pages 495, XP037052082, DOI: 10.1038/256495a0
LE DOUSSAL ET AL.: "Enhanced in vivo targeting of an asymmetric bivalent hapten to double-antigen-positive mouse B cells with monoclonal antibody conjugate cocktails", J. IMMUNOL., vol. 146, 1991, pages 169 - 175
LOUIS ET AL., BASIC HISTOLOGY: TEXT AND ATLAS, 2002
MARKS ET AL.: "By-passing immunization: human antibodies from V-gene libraries displayed on phage", J. MOL. BIOL., vol. 222, 1991, pages 581 - 597, XP024010124, DOI: 10.1016/0022-2836(91)90498-U
MENDEZ ET AL.: "Functional transplant of megabase human immunoglobulin loci recapitulates human antibody response in mice", NATURE GENETICS, vol. 15, 1997, pages 146 - 156, XP002067603, DOI: 10.1038/ng0297-146
MENNE ET AL.: "A comparison of signal sequence prediction methods using a test set of signal peptides", BIOINFORMATICS, vol. 16, 2000, pages 741 - 742
MENNE,K.M.L.: "A comparisonof signal sequence prediction methods using a test set of signal peptides", BIOINFORMATICS, vol. 16, 2000, pages 741 - 742
MEYAARD, L.: "LAIR-1, a novel inhibitory receptor expressed on human mononuclear leukocytes", IMMUNITY, vol. 7, 1997, pages 283 - 290, XP002061091, DOI: 10.1016/S1074-7613(00)80530-0
MOLECULAR PROBES, CATALOGUE, 2003
OWENS ET AL., FLOW CYTOMETRY PRINCIPLES FOR CLINICAL LABORATORY PRACTICE, 1994, pages 71
PRESTA: "Selection, design, and engineering of therapeutic antibodies", J. ALLERGY CLIN. IMMUNOL., vol. 116, 2005, pages 731, XP005094459, DOI: 10.1016/j.jaci.2005.08.003
PRESTON ET AL.: "The leukocyte/neuron cell surface antigen OX2 binds to a ligand on macrophages", EUR. J. IMMUNOL., vol. 27, 1997, pages 1911 - 1918, XP002094135, DOI: 10.1002/eji.1830270814
SHAPIRO, PRACTICAL FLOW CYTOMETRY, 2003
SIGMA-ALDRICH, CATALOGUE, 2003
SIGMA-ALDRICH, CO. PRODUCTS FOR LIFE SCIENCE RESEARCH, 2001, pages 45 - 89
SUNSHINE ET AL.: "Pd-1 /Pd-L1 Inhibitors", CURR. OPIN. IN PHARMACOL., 2015, pages 23
TANG ET AL.: "Use of a peptide mimotope to guide the humanization of MRK-16, an anti-P-glycoprotein monoclonal antibody", J. BIOL. CHEM., vol. 274, 1999, pages 27371 - 27378
TOPALIAN ET AL.: "Safety, activity, and immune correlates of anti-PD-1 antibody in cancer", NEW ENGL. J. MED., vol. 366, 2012, pages 2443, XP055098235, DOI: 10.1056/NEJMoa1200690
VAUGHAN ET AL.: "Human antibodies with sub-nanomolar affinities isolated from a large non-immunized phage display library", NATURE BIOTECHNOL, vol. 14, 1996, pages 309 - 314, XP000196144, DOI: 10.1038/nbt0396-309
VON HEIJNE: "A new method for predicting signal sequence cleavage sites", NUCLEIC ACIDS RES, vol. 14, 1986, pages 4683 - 4690
VON HEIJNE: "Patterns of amino acids near signal-sequence cleavage sites", EUR. J. BIOCHEM., vol. 133, 1983, pages 17 - 21, XP008043344, DOI: 10.1111/j.1432-1033.1983.tb07424.x
WREN ET AL.: "SIGNAL-sequence information and GeNomic", ANALYSISCOMPUT. METHODS PROGRAMS BIOMED., vol. 68, 2002, pages 177 - 181
WRIGHT ET AL.: "Inhibition of chicken adipocyte differentiation by in vitro exposure to monoclonal antibodies against embryonic chicken adipocyte plasma membranes", IMMUNITY, vol. 13, 2000, pages 233 - 242
ZHANG FWEI HWANG XBAI YWANG PWU J ET AL.: "Structural basis of a novel PD-L1 nanobody for immune checkpoint blockade", CELL DISCOV, vol. 3, 2017, pages 1700, XP055490308, DOI: 10.1038/celldisc.2017.4

Similar Documents

Publication Publication Date Title
JP6894952B2 (en) Combination of PD-1 antagonist and VEGFR inhibitor for cancer treatment
US11312772B2 (en) Combinations of PD-1 antagonists and benzo [b] thiophene STING agonists for cancer treatment
RU2766890C2 (en) Combined methods for treating with pd-l1 antagonists
JP6637439B2 (en) Anti-OX40 antibody and method of use
KR20170122809A (en) A combination of a PD-1 antagonist and a VEGFR / FGFR / RET tyrosine kinase inhibitor to treat cancer
JP2018521019A (en) Method of treating cancer using anti-OX40 antibody
US20170306050A1 (en) Compositions and methods for treating cancer, inflammatory diseases and autoimmune diseases
JP2020023499A (en) Treatment of c1013g/cxcr4-associated waldenstrom&#39;s macroglobulinemia with anti-cxcr4 antibody
KR20170122810A (en) A combination of PD-1 antagonist and eribulin for treating cancer
AU2017210224A1 (en) Treatment of cancer with combinations of immunoregulatory agents
US20150079089A1 (en) Emp2 regulates angiogenesis in cancer cells through induction of vegf
US20210054063A1 (en) Combination therapy with anti-il-8 antibodies and anti-pd-1 antibodies for treating cancer
US20220241412A1 (en) Combination therapies using cdk inhibitors
TW202137984A (en) Combination of a pd-1 antagonist, a vegfr/fgfr/ret tyrosine kinase inhibitor and a cbp/beta-catenin inhibitor for treating cancer
AU2016317378A1 (en) Agent for enhancing immunity to cancer by using Allergin-1 antagonist
WO2023079428A1 (en) Combination therapies using tlr7/8 agonist
US20230250182A1 (en) Methods for treating cancer or von-hippel lindau disease using a combination of a pd-1 antagonist, a hif-2 alpha inhibitor, and lenvatinib or a pharmaceutically acceptable salt thereof
EP4213846A1 (en) Combination therapy of a pd-1 antagonist and lag3 antagonist and lenvatinib or a pharmaceutically acceptable salt thereof for treating patients with cancer
CN110603447A (en) Compositions and methods for treating cancer with anti-renalase antibodies and anti-PD 1 antibodies
JP2023549835A (en) Materials and methods for treating cancer
JP2022533833A (en) Combination therapy using CDK inhibitors

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: 22802270

Country of ref document: EP

Kind code of ref document: A1