WO2024102859A1 - Inhibiteurs oncologiques immunitaires de mek et utilisations thérapeutiques associées - Google Patents

Inhibiteurs oncologiques immunitaires de mek et utilisations thérapeutiques associées Download PDF

Info

Publication number
WO2024102859A1
WO2024102859A1 PCT/US2023/079156 US2023079156W WO2024102859A1 WO 2024102859 A1 WO2024102859 A1 WO 2024102859A1 US 2023079156 W US2023079156 W US 2023079156W WO 2024102859 A1 WO2024102859 A1 WO 2024102859A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
compound
pharmaceutically acceptable
acceptable salt
formula
Prior art date
Application number
PCT/US2023/079156
Other languages
English (en)
Inventor
Brett Matthew HALL
Bart Lieven DECORTE
Peter John King
Ruben Leenders
Anita Wegert
Kevin Fowler
Sarah KOLITZ
Robin DOODEMAN
Jarno POELAKKER
Rutger Henk Adriaan Folmer
Original Assignee
Immuneering Corporation
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
Priority claimed from PCT/US2023/060181 external-priority patent/WO2023133472A1/fr
Application filed by Immuneering Corporation filed Critical Immuneering Corporation
Publication of WO2024102859A1 publication Critical patent/WO2024102859A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • 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
    • A61K31/397Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having four-membered rings, e.g. azetidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • the present invention relates to MEK inhibitors, MEK immune oncology inhibitors, techniques for designing and synthesizing such MEK inhibitors, MEK immune oncology compositions comprising MEK inhibitors, MEK immune oncology inhibitors, and methods of treating disease comprising administering MEK inhibitors.
  • MEK inhibitors In healthy cells, the Mitogen Activated Protein Kinase (MAPK) pathway utilizes parallel signaling streams to decipher complex extracellular stimuli and drive cellular programs that promote proliferation, differentiation, survival, motility, apoptosis, and stress response.
  • MAPK Mitogen Activated Protein Kinase
  • the RAS-RAF-MEK-ERK cascade is one of three distinct MAPK pathways and is the one most often exploited in cancer.
  • Gain of function mutations in RAS (KRAS, NRAS, HRAS) or RAF (ARAF, BRAF, CRAF/RAF1) are common in cancers.
  • RAS mutations alone represent up to 95% of pancreatic cancer (KRAS), 20-30% of melanoma (NRAS), 40% of non- small cell lung cancer (KRAS) and 45% of colorectal cancer (KRAS).
  • KRAS pancreatic cancer
  • NRAS melanoma
  • KRAS non- small cell lung cancer
  • KRAS colorectal cancer
  • MEK1 and MEK2 are closely related dual-specificity kinases that are activated by upstream mediators including RAF (ARAF, BRAF, RAF1[also known as CRAF]), KSR (KSR1, KSR2) and RAS (KRAS, NRAS, HRAS).
  • pMEK When activated by phosphorylation on two serine residues, pMEK in turn facilitates phosphorylation of ERK1 and ERK2 (pERK), which leads to regulation of multiple downstream targets. Inappropriate activation of this pathway is associated with multiple oncogenic cell processes including proliferation, survival, growth, tumor metabolism, migration and immune evasion. Multiple targeted agents have been, and continue to be, developed with the goal of reducing MAPK pathway activity at each level from RAS to ERK, and clinical proof-of-concept has been achieved for several drugs in this area, including those that target KRAS G12C , BRAF V600E/K and MEK.
  • Second generation MEK inhibitors resist pathway reactivation by engaging the allosteric pocket in MEK in a unique way that prevents RAF activation of MEK itself but still have long half-lives, leading to chronic pathway ablation.
  • This continuous disruption of this core biologic pathway creates at least three well-documented challenges for first and second generation MEK inhibitors: (1) tolerability: clinically limiting, class effect safety issues (Heinzerling 2019), (2) acquired/adaptive resistance: selective pressure for escape mutations (Corcoran 2011), and (3) clinical utility: reduced drug-drug combination potential due to limitations on drug-related safety and toxicity.
  • first generation MEK inhibitors suffer from multiple shortcomings: (1) sustained on-target occupancy drives acquired and/or adaptive resistance and dose-limiting toxicities, (2) mechanistic drug-target interaction fails to effectively control pathway reactivation (e.g., CRAF-bypass) and (3) limited clinical utility for drug combinations due to high baseline drug-related toxicity.
  • a common feature of nearly all MEK inhibitors is their allosteric target engagement, which is highly selective for MEK and non-ATP competitive, commonly referred to as a Type-III allosteric inhibitor.
  • First generation MEK inhibitors exemplified by trametinib, cobimetinib, binimetinib and selumetinib, sustainably suppress MAPK pathway activity through chronic occupancy of MEK1 and MEK2. Yet, they display dose-limiting class effect toxicities and are sensitive to pathway reactivation events. While second generation MEK inhibitors, exemplified by VS-6766 (CH5126766, that has a mean terminal half-life of 53.6-hours, Guo 2022), display mechanistic resistance to pathway reactivation, they also sustainably suppress MAPK pathway activity through chronic occupancy of MEK1 and MEK2 and, as such, share similar class effect toxicities as first-generation inhibitors.
  • VS-6766 CH5126766, that has a mean terminal half-life of 53.6-hours, Guo 2022
  • Table 1 is a summary of certain characteristics of MEK inhibitors for RAS mutant disease treatment.
  • Table 1 Characteristics of MEK Inhibitors by Generation in RAS Mutant Disease MEKi Type MoA Pathway Pathway Class-effect tor compounds, such as MEK inhibitor compounds with shorter half-life, such compounds having a shorter half-life in mouse and/or human liver microsome stability testing.
  • SUMMARY OF THE DISCLOSURE [0008] The compounds disclosed in the present application have been discovered to exhibit surprising and unexpected biological effects. In some embodiments, the chemical compounds of the present application are useful as dual-MEK inhibitors exhibiting surprising and unexpected biological effects. [0009] Aspects of the disclosure relate to a method of treating a patient with a RAS- or RAF-mutated cancer.
  • the method includes administering a therapeutically effective amount of an immune checkpoint inhibitor in combination with a compound having the chemical structure of Formula (IV)
  • R 6 is hydrogen, fluoro or chloro
  • R 13 is ethyl or -NR A R B wherein R A is hydrogen and R B is methyl
  • Z 2 is -
  • R 5 is C1 to R 5’ is C 1 to C 6 alkyl.
  • R 5 is methyl.
  • R 5’ is methyl.
  • R 5’ is ethyl.
  • Z2 is -NR 5 R 5’ .
  • R13 is -NRARB.
  • the compound is , or a pharmaceutically acceptable salt thereof. In some embodiments, the compound , or a pharmaceutically acceptable salt thereof. the compound is , or a pharmaceutically acceptable salt thereof. In some embodiments, the the compound , or a pharmaceutically acceptable salt is , or, or a pharmaceutically acceptable salt thereof. In some embodiments, the compound , or a pharmaceutically acceptable salt In some O O O N embodiments, the compound , or a pharmaceutically acceptable salt is , or a pharmaceutically acceptable salt thereof. In some embodiments, the compound , or a pharmaceutically acceptable salt thereof. In some embodiments, Z2 . In some embodiments, R13 is ethyl. In some is , or a pharmaceutically acceptable salt thereof.
  • the compound , or a pharmaceutically acceptable salt thereof. the compound is , or a pharmaceutically acceptable salt thereof.
  • R13 is -NRARB. In some embodiments, the compound is , or a pharmaceutically acceptable salt thereof. In some embodiments, the compound , or a pharmaceutically acceptable salt thereof. the compound is , or a pharmaceutically acceptable salt thereof. In some embodiments, the Z2 . In some embodiments, R13 is ethyl. In some embodiments, R13 is some embodiments, the compound is , or a pharmaceutically acceptable salt thereof. In some embodiments, the compound is , or a pharmaceutically acceptable salt thereof. In some embodiments, the compound is , or a pharmaceutically acceptable salt thereof. In some embodiments, the compound is , or a pharmaceutically acceptable salt thereof. In some embodiments, the compound is , or a pharmaceutically acceptable salt thereof.
  • R 2 is L
  • R 6 is selected from the group consisting of H or fluoro, chloro or bromo
  • R 7 is H
  • R 13 is selected from the group consisting of optionally substituted optionally substituted amin, C1 to C6 alkyl, H, deuterium, hydroxyl, halogen, cyano, nitro, optionally substituted amino, optionally substituted C-amido, optionally substituted N-amido, optionally substituted ester, optionally substituted sulfonyl, optionally substituted S-sulfonamido, optionally substituted N-sulfonamido, optionally substituted sulfonate, optionally substituted O-thiocarbamyl, optionally substituted N-thiocarbamyl, optionally substituted
  • Z2 is –NR 5 R 5 ⁇ .
  • R 5 is methyl.
  • R 5’ is methyl.
  • R 5’ is ethyl.
  • Z 2 is .
  • Z2 is optionally , 4.
  • n is 1.
  • RA and RB are each independently selected from hydrogen, or C1-6 alkyl.
  • R A is hydrogen and R B is methyl.
  • R 13 is C 1 to C 6 alkyl.
  • R 13 is ethyl.
  • the R 6 is fluoro.
  • R 6 is chloro.
  • R 6 is H.
  • aspects of the disclosure relate to a method of treating a patient with a RAS- or RAF-mutated cancer including administering a therapeutically effective amount of an immune checkpoint inhibitor in combination with a compound selected from the list consisting of: , and .
  • aspects of the disclosure relate to a method of treating a patient with a RAS- or RAF-mutated cancer including administering a therapeutically effective amount of an immune checkpoint inhibitor in combination with a compound selected from the list consisting of: .
  • the immune checkpoint inhibitor is an inhibitor of PD-1, PD-L1, PD-L2, PD-L3, PD-L4, CTLA-4, LAG3, B7-H3, B7-H4, KIR or TIM3.
  • the immune checkpoint inhibitor is a PD-1 inhibitor.
  • the immune checkpoint inhibitor is a PD-L2 inhibitor.
  • the immune checkpoint inhibitor is a CTLA-4 inhibitor.
  • the immune checkpoint inhibitor is nivolumab, pembrolizumab, pidilizumab, ipilimumab, BMS 936559, tremelimumab, relatlimab, atezolizumab, avelumab, cemiplimab, durvalumab, tislelizumab, spartalizumab, or any combinations thereof.
  • the RAS- or RAF-mutated cancer is associated with a RAS mutation.
  • the RAS- or RAF-mutated cancer has a RAS mutation that is a KRAS mutation selected from the group consisting of G12C, G12S, G12R, G12F, G12L, G12N, G12A, G12D, G12V, G13C, G13S, G13D, G13V, G13P, S17G, P34S, A59E, A59G, A59T, Q61K, Q61L, Q61R, and Q61H.
  • the immune checkpoint inhibitor is an inhibitor of CTLA-4.
  • the immune checkpoint inhibitor is tremelimumab or ipilimumab.
  • the immune checkpoint inhibitor is tremelimumab. In some embodiments, the immune checkpoint inhibitor is ipilimumab. In some embodiments, the immune checkpoint inhibitor is an inhibitor of PD-1 or PD-L1. In some embodiments, the immune checkpoint inhibitor is pembrolizumab. In some embodiments, the immune checkpoint inhibitor is nivolumab. In some embodiments, the immune checkpoint inhibitor is cemiplimab. In some embodiments, the RAS- or RAF-mutated cancer is Pancreatic adenocarcinoma (PDAC). In some embodiments, the RAS- or RAF-mutated cancer is a RAS-mutated cutaneous melanoma.
  • PDAC Pancreatic adenocarcinoma
  • the RAS- or RAF-mutated cancer is a RAF-mutated cutaneous melanoma. In some embodiments, the RAS- or RAF-mutated cancer is a RAS-mutated NSCLC. In some embodiments, the RAS- or RAF-mutated cancer is an RAS-mutated GI solid tumors other than CRC. In some embodiments, the RAS- or RAF-mutated cancer is a RAF- mutated solid tumor. In some embodiments, the RAF mutation is a class I RAF mutation. In some embodiments, the RAF mutation is BRAF-V600E and BRAF-V600 K. In some embodiments, the RAF mutation is BRAF class II mutation.
  • the RAF mutation is G464V, K601, L597, G464R, G464E, G469, or a frameshift between positions 480 and 495.
  • the RAS- or RAF-mutated cancer is characterized by a ARAF, RAF1 or CRAF) mutation.
  • FIG.1 illustrates exemplary compounds of the disclosure pharmacokinetics compared to analog compounds.
  • FIG. 2A illustrates a graph of pERK:total ERK (activation) in A549 lung cancer model
  • FIG.2B illustrates a graph of pERK:total ERK (activation) in A375 model
  • FIG. 2C illustrates a graph of pERK:total ERK in a SK-MEL-2Melanoma model.
  • FIG.3 illustrates a graph of a colon-26 syngeneic CRC tumor mouse model (BID) study.
  • FIG.4 illustrates a graph of a colon-26 syngeneic CRC tumor mouse model (QD) study.
  • FIG. 5 illustrates a table of exemplary compounds of the disclosure.
  • FIG.6 illustrates a graph of a colon-26 syngeneic CRC tumor mouse model (BID) study.
  • FIG.7 illustrates a graph of a colon-26 syngeneic CRC tumor mouse model (QD) study.
  • FIG.8A illustrates graphs depicting compound 274 with a short plasma and tumor PK half-life in vivo
  • FIG.8B illustrates graphs depicting compound 274 with a short plasma and tumor PK half-life in vivo.
  • FIG. 9A-D illustrates graphs depicting drug pharmacology and maximum effective dose (MED) in mice.
  • FIG.10A-10B illustrates graphs depicting MEKi, ⁇ -PD-1, ⁇ -CTLA-4 alone and combinations in CT-26/MC38. DETAILED DESCRIPTION [0026] In some embodiments, MEK inhibitors are provided.
  • these compounds include compounds having the structure of Formula I as described herein or pharmaceutically acceptable salts thereof.
  • prodrugs, metabolites, stereoisomers, hydrates, solvates, polymorphs, and pharmaceutically acceptable salts of the compounds disclosed herein are provided.
  • therapeutic methods or uses are providing herein for the treatement, prevention, or amelioration of a disease or condition in a subject, these methods comprising administering at least one compound disclosed herein to the subject.
  • therapeutic methods or uses are provided for the treatment, prevention or amelioration of cancer comprising administering of a compound having the structures of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), as described herein.
  • therapeutic methods or uses are provided for the treatment of cancer cachexia comprising administering a compound having the structures of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), as described herein.
  • the term “comprising” means that the compound, composition, or device includes at least the recited features or components, but may also include additional features or components.
  • prodrug refers to an agent that is converted into the parent drug in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent is not. The prodrug may also have improved solubility in pharmaceutical compositions over the parent drug.
  • prodrug a compound which is administered as an ester (the “prodrug”) to facilitate transmittal across a cell membrane where water solubility is detrimental to mobility, but which then is metabolically hydrolyzed to the carboxylic acid, the active entity, once inside the cell where water-solubility is beneficial.
  • prodrug a compound which is administered as an ester (the “prodrug”) to facilitate transmittal across a cell membrane where water solubility is detrimental to mobility, but which then is metabolically hydrolyzed to the carboxylic acid, the active entity, once inside the cell where water-solubility is beneficial.
  • a further example of a prodrug might be a short peptide (polyaminoacid) bonded to an acid group where the peptide is metabolized to reveal the active moiety.
  • Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in Design of Prodrugs, (ed. H.
  • Metabolites of the compounds disclosed herein include active species that are produced upon introduction of the compounds into the biological milieu.
  • Compounds disclosed herein having at least one chiral center they may exist as a racemate or as each enantiomer, and may exist as enantiomeric-enriched mixtures of the enantimoers. It should be noted that all such isomers and mixtures thereof are included in the scope of the present invention.
  • the crystalline forms for the compounds disclosed herein may exist as alternative polymorphs. Such polymorphs are included in one embodiment of the present invention.
  • the compounds of the present invention may form solvates with water (i.e., hydrates) or common organic solvents. Such solvates are included in one embodiment of the present invention.
  • pharmaceutically acceptable salt refers to a salt of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound.
  • the salt is an acid addition salt of the compound.
  • Pharmaceutical salts can be obtained by reacting a compound with inorganic acids such as hydrohalic acid (e.g., hydrochloric acid or hydrobromic acid), sulfuric acid, nitric acid, phosphoric acid and the like.
  • compositions can also be obtained by reacting a compound with an organic acid such as aliphatic or aromatic carboxylic or sulfonic acids, for example acetic, succinic, lactic, malic, tartaric, citric, ascorbic, nicotinic, methanesulfonic, ethanesulfonic, p-toluensulfonic, salicylic or naphthalenesulfonic acid.
  • organic acid such as aliphatic or aromatic carboxylic or sulfonic acids, for example acetic, succinic, lactic, malic, tartaric, citric, ascorbic, nicotinic, methanesulfonic, ethanesulfonic, p-toluensulfonic, salicylic or naphthalenesulfonic acid.
  • Pharmaceutical salts can also be obtained by reacting a compound with a base to form a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, N-methyl-D-glucamine, tris(hydroxymethyl)methylamine, C 1 -C 7 alkylamine, cyclohexylamine, triethanolamine, ethylenediamine, and salts with amino acids such as arginine, lysine, and the like.
  • a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, N-methyl-D-glucamine, tris(hydroxymethyl)methylamine, C 1 -C 7 alkylamine, cycl
  • halogen atom means any one of the radio-stable atoms of column 7 of the Periodic Table of the Elements, e.g., fluorine, chlorine, bromine, or iodine, with fluorine and chlorine being preferred.
  • esters refers to a chemical moiety with formula -(R) n -COOR’, where R and R’ are independently selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon), and where n is 0 or 1.
  • amide refers to a chemical moiety with formula -(R) n -C(O)NHR’ or -(R) n -NHC(O)R’, where R and R’ are independently selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon), and where n is 0 or 1.
  • An amide may be an amino acid or a peptide molecule attached to a molecule of the present invention, thereby forming a prodrug.
  • Any amine, hydroxyl, or carboxyl side chain on the compounds disclosed herein can be esterified or amidified.
  • aromatic refers to an aromatic group which has at least one ring having a conjugated pi electron system and includes both carbocyclic aryl (e.g., phenyl) and heterocyclic aryl groups (e.g., pyridine).
  • carbocyclic aryl e.g., phenyl
  • heterocyclic aryl groups e.g., pyridine
  • the term includes monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of carbon atoms) groups.
  • Carbocyclic refers to a compound which contains one or more covalently closed ring structures, and that the atoms forming the backbone of the ring are all carbon atoms. The term thus distinguishes carbocyclic from heterocyclic rings in which the ring backbone contains at least one atom which is different from carbon.
  • heterocyclic refers to an aromatic group which contains at least one heterocyclic ring.
  • C a to C b in which “a” and “b” are integers refer to the number of carbon atoms in an alkyl, alkenyl or alkynyl group, or the number of carbon atoms in the ring of a cycloalkyl, aryl, heteroaryl or heterocyclyl group. That is, the alkyl, alkenyl, alkynyl, ring of the cycloalkyl, ring of the aryl, ring of the heteroaryl or ring of the heterocyclyl can contain from “a” to “b”, inclusive, carbon atoms.
  • a “C 1 to C 4 alkyl” group or a “C1-C4 alkyl” group refers to all alkyl groups having from 1 to 4 carbons, that is, CH3-, CH3CH2-, CH3CH2CH2-, (CH3)2CH-, CH3CH2CH2CH2-, CH3CH2CH(CH3)- and (CH 3 ) 3 C-.
  • cycloalkyl group may contain from “a” to “b”, inclusive, total atoms, such as a C3-C8 cycloalkyl group, 3 to 8 carbon atoms in the ring(s).
  • a “4 to 7 membered heterocyclyl” group refers to all heterocyclyl groups with 4 to 7 total ring atoms, for example, azetidine, oxetane, oxazoline, pyrrolidine, piperidine, piperazine, morpholine, and the like.
  • C1-C6 includes C1, C2, C3, C4, C5 and C6, and a range defined by any of the two preceding numbers.
  • C 1 -C 6 alkyl includes C 1 , C 2 , C 3 , C 4 , C 5 and C 6 alkyl, C 2 - C6 alkyl, C1-C3 alkyl, etc.
  • C3-C8 carbocyclyl or cycloalkyl each includes hydrocarbon ring containing 3, 4, 5, 6, 7 and 8 carbon atoms, or a range defined by any of the two numbers, such as C 3 -C 7 cycloalkyl or C 5 -C 6 cycloalkyl.
  • 3 to 10 membered heterocyclyl includes 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms, or a range defined by any of the two preceding numbers, such as 4 to 6 membered or 5 to 7 membered heterocyclyl.
  • alkyl refers to a straight or branched hydrocarbon chain fully saturated (no double or triple bonds) hydrocarbon group.
  • the alkyl group may have 1 to 20 carbon atoms (whenever it appears herein, a numerical range such as “1 to 20” refers to each integer in the given range; e.g., “1 to 20 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 20 carbon atoms, although the present definition also covers the occurrence of the term “alkyl” where no numerical range is designated).
  • the alkyl group may also be a medium size alkyl having 1 to 10 carbon atoms.
  • the alkyl group could also be a lower alkyl having 1 to 5 carbon atoms.
  • the alkyl group of the compounds may be designated as “C 1 -C 4 alkyl” or similar designations.
  • “C1-C4 alkyl” indicates that there are one to four carbon atoms in the alkyl chain, i.e., the alkyl chain is selected from the group consisting of methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl.
  • Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl, hexyl, ethenyl, propenyl, butenyl, and the like. [0047] The alkyl group may be substituted or unsubstituted.
  • the substituent group(s) is(are) one or more group(s) individually and independently selected from alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyana
  • alkenyl refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more double bonds. An alkenyl group may be unsubstituted or substituted. When substituted, the substituent(s) may be selected from the same groups disclosed above with regard to alkyl group substitution.
  • the alkenyl group may have 2 to 20 carbon atoms, although the present definition also covers the occurrence of the term “alkenyl” where no numerical range is designated.
  • the alkenyl group may also be a medium size alkenyl having 2 to 9 carbon atoms.
  • the alkenyl group could also be a lower alkenyl having 2 to 4 carbon atoms.
  • the alkenyl group of the compounds may be designated as “C2-4 alkenyl” or similar designations.
  • C2-4 alkenyl indicates that there are two to four carbon atoms in the alkenyl chain, i.e., the alkenyl chain is selected from the group consisting of ethenyl, propen-1-yl, propen-2-yl, propen-3-yl, buten-1-yl, buten-2-yl, buten-3-yl, buten-4-yl, 1-methyl-propen-1-yl, 2-methyl-propen-1-yl, 1-ethyl-ethen-1-yl, 2- methyl-propen-3-yl, buta-1,3-dienyl, buta-1,2,-dienyl, and buta-1,2-dien-4-yl.
  • alkenyl groups include, but are in no way limited to, ethenyl, propenyl, butenyl, pentenyl, and hexenyl, and the like.
  • alkynyl refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more triple bonds. An alkynyl group may be unsubstituted or substituted. When substituted, the substituent(s) may be selected from the same groups disclosed above with regard to alkyl group substitution.
  • the alkynyl group may have 2 to 20 carbon atoms, although the present definition also covers the occurrence of the term “alkynyl” where no numerical range is designated.
  • the alkynyl group may also be a medium size alkynyl having 2 to 9 carbon atoms.
  • the alkynyl group could also be a lower alkynyl having 2 to 4 carbon atoms.
  • the alkynyl group of the compounds may be designated as “C 2-4 alkynyl” or similar designations.
  • C 2-4 alkynyl indicates that there are two to four carbon atoms in the alkynyl chain, i.e., the alkynyl chain is selected from the group consisting of ethynyl, propyn-1-yl, propyn-2-yl, butyn-1-yl, butyn-3-yl, butyn- 4-yl, and 2-butynyl.
  • Typical alkynyl groups include, but are in no way limited to, ethynyl, propynyl, butynyl, pentynyl, and hexynyl, and the like.
  • heteroalkyl refers to a straight or branched hydrocarbon chain containing one or more heteroatoms, that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur, in the chain backbone.
  • the heteroalkyl group may have 1 to 20 carbon atoms although the present definition also covers the occurrence of the term “heteroalkyl” where no numerical range is designated.
  • the heteroalkyl group may also be a medium size heteroalkyl having 1 to 9 carbon atoms.
  • the heteroalkyl group could also be a lower heteroalkyl having 1 to 4 carbon atoms.
  • the heteroalkyl group of the compounds may be designated as “C1-4 heteroalkyl” or similar designations.
  • the heteroalkyl group may contain one or more heteroatoms.
  • C 1-4 heteroalkyl indicates that there are one to four carbon atoms in the heteroalkyl chain and additionally one or more heteroatoms in the backbone of the chain.
  • aryl refers to a carbocyclic (all carbon) ring or two or more fused rings (rings that share two adjacent carbon atoms) that have a fully delocalized pi- electron system. Examples of aryl groups include, but are not limited to, benzene, naphthalene and azulene. An aryl group may be substituted or unsubstituted.
  • substituent group(s) that is(are) one or more group(s) independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, iso
  • substituents on an aryl group may form a non-aromatic ring fused to the aryl group, including a cycloalkyl, cycloalkenyl, cycloalkynyl, and heterocyclyl.
  • heteroaryl refers to a monocyclic or multicyclic aromatic ring system (a ring system with fully delocalized pi-electron system), one or two or more fused rings that contain(s) one or more heteroatoms, that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur.
  • heteroaryl rings include, but are not limited to, furan, thiophene, phthalazine, pyrrole, oxazole, thiazole, imidazole, pyrazole, isoxazole, isothiazole, triazole, thiadiazole, pyridine, pyridazine, pyrimidine, pyrazine and triazine.
  • a heteroaryl group may be substituted or unsubstituted.
  • substituent group(s) that is(are) one or more group(s) independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, iso
  • substituents on a heteroayl group may form a non-aromatic ring fused to the aryl group, including a cycloalkyl, cycloalkenyl, cycloalkynyl, and heterocyclyl.
  • an “aralkyl” or “arylalkyl” refers to an aryl group connected, as a substituent, via an alkylene group.
  • the alkylene and aryl group of an aralkyl may be substituted or unsubstituted. Examples include but are not limited to benzyl, substituted benzyl, 2-phenylethyl, 3-phenylpropyl, and naphtylalkyl.
  • the alkylene group is a lower alkylene group.
  • a “heteroaralkyl” or “heteroarylalkyl” is heteroaryl group connected, as a substituent, via an alkylene group.
  • the alkylene and heteroaryl group of heteroaralkyl may be substituted or unsubstituted. Examples include but are not limited to 2- thienylmethyl, 3-thienylmethyl, furylmethyl, thienylethyl, pyrrolylalkyl, pyridylalkyl, isoxazollylalkyl, and imidazolylalkyl, and their substituted as well as benzo-fused analogs.
  • the alkylene group is a lower alkylene group.
  • a “alkylene” refers to a branched, or straight chain fully saturated di-radical chemical group containing only carbon and hydrogenthat is attached to the rest of the molecule via two points of attachment (i.e., an alkanediyl).
  • the alkylene group may have 1 to 20 carbon atoms, although the present definition also covers the occurrence of the term alkylene where no numerical range is designated.
  • the alkylene group may also be a medium size alkylene having 1 to 9 carbon atoms.
  • the alkylene group could also be a lower alkylene having 1 to 4 carbon atoms.
  • the alkylene group may be designated as “C1-4 alkylene” or similar designations.
  • C1-4 alkylene indicates that there are one to four carbon atoms in the alkylene chain, i.e., the alkylene chain is selected from the group consisting of methylene, ethylene, ethan-1,1-diyl, propylene, propan-1,1-diyl, propan-2,2-diyl, 1-methyl-ethylene, butylene, butan-1,1-diyl, butan-2,2-diyl, 2-methyl-propan-1,1-diyl, 1- methyl-propylene, 2-methyl-propylene, 1,1-dimethyl-ethylene, 1,2-dimethyl-ethylene, and 1- ethyl-ethylene.
  • alkenylene refers to a straight or branched chain di-radical chemical group containing only carbon and hydrogen and containing at least one carbon- carbon double bond that is attached to the rest of the molecule via two points of attachment.
  • the alkenylene group may have 2 to 20 carbon atoms, although the present definition also covers the occurrence of the term alkenylene where no numerical range is designated.
  • the alkenylene group may also be a medium size alkenylene having 2 to 9 carbon atoms.
  • the alkenylene group could also be a lower alkenylene having 2 to 4 carbon atoms.
  • the alkenylene group may be designated as “C2-4 alkenylene” or similar designations.
  • C2-4 alkenylene indicates that there are two to four carbon atoms in the alkenylene chain, i.e., the alkenylene chain is selected from the group consisting of ethenylene, ethen-1,1- diyl, propenylene, propen-1,1-diyl, prop-2-en-1,1-diyl, 1-methyl-ethenylene, but-1-enylene, but-2-enylene, but-1,3-dienylene, buten-1,1-diyl, but-1,3-dien-1,1-diyl, but-2-en-1,1-diyl, but- 3-en-1,1-diyl, 1-methyl-prop-2-en-1,1-diyl, 2-methyl-prop-2-en-1,1-diyl, 1-ethyl-ethenylene, 1,2-dimethyl-ethenylene, 1-methyl-propenylene, 2-methyl-propen
  • arylalkylidene refers to an alkylidene group in which either R’ and R’’ is an aryl group. An alkylidene group may be substituted or unsubstituted.
  • alkoxy refers to the formula –OR wherein R is an alkyl is defined as above, e.g.
  • alkoxy may be substituted or unsubstituted.
  • alkylthio refers to the formula –SR wherein R is an alkyl is defined as above, e.g.
  • aryloxy and “arylthio” refers to RO- and RS-, respectively, in which R is an aryl, such as but not limited to phenyl. Both an aryloxyl and arylthio may be substituted or unsubstituted.
  • Non-limiting examples include formyl, acetyl, propanoyl, benzoyl, and acryl.
  • cycloalkyl refers to a completely saturated (no double bonds) mono- or multi- cyclic hydrocarbon ring system.
  • Cycloalkyl groups may range from C3 to C10, in other embodiments it may range from C3 to C6.
  • a cycloalkyl group may be unsubstituted or substituted.
  • Typical cycloalkyl groups include, but are in no way limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like. If substituted, the substituent(s) may be an alkyl or selected from those indicated above with regard to substitution of an alkyl group unless otherwise indicated.
  • cycloalkenyl refers to a cycloalkyl group that contains one or more double bonds in the ring although, if there is more than one, they cannot form a fully delocalized pi-electron system in the ring (otherwise the group would be “aryl,” as defined herein).
  • the rings When composed of two or more rings, the rings may be connetected together in a fused, bridged or spiro-connected fashion.
  • a cycloalkenyl group may be unsubstituted or substituted.
  • substituent(s) When substituted, the substituent(s) may be an alkyl or selected from the groups disclosed above with regard to alkyl group substitution unless otherwise indicated.
  • substituents on a cycloalkenyl group When substituted, substituents on a cycloalkenyl group may form an aromatic ring fused to the cycloalkenyl group, including an aryl and a heteroaryl.
  • cycloalkynyl refers to a cycloalkyl group that contains one or more triple bonds in the ring. When composed of two or more rings, the rings may be joined together in a fused, bridged or spiro-connected fashion. A cycloalkynyl group may be unsubstituted or substituted.
  • substituent(s) When substituted, the substituent(s) may be an alkyl or selected from the groups disclosed above with regard to alkyl group substitution unless otherwise indicated. When substituted, substituents on a cycloalkynyl group may form an aromatic ring fused to the cycloalkynyl group, including an aryl and a heteroaryl.
  • heteroalicyclic or “heteroalicyclyl” refers to a stable 3- to 18 membered ring which consists of carbon atoms and from one to five heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur.
  • heteroalicyclic or “heteroalicyclyl” may be monocyclic, bicyclic, tricyclic, or tetracyclic ring system, which may be joined together in a fused, bridged or spiro-connected fashion; and the nitrogen, carbon and sulfur atoms in the “heteroalicyclic” or “heteroalicyclyl” may be optionally oxidized; the nitrogen may be optionally quaternized; and the rings may also contain one or more double bonds provided that they do not form a fully delocalized pi-electron system throughout all the rings.
  • Heteroalicyclyl groups may be unsubstituted or substituted.
  • the substituent(s) may be one or more groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, ester, mercapto, alkylthio, arylthio, cyano, halogen, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyan
  • heteroalicyclic or “heteroalicyclyl” include but are not limited to, azepinyl, acridinyl, carbazolyl, cinnolinyl, dioxolanyl, imidazolinyl, morpholinyl, oxiranyl, piperidinyl N-oxide, piperidinyl, piperazinyl, pyrrolidinyl, 4-piperidonyl, pyrazolidinyl, 2-oxopyrrolidinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, and thiamorpholinyl sulfone.
  • substituents on a heteroalicyclyl group may form an aromatic ring fused to the heteroalicyclyl group, including an aryl and a heteroaryl.
  • substituents on a heteroalicyclyl group may form an aromatic ring fused to the heteroalicyclyl group, including an aryl and a heteroaryl.
  • the term “(cycloalkenyl)alkyl” refers to a cycloalkenyl group connected, as a substituent, via an alkylene group.
  • the alkylene and cycloalkenyl of a (cycloalkenyl)alkyl may be substituted or unsubstituted.
  • the alkylene group is a lower alkylene group.
  • (cycloalkynyl)alkyl to a cycloalkynyl group connected, as a substituent, via an alkylene group.
  • the alkylene and cycloalkynyl of a (cycloalkynyl)alkyl may be substituted or unsubstituted.
  • the alkylene group is a lower alkylene group.
  • R can be hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, or (heteroalicyclyl)alkyl, as defined herein.
  • An O-carboxy may be substituted or unsubstituted.
  • a C-carboxy may be substituted or unsubstituted.
  • trimihalomethanesulfonyl refers to an “X3CSO2-“ group wherein X is a halogen.
  • cyano refers to a “-CN” group.
  • cyanato refers to an “-OCN” group.
  • isocyanato refers to a “-NCO” group.
  • thiocyanato refers to a “-SCN” group.
  • the term “isothiocyanato” refers to an “-NCS” group.
  • the term “sulfonyl” refers to an “-SO2R” group in which R can be the same as defined with respect to O-carboxy. A sulfonyl may be substituted or unsubstituted.
  • S-sulfonamido refers to a “-SO2NRARB” group in which R A and R B can be the same as defined with respect to O-carboxy. An S-sulfonamido may be substituted or unsubstituted.
  • N-sulfonamido refers to a “-SO2N(RA)(RB)” group in which R, RA, and RB can be the same as defined with respect to O-carboxy. A sulfonyl may be substituted or unsubstituted.
  • trihalomethanesulfonamido refers to an “X3CSO2N(R)-“ group with X as halogen and R can be the same as defined with respect to O- carboxy.
  • a trihalomethanesulfonamido may be substituted or unsubstituted.
  • amino refers to a “-NRARB” group in which RA and R B are each independently selected from hydrogen, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C3-7 carbocyclyl, C6-10 aryl, 5-10 membered heteroaryl, and 5-10 membered heterocyclyl, as defined herein.
  • aminoalkyl refers to an amino group connected via an alkylene group.
  • lower aminoalkyl refers to an amino group connected via a lower alkylene group. A lower aminoalkyl may be substituted or unsubstituted.
  • the term “lower alkoxyalkyl” refers to an alkoxy group connected via a lower alkylene group.
  • a lower alkoxyalkyl may be substituted or unsubstituted.
  • R can be hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, or (heteroalicyclyl)alkyl, as defined herein.
  • An O-carbamyl can be substituted or unsubstituted.
  • R can be hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, or (heteroalicyclyl)alkyl, as defined herein.
  • An N-carbamyl can be substituted or unsubstituted.
  • R can be hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, or (heteroalicyclyl)alkyl, as defined herein.
  • An O- thiocarbamyl can be substituted or unsubstituted.
  • R can be hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, or (heteroalicyclyl)alkyl, as defined herein.
  • An N-thiocarbamyl can be substituted or unsubstituted.
  • perhaloalkyl refers to an alkyl group where all of the hydrogen atoms are replaced by halogen atoms.
  • halogen refers to any one of the radio- stable atoms of column 7 of the Periodic Table of the Elements, e.g., fluorine, chlorine, bromine, or iodine, with fluorine and chlorine being preferred.
  • carbocyclyl refers to a non-aromatic cyclic ring or ring system containing only carbon atoms in the ring system backbone. When the carbocyclyl is a ring system, two or more rings may be joined together in a fused, bridged or spiro-connected fashion.
  • Carbocyclyls may have any degree of saturation provided that at least one ring in a ring system is not aromatic.
  • carbocyclyls include cycloalkyls, cycloalkenyls, and cycloalkynyls.
  • the carbocyclyl group may have 3 to 20 carbon atoms, although the present definition also covers the occurrence of the term “carbocyclyl” where no numerical range is designated.
  • the carbocyclyl group may also be a medium size carbocyclyl having 3 to 10 carbon atoms.
  • the carbocyclyl group could also be a carbocyclyl having 3 to 6 carbon atoms.
  • the carbocyclyl group may be designated as “C3-6 carbocyclyl” or similar designations.
  • carbocyclyl rings include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, 2,3-dihydro-indene, bicycle[2.2.2]octanyl, adamantyl, and spiro[4.4]nonanyl.
  • (cycloalkyl)alkyl refers to a cycloalkyl group connected, as a substituent, via an alkylene group. The alkylene and cycloalkyl of a (cycloalkyl)alkyl may be substituted or unsubstituted.
  • Examples include but are not limited cyclopropylmethyl, cyclobutylmethyl, cyclopropylethyl, cyclopropylbutyl, cyclobutylethyl, cyclopropylisopropyl, cyclopentylmethyl, cyclopentylethyl, cyclohexylmethyl, cyclohexylethyl, cycloheptylmethyl, and the like.
  • the alkylene group is a lower alkylene group.
  • cycloalkyl refers to a fully saturated carbocyclyl ring or ring system.
  • cycloalkenyl means a carbocyclyl ring or ring system having at least one double bond, wherein no ring in the ring system is aromatic.
  • An example is cyclohexenyl.
  • heterocyclyl refers to three-, four-, five-, six-, seven-, and eight- or more membered rings wherein carbon atoms together with from 1 to 3 heteroatoms constitute said ring.
  • a heterocyclyl can optionally contain one or more unsaturated bonds situated in such a way, however, that an aromatic pi-electron system does not arise.
  • the heteroatoms are independently selected from oxygen, sulfur, and nitrogen.
  • a heterocyclyl can further contain one or more carbonyl or thiocarbonyl functionalities, so as to make the definition include oxo-systems and thio-systems such as lactams, lactones, cyclic imides, cyclic thioimides, cyclic carbamates, and the like.
  • heterocyclyl refers to a non-aromatic cyclic ring or ring system containing at least one heteroatom in the ring backbone.
  • Heterocyclyls may be joined together in a fused, bridged or spiro-connected fashion. Heterocyclyls may have any degree of saturation provided that at least one ring in the ring system is not aromatic.
  • the heteroatom(s) may be present in either a non-aromatic or aromatic ring in the ring system.
  • the heterocyclyl group may have 3 to 20 ring members (i.e., the number of atoms making up the ring backbone, including carbon atoms and heteroatoms), although the present definition also covers the occurrence of the term “heterocyclyl” where no numerical range is designated.
  • the heterocyclyl group may also be a medium size heterocyclyl having 3 to 10 ring members.
  • the heterocyclyl group could also be a heterocyclyl having 3 to 6 ring members.
  • the heterocyclyl group may be designated as “3-6 membered heterocyclyl” or similar designations.
  • the heteroatom(s) are selected from one up to three of O, N or S, and in preferred five membered monocyclic heterocyclyls, the heteroatom(s) are selected from one or two heteroatoms selected from O, N, or S.
  • heterocyclyl rings include, but are not limited to, azepinyl, acridinyl, carbazolyl, cinnolinyl, dioxolanyl, imidazolinyl, imidazolidinyl, morpholinyl, oxiranyl, oxepanyl, thiepanyl, piperidinyl, piperazinyl, dioxopiperazinyl, pyrrolidinyl, pyrrolidonyl, pyrrolidionyl, 4-piperidonyl, pyrazolinyl, pyrazolidinyl, 1,3-dioxinyl, 1,3-dioxanyl, 1,4-dioxinyl, 1,4-dioxanyl, 1,3- oxathianyl, 1,4-oxathianyl, 2H-1,2-oxazinyl, trioxanyl, hexazepinyl, acridinyl,
  • heterocyclylalkyl refers to a heterocyclyl group connected, as a substituent, via an alkylene group. Examples include, but are not limited to, imidazolinylmethyl and indolinylethyl.
  • purified refers to compounds disclosed herein being free of other, dissimilar compounds with which the compounds of the invention are normally associated in their natural state, so that the compounds of the invention comprise at least 0.5%, 1%, 5%, 10%, or 20%, and most preferably at least 50% or 75% of the mass, by weight, of a given sample.
  • Substituted groups are based upon or derived from the unsubstituted parent group in which there has been an exchange of one or more hydrogen atoms for another atom or group. Unless otherwise indicated, when a group is deemed to be “substituted,” the group is substituted with one or more substituents independently selected from C 1 -C 6 alkyl, C 1 -C 6 alkenyl, C 1 -C 6 alkynyl, C 1 -C 6 heteroalkyl, C 3 -C 7 carbocyclyl (optionally substituted with halo, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkyl, and C 1 -C 6 haloalkoxy), C 3 -C 7 -carbocyclyl-C 1 -C 6 - alkyl (optionally substituted with halo, C1-C6 alkyl, C1-C6 alkoxy, C1-C
  • a substituted group is substituted with one or more substituent(s) individually and independently selected from C1-C4 alkyl, amino, hydroxy, and halogen.
  • substituent(s) individually and independently selected from C1-C4 alkyl, amino, hydroxy, and halogen.
  • certain radical naming conventions can include either a mono-radical or a di-radical, depending on the context. For example, where a substituent requires two points of attachment to the rest of the molecule, it is understood that the substituent is a di-radical.
  • a substituent identified as alkyl that requires two points of attachment includes di-radicals such as –CH2–, –CH2CH2–, –CH2CH(CH3)CH2–, and the like.
  • Other radical naming conventions clearly indicate that the radical is a di-radical such as “alkylene” or “alkenylene.”
  • substituent is a group that may be substituted with one or more group(s) individually and independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxyl, alkoxy, aryloxy, mercapto, alkylthio, arylthio, cyano, halo, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N- thio
  • agent or “test agent,” as used herein, includes any substance, molecule, element, compound, entity, or a combination thereof. It includes, but is not limited to, e.g., protein, polypeptide, peptide or mimetic, small organic molecule, polysaccharide, polynucleotide, and the like. It can be a natural product, a synthetic compound, or a chemical compound, or a combination of two or more substances. Unless otherwise specified, the terms “agent”, “substance”, and “compound” are used interchangeably herein.
  • analog refers to a molecule that structurally resembles a reference molecule but which has been modified in a targeted and controlled manner, by replacing a specific substituent of the reference molecule with an alternate substituent. Compared to the reference molecule, an analog would be expected, by one skilled in the art, to exhibit the same, similar, or improved utility. Synthesis and screening of analogs, to identify variants of known compounds having improved characteristics (such as higher binding affinity for a target molecule) is an approach that is well known in pharmaceutical chemistry.
  • mamal as used herein, is used in its usual biological sense.
  • microbial infection refers to the invasion of the host organism, whether the organism is a vertebrate, invertebrate, fish, plant, bird, or mammal, by pathogenic microbes. This includes the excessive growth of microbes that are normally present in or on the body of a mammal or other organism.
  • a microbial infection can be any situation in which the presence of a microbial population(s) is damaging to a host mammal.
  • a mammal is “suffering” from a microbial infection when excessive numbers of a microbial population are present in or on a mammal’s body, or when the effects of the presence of a microbial population(s) is damaging the cells or other tissue of a mammal.
  • this description applies to a bacterial infection.
  • the compounds of preferred embodiments are also useful in treating microbial growth or contamination of cell cultures or other media, or inanimate surfaces or objects, and nothing herein should limit the preferred embodiments only to treatment of higher organisms, except when explicitly so specified in the claims.
  • pharmaceutically acceptable carrier or “pharmaceutically acceptable excipient,” as used herein, includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated.
  • various adjuvants such as are commonly used in the art may be included. Considerations for the inclusion of various components in pharmaceutical compositions are described, e.g., in Gilman et al.
  • subject refers to a human or a non-human mammal, e.g., a dog, a cat, a mouse, a rat, a cow, a sheep, a pig, a goat, a non-human primate or a bird, e.g., a chicken, as well as any other vertebrate or invertebrate.
  • prophylactic treatment refers to treating a subject who does not yet exhibit symptoms of a disease or condition, but who is susceptible to, or otherwise at risk of, a particular disease or condition, whereby the treatment reduces the likelihood that the patient will develop the disease or condition.
  • therapeutic treatment refers to administering treatment to a subject.
  • each chemical element as represented in a compound structure may include any isotope of said element.
  • a hydrogen atom may be explicitly disclosed or understood to be present in the compound.
  • the hydrogen atom can be any isotope of hydrogen, including but not limited to hydrogen-1 (protium), hydrogen-2 (deuterium), and hydrogen-3 (tritium).
  • reference herein to a compound encompasses all potential isotopic forms unless the context clearly dictates otherwise.
  • immune checkpoint inhibitor refers to a molecule (e.g., small molecule, peptide, polypeptide, protein, antibody, antibody fragment and the like) that acts as an inhibitor (antagonist) of an immune checkpoint pathway. Inhibition of a pathway can include blockade of the pathway through binding to a receptor or signaling molecule that is part of the immune checkpoint pathway.
  • Formula (I) is a pharmaceutically acceptable salt as described herein.
  • Formula (I) is represented by Formula (Ia), Formula (Ib), Formula (Ic), or Formula (Id): , , , (Id) are a pharmaceutically acceptable salt as described herein.
  • Some embodiments provide a compound of Formula (II): Q B Y1 Y2 R1 Q A Q C [0130] In acceptable salt as described herein.
  • Formula (II) is represented by Formula (IIa), Formula (IIb), Formula (IIc): , , , .
  • Formula (IIa), Formula (IIb), Formula (IIc), Formula (IId) may be a pharmaceutically acceptable salt as described herein.
  • Some embodiments provide a compound of Formula (III): salt as described herein.
  • Ring A is , , , be cyano, optionally substituted C-amido, optionally substituted N-amido, optionally substituted ester, optionally substituted sulfonyl, optionally substituted S-sulfonamido, optionally substituted N-sulfonamido, optionally substituted sulfonate, optionally substituted O-thiocarbamyl, optionally substituted N-thiocarbamyl, optionally substituted N-carbamyl, optionally substituted O-carbamyl, O-aryl, O-heteroaryl, optionally substituted urea, optionally substituted C 1 to C 6 alkoxy, optionally substituted C 1 to C 6 alkyl, optionally substituted C 2 to C6 alkenyl, optionally substituted C2 to C6 alkynyl, optionally substituted C3 to C8 cycloalkyl, optionally substituted C6 to C10 aryl, optional
  • R 1 is not O-pyrimidinyl. In some embodiments, R 1 is not an ether-linked pyrimidyl.
  • R 2 may be selected from H, deuterium, hydroxyl, halogen, cyano, nitro, optionally substituted amino, optionally substituted C-amido, optionally substituted N-amido, optionally substituted ester, optionally substituted sulfonyl, optionally substituted S-sulfonamido, optionally substituted N-sulfonamido, optionally substituted sulfonate, optionally substituted O-thiocarbamyl, optionally substituted N-thiocarbamyl, optionally substituted N-carbamyl, optionally substituted O-carbamyl, O-aryl, O-heteroaryl
  • R 2 is L. In some further embodiments, R 2 is –CH 3 .
  • R 3 may be selected from H, deuterium, hydroxyl, halogen, cyano, nitro, optionally substituted amino, optionally substituted C-amido, optionally substituted N- amido, optionally substituted ester, optionally substituted sulfonyl, optionally substituted S-sulfonamido, optionally substituted N-sulfonamido, optionally substituted sulfonate, optionally substituted O-thiocarbamyl, optionally substituted N-thiocarbamyl, optionally substituted N-carbamyl, optionally substituted O-carbamyl, optionally substituted urea, optionally substituted C1 to C6 alkoxy, optional
  • R 2 is L. In some further embodiments, R 2 is –CH3. [0140] In some embodiments of the compounds of Formula (I), (Ia), (Ib), (Ic), (Id), (II), or (IIa), R 4 may be selected from H, deuterium, hydroxyl, halogen, cyano, nitro, optionally substituted amino, optionally substituted C-amido, optionally substituted N-amido, optionally substituted ester, optionally substituted sulfonyl, optionally substituted S-sulfonamido, optionally substituted N-sulfonamido, optionally substituted sulfonate, optionally substituted O-thiocarbamyl, optionally substituted N-thiocarbamyl, optionally substituted N-carbamyl, optionally substituted O-carbamyl, optionally substituted urea, optionally substituted C1 to C6 alkoxy, optionally substituted C 1 to C 6
  • R 5 may be selected from H, deuterium, hydroxyl, halogen, cyano, nitro, optionally substituted amino, optionally substituted C 1 to C 6 alkoxy, optionally substituted C 1 to C 6 alkyl, optionally substituted C 2 to C 6 alkenyl, optionally substituted C 2 to C 6 alkynyl.
  • R 5 is H, deuterium, halo, or an optionally susbstituted C1 to C6 alkyl.
  • R 5 ⁇ may be selected from H, deuterium, hydroxyl, halogen, cyano, nitro, optionally substituted amino, optionally substituted C1 to C6 alkoxy, optionally substituted C1 to C6 alkyl, optionally substituted C2 to C6 alkenyl, optionally substituted C2 to C6 alkynyl.
  • R 5 ⁇ is H, deuterium, halo, or an optionally susbstituted C 1 to C 6 alkyl.
  • R 6 may be selected from H, deuterium, hydroxyl, halogen, cyano, nitro, optionally substituted amino, optionally substituted C-amido, optionally substituted N-amido, optionally substituted ester, optionally substituted sulfonyl, optionally substituted S-sulfonamido, optionally substituted N-sulfonamido, optionally substituted sulfonate, optionally substituted O-thiocarbamyl, optionally substituted N-thiocarbamyl, optionally substituted N-carbamyl, optionally substituted O-carbamyl, optionally substituted urea, optionally substituted C1 to C6 alkoxy, optionally substituted C1 to C6 alkyl, optionally substituted C2 to
  • R 6 is selected from the group consisting of H or fluoro, chloro or bromo.
  • R 7 may be selected from H, deuterium, hydroxyl, halogen, cyano, nitro, optionally substituted amino, optionally substituted C-amido, optionally substituted N-amido, optionally substituted ester, optionally substituted sulfonyl, optionally substituted S-sulfonamido, optionally substituted N-sulfonamido, optionally substituted sulfonate, optionally substituted O-thiocarbamyl, optionally substituted N-thiocarbamyl, optionally substituted N-carbamyl, optionally substituted O-carbamyl, optionally substituted urea,
  • R 7 is F, Cl, or Br. In some embodiments, R 7 is Cl. In some embodiments, R 7 may be selected from H, deuterium, hydroxyl, halogen, cyano, nitro, optionally substituted amino, optionally substituted C1 to C6 alkoxy, optionally substituted C1 to C6 alkyl, optionally substituted C2 to C6 alkenyl, optionally substituted C2 to C6 alkynyl. In some embodiments, R 7 is halo. In some embodiments, R 7 is H. In some embodiments, R 7 is selected from the group consisting of H, F, methyl, or methoxy. In some embodiments, R 7 is H or F.
  • R 8 selected from H, deuterium, optionally substituted C 1 to C 6 alkyl, optionally substituted C 2 to C 6 alkenyl, optionally substituted C2 to C6 alkynyl, optionally substituted C3 to C8 carbocyclyl, optionally substituted C6 to C10 aryl, optionally substituted C3 to C8 heterocyclyl, optionally substituted C 3 to C 10 heteroaryl.
  • R 8 is selected from halo, H, deuterium, or CH 3 .
  • R 9 may be selected from hydrogen, deuterium, optionally substituted C1 to C6 alkyl, optionally substituted C 3 to C 8 cycloalkyl, optionally substituted C 6 to C 10 aryl, optionally substituted C 3 to C 8 heterocyclyl, optionally substituted C3 to C10 heteroaryl, -CH2-(optionally substituted aryl), - CH2-(optionally substituted C3 to C8 cycloalkyl) or -CH2-(optionally substituted C3 to C10 heteroaryl).
  • Z 2 is C 3 to C 8 cycloalkyl, optionally substituted C 3 to C8 heterocyclyl, optionally substituted C3 to C8 heteroaryl, –NR 5 R 5 ⁇ , –CH2CH, or –CH2CN.
  • R 10 may be selected from hydrogen, deuterium, optionally substituted C 1 to C 6 alkyl, optionally substituted C 3 to C 8 cycloalkyl, optionally substituted C 6 to C 10 aryl, optionally substituted C 3 to C 8 heterocyclyl, optionally substituted C3 to C10 heteroaryl, -CH2-(optionally substituted aryl), - CH2-(optionally substituted C3 to C8 cycloalkyl) or -CH2-(optionally substituted C3 to C10 heteroaryl).
  • Z 2 is C 3 to C 8 cycloalkyl, optionally substituted C 3 to C8 heterocyclyl, optionally substituted C3 to C8 heteroaryl, –NR 5 R 5 ⁇ , –CH2CH, or –CH2CN.
  • R 11 is independently H, deuterium, optionally substituted C 1 to C 6 alkyl, optionally substituted C 2 to C6 alkenyl, optionally substituted C2 to C6 alkynyl, optionally substituted C3 to C8 carbocyclyl, optionally substituted C6 to C10 aryl, optionally substituted C3 to C8 heterocyclyl, optionally substituted C 3 to C 10 heteroaryl, or L.
  • R 11 is selected from halo, H or CH 3 .
  • R 13 may be selected from H, deuterium, hydroxyl, halogen, cyano, nitro, optionally substituted amino, optionally substituted C-amido, optionally substituted N-amido, optionally substituted ester, optionally substituted sulfonyl, optionally substituted S-sulfonamido, optionally substituted N-sulfonamido, optionally substituted sulfonate, optionally substituted O-thiocarbamyl, optionally substituted N-thiocarbamyl, optionally substituted N-carbamyl, optionally substituted O-carbamyl, optionally substituted urea, optionally substituted C 1 to C 6 alkoxy, optionally substituted C1 to C6 alkyl, optionally substituted C2 to C6 alkenyl, optionally substituted C2 to C6 alkynyl, optionally substituted C3 to C8 cycloalkyl, optional
  • R 13 is C 1 to C 6 alkyl.
  • X may be selected from C(R 5 ) 2 , CH(R 5 ), CH 2 , . In some further embodiments, X is CH2 or –O–.
  • X 1 is N or CH.
  • n is 1, 2, 3, or 4.
  • Y 1 may be selected from C(R 5 )2, CH(R 5 ), CH2, .
  • Y 1 is CH2 or –O–. In some embodiments, Y 1 is . In some embodiments of the compounds of Formula (I), (Ia), 2 (II), or (IIa), Y may from C(R 5 ) 2 , CH(R 5 ), CH 2 , , or In some embodiments, Y 2 is -O-. In some embodiments, . In some embodiments of the compounds of , (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), or (III), L may be selected from –Z1-Z2.
  • L may be selected from –Z 1 -Z 2 -Z 3 .
  • Z1 is –CH2–.
  • Z 2 is C 3 to C 8 cycloalkyl, optionally substituted C 3 to C 8 heterocyclyl, optionally substituted C 3 to C 8 heteroaryl, –NR 5 R 5 , –CH 2 CH, or –CH 2 CN.
  • Z2 is optionally substituted C3 to C8 heterocyclyl.
  • Z2 is –NR 5 R 5 ⁇ .
  • Z1 is –CH2- and Z2 is –NR 5 R 5 ⁇ .
  • Z3 may be selected from hydrogen, deuterium, halo, –COH, –CO2H, –NO2, –CH2CCH, –CH2CN, –NR 5 R 5 ⁇ , –(CO)NH2, –(CO)NR 5 R 5 ⁇ , –SO2-NH2, –R 5 CH3, –R 5 -COH, – R 5 CO 2 H, – R 5 NH 2 , – R 5 NH(COH) , –R 5 (CO)NH 2 , – R 5 NH-SO 2 H, – R 5 SO 2 -NH 2 , –CH2R 5 , –OR 5 , –SO2R 5 –, –CO2R 5 , –NHR 5 ,
  • Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), or (III) is a compound of a disclosed formula, for example Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), or (III), but excluding the compounds: salt as described herein.
  • R6 is hydrogen, fluoro or chloro.
  • R 13 is ethyl or - NRARB wherein RA is hydrogen and RB is methyl.
  • Z2 is -NR 5 R 5’ , .
  • R 5 is C 1 to C 6 alkyl.
  • R 5’ is C1 to C6 alkyl.
  • the compounds of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (IId), (III), or (IV) are selected from Compounds of FIG. 5 and Tables B, C, D, E, and pharmaceutically acceptable salts thereof.
  • Table B Exemplary compounds of the disclosure. No Structure No Structure Table C. Exemplary Compounds of the disclosure.
  • Table D Exemplary Compounds of the disclosure.
  • Table E Exemplary compounds of the disclosure.
  • the pharmaceutically acceptable salt can be an alkaline metal salt.
  • the pharmaceutically acceptable salt can be an alkali metal salt.
  • the pharmaceutically acceptable salt can be an alkali earth metal salt. In some embodiments, the pharmaceutically acceptable salt can be an ammonium salt. Syntheses [0168] Compounds of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or pharmaceutically acceptable salts thereof, described herein may be prepared in various ways, including those known to those skilled in the art. The routes shown and described herein are illustrative only and are not intended, nor are they to be construed, to limit the scope of the claims in any manner whatsoever.
  • compositions comprise a physiologically acceptable surface active agents, carriers, diluents, excipients, smoothing agents, suspension agents, film forming substances, and coating assistants, or a combination thereof; and a compound disclosed herein.
  • Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington's Pharmaceutical Sciences, 18th Ed., Mack Publishing Co., Easton, PA (1990), which is incorporated herein by reference in its entirety.
  • Preservatives Preservatives, stabilizers, dyes, sweeteners, fragrances, flavoring agents, and the like may be provided in the pharmaceutical composition.
  • sodium benzoate, ascorbic acid and esters of p-hydroxybenzoic acid may be added as preservatives.
  • antioxidants and suspending agents may be used.
  • alcohols, esters, sulfated aliphatic alcohols, and the like may be used as surface active agents; sucrose, glucose, lactose, starch, crystallized cellulose, mannitol, light anhydrous silicate, magnesium aluminate, magnesium methasilicate aluminate, synthetic aluminum silicate, calcium carbonate, sodium acid carbonate, calcium hydrogen phosphate, calcium carboxymethyl cellulose, and the like may be used as excipients; magnesium stearate, talc, hardened oil and the like may be used as smoothing agents; coconut oil, olive oil, sesame oil, peanut oil, soya may be used as suspension agents or lubricants; cellulose acetate phthalate as a derivative of a carbohydrate such as cellulose or sugar, or methylacetate-methacrylate copolymer as a derivative of polyvinyl may be used as suspension agents; and plasticizers such as ester phthalates and the like may be used as suspension agents.
  • composition refers to a mixture of a compound disclosed herein with other chemical components, such as diluents or carriers.
  • the pharmaceutical composition facilitates administration of the compound to an organism. Multiple techniques of administering a compound exist in the art including, but not limited to, oral, injection, aerosol, parenteral, and topical administration.
  • Pharmaceutical compositions can also be obtained by reacting compounds with inorganic or organic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.
  • carrier refers to a chemical compound that facilitates the incorporation of a compound into cells or tissues.
  • DMSO dimethyl sulfoxide
  • carrier refers to chemical compounds diluted in water that will dissolve the compound of interest as well as stabilize the biologically active form of the compound. Salts dissolved in buffered solutions are utilized as diluents in the art.
  • buffered solution is phosphate buffered saline because it mimics the salt conditions of human blood.
  • an “excipient” refers to an inert substance that is added to a pharmaceutical composition to provide, without limitation, bulk, consistency, stability, binding ability, lubrication, disintegrating ability etc., to the composition.
  • a “diluent” is a type of excipient.
  • compositions comprising the compound, alone or in a mixture with other compounds of the genus or sub- genus, or with alternative compounds described herein, or with one or more alternative pharmaceutically active compounds, and one or more pharmaceutically acceptable carrier, diluent, excipient or combination thereof.
  • the pharmaceutical compositions described herein can be administered to a human patient per se, or in pharmaceutical compositions where they are mixed with other active ingredients, as in combination therapy, or carriers, diluents, excipients or combinations thereof. Proper formulation is dependent upon the route of administration chosen. Techniques for formulation and administration of the compounds described herein are known to those skilled in the art.
  • compositions disclosed herein may be manufactured in any manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or tableting processes. Additionally, the active ingredients are contained in an amount effective to achieve its intended purpose. Many of the compounds used in the pharmaceutical combinations disclosed herein may be provided as salts with pharmaceutically compatible counterions. [0179] The pharmaceutical compositions described herein can be administered to a human patient per se, or in pharmaceutical compositions where they are mixed with other active ingredients, as in combination therapy, or suitable carriers or excipient(s).
  • Suitable routes of administration may, for example, include oral, rectal, transmucosal, topical, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intranasal, or intraocular injections.
  • the compounds can also be administered in sustained or controlled release dosage forms, including depot injections, osmotic pumps, pills, transdermal (including electrotransport) patches, and the like, for prolonged and/or timed, pulsed administration at a predetermined rate.
  • compositions of the present invention may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or tabletting processes.
  • Pharmaceutical compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g., in Remington’s Pharmaceutical Sciences, above.
  • Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions.
  • Suitable excipients are, for example, water, saline, dextrose, mannitol, lactose, lecithin, albumin, sodium glutamate, cysteine hydrochloride, and the like.
  • the injectable pharmaceutical compositions may contain minor amounts of nontoxic auxiliary substances, such as wetting agents, pH buffering agents, and the like.
  • Physiologically compatible buffers include, but are not limited to, Hanks’s solution, Ringer’s solution, or physiological saline buffer.
  • absorption enhancing preparations for example, liposomes
  • penetrants appropriate to the barrier to be permeated may be used in the formulation.
  • Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water- soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or other organic oils such as soybean, grapefruit or almond oils, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • a suitable vehicle e.g., sterile pyrogen-free water
  • the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Pharmaceutical preparations for oral use can be obtained by combining the active compounds with solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • compositions may take the form of tablets or lozenges formulated in conventional manner.
  • suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • a suitable powder base such as lactose or starch.
  • suitable penetrants for these uses are generally known in the art.
  • Pharmaceutical compositions for intraocular delivery include aqueous ophthalmic solutions of the active compounds in water-soluble form, such as eyedrops, or in gellan gum (Shedden et al., Clin.
  • compositions for intranasal delivery may also include drops and sprays often prepared to simulate in many respects nasal secretions to ensure maintenance of normal ciliary action.
  • suitable formulations are most often and preferably isotonic, slightly buffered to maintain a pH of 5.5 to 6.5, and most often and preferably include antimicrobial preservatives and appropriate drug stabilizers.
  • Pharmaceutical formulations for intraauricular delivery include suspensions and ointments for topical application in the ear. Common solvents for such aural formulations include glycerin and water.
  • the compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • a suitable pharmaceutical carrier may be a cosolvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase.
  • a common cosolvent system used is the VPD co-solvent system, which is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant Polysorbate 80TM, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol.
  • VPD co-solvent system is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant Polysorbate 80TM, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol.
  • the proportions of a co-solvent system may be varied considerably without destroying its solubility and toxicity characteristics.
  • co-solvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of POLYSORBATE 80TM; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g., polyvinyl pyrrolidone; and other sugars or polysaccharides may substitute for dextrose.
  • other delivery systems for hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs. Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity.
  • the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
  • sustained-release materials have been established and are well known by those skilled in the art.
  • Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days.
  • additional strategies for protein stabilization may be employed.
  • Agents intended to be administered intracellularly may be administered using techniques well known to those of ordinary skill in the art. For example, such agents may be encapsulated into liposomes. All molecules present in an aqueous solution at the time of liposome formation are incorporated into the aqueous interior.
  • the liposomal contents are both protected from the external micro-environment and, because liposomes fuse with cell membranes, are efficiently delivered into the cell cytoplasm.
  • the liposome may be coated with a tissue-specific antibody.
  • the liposomes will be targeted to and taken up selectively by the desired organ.
  • small hydrophobic organic molecules may be directly administered intracellularly.
  • Additional therapeutic or diagnostic agents may be incorporated into the pharmaceutical compositions.
  • pharmaceutical compositions may be combined with other compositions that contain other therapeutic or diagnostic agents.
  • Parenteral Pharmaceutical Composition [0197] To prepare a parenteral pharmaceutical composition suitable for administration by injection (subcutaneous, intravenous, or the like), 0.1 mg to 120 mg of a water-soluble salt/soluble material itself/solubilized complex of a compound of a preferred embodiment is dissolved in sterile water and then mixed with 10 ⁇ L of 0.9% sterile saline. The mixture is incorporated into a dosage unit form suitable for administration by injection.
  • Injectable Pharmaceutical Composition [0198] To prepare an injectable formulation, 0.1 mg to 100 mg of a compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), 2.0 mL of sodium acetate buffer solution (0.4 M), HCl (1 N) or NaOH (1 M) (q.s. to suitable pH), water (distilled, sterile) (q.s. to 20 mL) are mixed. All of the above ingredients, except water, are combined and stirred and if necessary, with slight heating if necessary. A sufficient quantity of water is then added.
  • Oral Pharmaceutical Composition To prepare a pharmaceutical composition for oral delivery, 0.1 mg to 120 mg of a compound of an embodiment is mixed with 750 mg of starch. The mixture is incorporated into an oral dosage unit, such as a hard gelatin capsule, or 0.1 mg to 120 mg of compound is granulated with binder solution such as starch solution along with suitable diluents such as microcrystalline cellulose or like, disintegrants such as croscaramellose sodium, dry the resultant mixture and add lubricant and compress into tablet which is suitable for oral administration.
  • binder solution such as starch solution along with suitable diluents such as microcrystalline cellulose or like, disintegrants such as croscaramellose sodium
  • Sublingual (Hard Lozenge) Pharmaceutical Composition [0200] To prepare a pharmaceutical composition for buccal delivery, such as a hard lozenge, 0.1 mg to 120 mg of a compound of a preferred embodiment is mixed with 420 mg of powdered sugar/mannitol/xylitol or such sugars that provide negative heat of solution to the system, 1.6 mL of light corn syrup, 2.4 mL distilled water, and 0.42 mL mint extract or other flavorants. The mixture is blended and poured into a mold to form a lozenge suitable for buccal administration.
  • a fast-disintegrating sublingual tablet is prepared by mixing 48.5% by weight of a compound of a preferred embodiment, 20% by weight of microcrystalline cellulose (KG-802), 24.5% by weight of either mannitol or modified dextrose or combination that help dissolve the compressed tablet faster in the mouth, 5% by weight of low-substituted hydroxypropyl cellulose (50 ⁇ m), and 2% by weight of magnesium stearate. Tablets are prepared by direct compression (AAPS PharmSciTech. 2006; 7(2):E41). The total weight of the compressed tablets is maintained at 150 mg.
  • the formulation is prepared by mixing the amount of the compound of a preferred embodiment with the total quantity of microcrystalline cellulose (MCC) and mannitol/modified dextrose or combination, and two-thirds of the quantity of low-substituted hydroxypropyl cellulose (L-HPC) by using a three dimensional manual mixer (Inversina, Bioengineering AG, Switzerland) for 4.5 minutes. All of the magnesium stearate (MS) and the remaining one-third of the quantity of L-HPC are added 30 seconds before the end of mixing.
  • MCC microcrystalline cellulose
  • L-HPC low-substituted hydroxypropyl cellulose
  • a pharmaceutical composition for inhalation delivery 0.1 mg to 100 mg of a compound of a preferred embodiment is mixed with 50 mg of anhydrous citric acid and 100 mL of 0.9% sodium chloride solution. The mixture is incorporated into an inhalation delivery unit, such as a nebulizer, which is suitable for inhalation administration.
  • an inhalation delivery unit such as a nebulizer, which is suitable for inhalation administration.
  • Nebulizer Suspension Pharmaceutical Composition [0203]
  • a compound of a preferred embodiment 0.1 mg to 100 mg
  • Span 85 (1 g) is added followed by addition of dextrose (5.5 g) and ascorbic acid (10 mg).
  • Transdermal Patch Pharmaceutical Composition 0.1 mg to 100 mg of a compound of a preferred embodiment is embedded in, or deposited on, a patch with a single adhesive face. The resulting patch is then attached to the skin via the adhesive face for transdermal administration.
  • Topical Gel Pharmaceutical Composition 0.1 mg to 100 mg of a compound of a preferred embodiment is mixed with 1.75 g of hydroxypropyl cellulose, 10 mL of propylene glycol, 10 mL of isopropyl myristate and 100 mL of purified alcohol USP. The resulting gel mixture is then incorporated into containers, such as tubes, which are suitable for topical administration.
  • Ophthalmic Solution 0.1 mg to 100 mg of a compound of a preferred embodiment is mixed with 0.9 g of NaCl in 100 mL of purified water and filtered using a 0.2 micron filter.
  • ophthalmic delivery units such as eye drop containers, which are suitable for ophthalmic administration.
  • Nasal Spray Solution To prepare a pharmaceutical nasal spray solution, 0.1 mg to 100 mg of a compound of a preferred embodiment is mixed with 30 mL of a 0.05M phosphate buffer solution (pH 4.4). The solution is placed in a nasal administrator designed to deliver 100 ⁇ l of spray for each application.
  • one or more immune checkpoint inhibitor may be co-administered with a compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV).
  • a review describing immune checkpoint pathways and the blockade of such pathways with immune checkpoint inhibitor compounds is provided by Pardoll in Nature Reviews Cancer (April, 2012), pages 252-264, which is incorporated herein by reference in its entirety.
  • Immune check point inhibitor compounds display anti-tumor activity by blocking one or more of the endogenous immune checkpoint pathways that downregulate an anti- tumor immune response.
  • the inhibition or blockade of an immune checkpoint pathway typically involves inhibiting a checkpoint receptor and ligand interaction with an immune checkpoint inhibitor compound to reduce or eliminate the down regulation signal and resulting diminishment of the anti-tumor response.
  • the immune checkpoint inhibitor compound inhibits the signaling interaction between an immune checkpoint receptor and the corresponding ligand of the immune checkpoint receptor.
  • the immune checkpoint inhibitor compound can act by blocking activation of the immune checkpoint pathway by inhibition (antagonism) of an immune checkpoint receptor (some examples of receptors include CTLA-4, PD-1, LAG-3, TIM-3, BTLA, and KIR) or by inhibition of a ligand of an immune checkpoint receptor (some examples of ligands include PD-L1 and PD-L2).
  • the effect of the immune checkpoint inhibitor compound is to reduce or eliminate down regulation of certain aspects of the immune system anti-tumor response in the tumor microenvironment.
  • the Programmed Death 1 (PD-1) protein is an inhibitory member of the extended CD28/CTLA-4 family of T cell regulators (Okazaki et al.
  • CD28 CD28
  • CTLA-4 CTLA-4
  • ICOS BTLA
  • PD-1 is suggested to exist as a monomer, lacking the unpaired cysteine residue characteristic of other CD28 family members. PD-1 is expressed on activated B cells, T cells, and monocytes. [0211] The PD-1 gene encodes a 55 kDa type I transmembrane protein (Agata et al. (1996) Int Immunol. 8:765-72, which is incorporated herein by reference in its entirety).
  • PD-1 Although structurally similar to CTLA-4, PD-1 lacks the MYPPY motif that is important for B7-1 and B7-2 binding.
  • Two ligands for PD-1 have been identified, PD-L1 (B7-H1) and PD- L2 (B7-DC), that have been shown to downregulate T cell activation upon binding to PD-1 (Freeman et al. (2000) J. Exp. Med. 192:1027-34; Carter et al. (2002) Eur. J. Immunol. 32:634- 43; which are incorporated herein by reference in their entirety).
  • Both PD-L1 and PD-L2 are B7 homologs that bind to PD-1, but do not bind to other CD28 family members.
  • PD-L1 is abundant in a variety of human cancers (Dong et al. (2002) Nat. Med. 8:787-9, which is incorporated herein by reference in its entirety).
  • PD-1 is known as an immunoinhibitory protein that negatively regulates TCR signals (Ishida, Y. et al. (1992) EMBO J. 11:3887-3895; Blank, C. et al. (Epub 2006 Dec. 29) Immunol. Immunother. 56(5):739-745; which are incorporated herein by reference in their entirety).
  • the interaction between PD-1 and PD-L1 can act as an immune checkpoint, which can lead to, e.g., a decrease in tumor infiltrating lymphocytes, a decrease in T-cell receptor mediated proliferation, and/or immune evasion by cancerous cells (Dong et al. (2003) J. Mol. Med. 81:281-7; Blank et al. (2005) Cancer Immunol. Immunother. 54:307-314; Konishi et al. (2004) Clin. Cancer Res. 10:5094-100; which are incorporated herein by reference in their entirety).
  • Immune suppression can be reversed by inhibiting the local interaction of PD-1 with PD-L1 or PD-L2; the effect is additive when the interaction of PD-1 with PD-L2 is blocked as well (Iwai et al. (2002) Proc. Nat'l. Acad. Sci. USA 99:12293-7; Brown et al. (2003) J. Immunol. 170:1257-66; which are incorporated herein by reference in their entirety).
  • CTLA-4 cytotoxic T-lymphocyte associated antigen 4
  • CTLA-4 can downregulate T-cell activation through competitive binding and sequestration of CD80 and CD86.
  • CTLA-4 has been shown to be involved in enhancing the immunosuppressive activity of T Reg cells.
  • the immune checkpoint receptor programmed death 1 (PD-1) is expressed by activated T-cells upon extended exposure to antigen. Engagement of PD-1 with its known binding ligands, PD-L1 and PD-L2, occurs primarily within the tumor microenvironment and results in downregulation of anti-tumor specific T-cell responses. Both PD-L1 and PD-L2 are known to be expressed on tumor cells. The expression of PD-L1 and PD-L2 on tumors has been correlated with decreased survival outcomes.
  • the immune checkpoint receptor T cell membrane protein 3 (TIM-3) is expressed on Th1 and Tc1 cells, but not other T-cells. Interaction of TIM-3 with its ligand, galectin-9, produces a Th1 cell death signal. TIM-3 has been reported to play a role in maintaining T-cell exhaustion and blockade of TIM-3 has been shown to restore activity to exhausted T-cells.
  • the immune checkpoint receptor B- and T-lymphocyte attenuator (BTLA) receptor is expressed on both resting and activated B-cells and T-cells. Activation of BTLA when combined with its ligand HVEM (herpes virus entry mediator) results in downregulation of both T-cell activation and proliferation.
  • HVEM herpes virus entry mediator
  • HVEM is expressed by certain tumors (e.g., melanoma) and tumor-associated endothelial cells.
  • the immune checkpoint receptors known as killer cell immunoglobulin- like receptors (KIR) are a polymorphic family of receptors expressed on NK cells and some T- cells and function as regulators of immune tolerance associated with natural killer (NK) cells. Blocking certain KIR receptors with inhibitor compounds can facilitate the destruction of tumors through the increased activity of NK cells.
  • the immune checkpoint inhibitor compound is a small organic molecule (molecular weight less than 1000 daltons), a peptide, a polypeptide, a protein, an antibody, an antibody fragment, or an antibody derivative.
  • the immune checkpoint inhibitor compound is an antibody.
  • the antibody is a monoclonal antibody, specifically a human or a humanized monoclonal antibody.
  • Monoclonal antibodies, antibody fragments, and antibody derivatives for blocking immune checkpoint pathways can be prepared by any of several methods known to those of ordinary skill in the art, including but not limited to, somatic cell hybridization techniques and hybridoma, methods. Hybridoma generation is described in Antibodies, A Laboratory Manual, Harlow and Lane, 1988, Cold Spring Harbor Publications, New York, which is incorporated herein by reference in its entirety. Human monoclonal antibodies can be identified and isolated by screening phage display libraries of human immunoglobulin genes by methods described for example in U.S. Pat. Nos.
  • Monoclonal antibodies can be prepared using the general methods described in U.S. Pat. No. 6,331,415 (Cabilly), which is incorporated herein by reference in its entirety.
  • human monoclonal antibodies can be prepared using a XenoMouseTM (Abgenix, Freemont, Calif.) or hybridomas of B cells from a XenoMouse.
  • a XenoMouse is a murine host having functional human immunoglobulin genes as described in U.S. Pat. No.
  • Patent Application No.2011/0271358 (Freeman), which are incorporated herein by reference in their entirety.
  • the preparation and therapeutic uses of anti-PD-L1 antibodies are described in U.S. Pat. No. 7,943,743 (Korman), which is incorporated herein by reference in its entirety.
  • the preparation and therapeutic uses of anti-TIM-3 antibodies are described in U.S. Pat. No.8,101,176 (Kuchroo) and U.S. Pat. No. 8,552,156 (Tagayanagi), which are incorporated herein by reference in their entirety.
  • the preparation and therapeutic uses of anti-LAG-3 antibodies are described in U.S. Patent Application No.
  • the one or more immune checkpoint inhibitor is an inhibitor of PD-1, PD-L1, or CTLA-4. In some embodiments, the immune checkpoint inhibitor is a PD-1 inhibitor.
  • the immune checkpoint inhibitor is a binding ligand of PD-L1. In some embodiments, the immune checkpoint inhibitor is a PD-L1 inhibitor. In some embodiments, the immune checkpoint inhibitor is a CTLA-4 inhibitor. [0223] In some embodiments, the one or more immune checkpoint inhibitor as described herein includes a first immune checkpoint inhibitor and a second immune checkpoint inhibitor, wherein the first immune checkpoint inhibitor is different from the second immune checkpoint inhibitor. In some embodiments, the first and the second immune checkpoint inhibitor are independently an inhibitor of PD-1, PD-L1 or CTLA-4. In some embodiments, the first immune checkpoint inhibitor is a PD-1 inhibitor, and the second immune checkpoint inhibitor is a CTLA-4 inhibitor.
  • the immune checkpoint inhibitor is pembrolizumab, nivolumab, cemiplimab, atezolizumab, avelumab, pembrolizumab, pidilizumab, ipilimumab, BMS 936559, durvalumab, or any combinations thereof.
  • the one or more immune checkpoint inhibitor may include an anti-PD-1 HuMAbs can be selected from 17D8, 2D3, 4H1, 5C4 (also referred to herein as nivolumab), 4A11, 7D3 and 5F4, all of which are described in U.S. Pat. No. 8,008,449, which is incorporated herein by reference in its entirety.
  • the anti-PD-1 HuMAbs can be selected from 3G10, 12A4 (also referred to herein as BMS-936559), 10A5, 5F8, 10H10, 1B12, 7H1, 11E6, 12B7, and 13G4, all of which are described in U.S. Pat. No.7,943,743, which is incorporated herein by reference in its entirety.
  • the one or more immune checkpoint inhibitor may be incorporated in a pharmaceutically acceptable formulation.
  • the one or more immune checkpoint inhibitor is incorporated in a pharmaceutically acceptable aqueous formulation.
  • the immune checkpoint inhibitor compound is incorporated in a pharmaceutically acceptable liposome formulation, wherein the formulation is a passive or targeted liposome formulation.
  • a pharmaceutically acceptable liposome formulation examples include isotonic buffered and pH 4.5-8 adjusted saline solutions such as Lactated Ringer's Solution and the like.
  • the immune checkpoint inhibitor compound is incorporated in a pharmaceutically acceptable liposome formulation, wherein the formulation is a passive or targeted liposome formulation. Examples of methods for the preparation of suitable liposome formulations of antibodies are described U.S. Pat. No. 5,399,331 (Loughrey), U.S. Pat. No. 8,304,565 (Wu) and U.S. Pat. No. 7,780,882 (Chang), which are incorporated herein by reference in their entirety.
  • the one or more immune checkpoint inhibitor may be an antibody.
  • the antibody is a dry, lyophilized solid that is reconstituted with an aqueous reconstitution solvent prior to use.
  • the antibody is incorporated in a pharmaceutically acceptable formulation and the pharmaceutically acceptable formulation is injected directly into a tumor.
  • the immune checkpoint inhibitor antibody is incorporated in a pharmaceutically acceptable formulation and the pharmaceutically acceptable formulation is injected into the peritumoral region surrounding a tumor. The peritumoral region may contain antitumor immune cells.
  • the antibody is incorporated in a pharmaceutically acceptable formulation and the pharmaceutically acceptable formulation is administered by intravenous injection or infusion.
  • the immune checkpoint inhibitor antibody is incorporated in a pharmaceutically acceptable formulation and the pharmaceutically acceptable formulation is administered by subcutaneous injection or intradermal injection. In some embodiments, the antibody is incorporated in a pharmaceutically acceptable formulation and the pharmaceutically acceptable formulation is administered by intraperitoneal injection or lavage.
  • the precise amount of immune checkpoint inhibitor compound incorporated in a particular method or therapeutic combination of the disclosure may vary according to factors known in art such as for example, the physical and clinical status of the subject, the method of administration, the content of the formulation, the physical and chemical nature of the immune checkpoint inhibitor compound, the intended dosing regimen or sequence. Those of ordinary skill in the art, however, can readily determine the appropriate amount with due consideration of such factors.
  • a compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof is co-administered with a CTLA-4 receptor inhibitor compound.
  • a compound of Formula (I) is co-administered a PD-1 or PD-L1 receptor inhibitor compound.
  • the method comprises treating a subject by co- administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and a LAG- 3 receptor inhibitor compound.
  • the method comprises treating a subject by co-administering a therapeutically effective amount of a compound of Formula (I) and a TIM-3 receptor inhibitor compound.
  • the method comprises treating a subject by co-administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and a BTLA receptor inhibitor compound.
  • the method comprises treating a subject by co-administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and a KIR receptor inhibitor compound.
  • the method comprises treating a subject by co-administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and a PD-L1 inhibitor compound.
  • the method comprises treating a subject by co-administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and a PD-L2 inhibitor compound.
  • the method comprises treating a subject by co-administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and a blocking antibody of an immune checkpoint pathway.
  • the method comprises treating a subject by co-administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and an anti-CTLA-4 receptor antibody.
  • the method comprises treating a subject by co-administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and an anti-PD-1 receptor antibody.
  • the method comprises co-administering to a subject having a tumor a therapeutically effective amount of the compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and an anti-LAG-3 receptor antibody.
  • the method comprises co-administering to a subject having a tumor a therapeutically effective amount of the compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and an anti-TIM-3 receptor antibody.
  • the method comprises co- administering to a subject having a tumor a therapeutically effective amount of the compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and an anti-BTLA receptor antibody.
  • the method comprises co-administering to a subject having a tumor a therapeutically effective amount of the compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and an anti-KIR receptor antibody.
  • the anti-KIR receptor antibody is lirilumab.
  • the method comprises co-administering to a subject having a tumor a therapeutically effective amount of the compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and an anti-PD-1 antibody.
  • the anti-PD-1 antibody is lambrolizumab, pidilizumab, or nivolumab.
  • the method comprises co-administering to a subject having a tumor a therapeutically effective amount of the compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and an anti-PD-L1 antibody.
  • the method comprises co-administering to a subject having a tumor a therapeutically effective amount of the compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and an anti-PD-L2 antibody.
  • the method comprises co-administering to a subject having a tumor a therapeutically effective amount of the compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and an anti-CTLA-4 antibody.
  • the anti-CTLA-4 antibody is ipilimumab or tremelimumab.
  • a method comprises co-administering a compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and an immune checkpoint inhibitor for treating, preventing, or ameliorating a cancer or tumor in a subject by administering a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and an immune checkpoint inhibitor.
  • the subject was resistant to prior treatment with only an immune checkpoint inhibitor.
  • a method for treating a subject with a cancer or tumor comprises administering a compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and a PD-1 inhibitor.
  • a method for treating a subject with a cancer or tumor comprises administering a compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and a PD-L1 inhibitor.
  • a method for treating a subject with a cancer or tumor comprises administering a compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and a PD-L2 inhibitor.
  • a method for treating a subject with a cancer or tumor comprises administering a compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), or (IV), or a pharmaceutically acceptable salt thereof, and a CTLA-4 inhibitor.
  • a method for treating a subject with a cancer or tumor comprises administering a compound of Formula (I), a PD-1 inhibitor and a CTLA-4 inhibitor. In some embodiments, a method for treating a subject with a cancer or tumor comprises administering a compound of Formula (I) and a LAG-3 inhibitor. In some embodiments, a method for treating a subject with a cancer or tumor comprises administering a compound of Formula (I) and a KIR inhibitor. In some embodiments, a method for treating a subject with a cancer or tumor comprises administering a compound of Formula (I) and a TIM-3 inhibitor.
  • a method for treating a subject with a cancer or tumor comprises administering a compound of Formula (I) and a BTLA inhibitor.
  • a method for treating a subject comprises treating a subject having exhibited resistance to a PD-1 inhibitor by administering a therapeutically effective amount of a compound of Formula (I).
  • a method comprises treating a subject having exhibited resistance to a PD-L1 inhibitor by administering a therapeutically effective amount of a compound of Formula (I).
  • a method comprises treating a subject having exhibited resistance to a PD-L2 inhibitor by administering a therapeutically effective amount of a compound of Formula (I).
  • a method comprises treating a subject having exhibited resistance to a CTLA-4 inhibitor by administering a therapeutically effective amount of a compound of Formula (I).
  • the method includes administering the compound of Formula (I) when the subject has exhibited resistance to two different immune checkpoint inhibitors.
  • the two different immune inhibitors can be selected from a CTLA-4 receptor inhibitor, a PD-1 receptor inhibitor, a LAG-3 receptor inhibitor, a TIM-3 receptor inhibitor, a BTLA receptor inhibitor, a KIR receptor inhibitor a PD-L1 inhibitor or a PD-L2 inhibitor.
  • a method comprises treating a subject having exhibited resistance to a PD-1 inhibitor and CTLA-4 inhibitor by administering a therapeutically effective amount of a compound of Formula (I). In some embodiments, a method comprises treating a subject having exhibited resistance to a PD-L1 inhibitor and a CTLA-4 inhibitor by administering a therapeutically effective amount of a compound of Formula (I).
  • a method comprises treating a subject having exhibited resistance to a PD-1 inhibitor, a PD-L1 inhibitor, and a CTLA-4 inhibitor by administering a therapeutically effective amount of a compound of Formula (I) (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), or a pharmaceutically acceptable salt thereof.
  • a method of treating a patient with a RAS- or RAF- mutated cancer comprises administering a therapeutically effective amount of an immune checkpoint inhibitor in combination with a compound having a structure of Compound 274: or a pharmaceutically thereof.
  • the immune checkpoint inhibitor is an inhibitor of PD-1, PD-L1, PD-L2, PD-L3, PD-L4, CTLA-4, LAG3, B7-H3, B7-H4, KIR or TIM3.
  • the immune checkpoint inhibitor is a PD-1 inhibitor.
  • the immune checkpoint inhibitor is a PD-L2 inhibitor.
  • the immune checkpoint inhibitor is a CTLA-4 inhibitor.
  • the immune checkpoint inhibitor is nivolumab, pembrolizumab, pidilizumab, ipilimumab, BMS 936559, tremelimumab, relatlimab, atezolizumab, avelumab, cemiplimab, durvalumab, tislelizumab, spartalizumab, or any combinations thereof.
  • the cancer is selected from the group consisting of brain cancer, breast cancer, lung cancer, non-small cell lung cancer, ovarian cancer, pancreatic cancer, stomach cancer, prostate cancer, renal cancer, colorectal cancer or leukemia.
  • the cancer is associated with a RAS mutation.
  • the cancer has a RAS mutation that is a KRAS mutation selected from the group consisting of G12C, G12S, G12R, G12F, G12L, G12N, G12A, G12D, G12V, G13C, G13S, G13D, G13V, G13P, S17G, P34S, A59E, A59G, A59T, Q61K, Q61L, Q61R, and Q61H.
  • the Compound 274 is administered orally to the patient in need thereof. In some embodiments, the Compound 274 is administered only twice per day to the patient in need thereof.
  • the cancer is associated with a RAS mutated cancer is a RAS-mutated cutaneous melanoma. In some embodiments, the cancer is a RAF-mutated cutaneous melanoma. In some embodiments, the cancer is a RAS- mutated NSCLC. In some embodiments, the cancer is an RAS-mutated GI solid tumors other than CRC. In some embodiments, the cancer is a RAF-mutated solid tumor. In some embodiments, the cancer comprises a RAF mutation and the RAF mutation is a class I RAF mutation. In some embodiments, the RAF mutation is BRAF-V600E and BRAF-V600 K. In some embodiments, the RAF mutation is BRAF class II mutation.
  • the RAF mutation is G464V, K601, L597, G464R, G464E, G469, or a frameshift between positions 480 and 495.
  • the RAS- or RAF-mutated cancer is characterized by a ARAF, RAF1 or CRAF) mutation.
  • the method comprises the treatment of a patient diagnosed with cancer by administration of a compound disclosed herein of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with a human cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) blocking therapy.
  • CTLA-4 human cytotoxic T-lymphocyte-associated antigen 4
  • CTLA-4 is a negative regulator of T-cell activity.
  • the CTLA-4 blocking therapy is tremelimumab.
  • the CTLA-4 blocking therapy comprises the administration of tremelimumab in combination with the compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV).
  • Tremelimumab is a monoclonal antibody that binds to CTLA- 4 and blocks the interaction with its ligands CD80 and CD86, releasing CTLA-4- mediated inhibition of T-cell activation.
  • Tremelimumab is a cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) blocking human IgG2 monoclonal antibody, is produced by recombinant DNA technology in NS0 cell suspension culture and has a molecular weight of 149 kDa.
  • CTLA-4 cytotoxic T-lymphocyte-associated antigen 4
  • Tremelimumab injection is a sterile, preservative-free, clear to slightly opalescent, colorless to slightly yellow solution, in a single- dose vial for intravenous infusion after dilution.
  • tremelimumab contains tremelimumab-actl at a concentration of 20 mg/mL in either a 25 mg/1.25 mL or a 300 mg/15 mL single-dose vial. Each mL contains 20 mg of tremelimumab-actl, and edetate disodium (0.09 mg), histidine (0.68 mg), L-histidine hydrochloride monohydrate (3.3 mg), polysorbate 80 (0.2 mg), trehalose (76 mg), and Water for Injection, USP. The pH is approximately 5.5.
  • the CTLA-4 blocking therapy is tremilumumab in combination with durvalumab, for the treatment of adult patients with unresectable hepatocellular carcinoma (uHCC).
  • the method comprising the administration of tremilumumab to a patient diagnosed with uHCC in an intravenous infusion over 60 minutes after dilution, as follows: 300 mg tremilumumab as a single dose to a patient weighing 30 kg or greater, in combination with durvalumab 1,500 mg at Cycle 1/Day 1, followed by durvalumab as a single agent every 4 weeks; or 4 mg/kg tremilumumab to a patient weighing less than 30 kg as a single dose in combination with durvalumab 20 mg/kg at Cycle 1/Day 1, followed by durvalumab as a single agent every 4 weeks.
  • the method comprising the administration of tremilumumab to a patient diagnosed with metastatic non-small cell lung cancer (NSCLC) with no sensitizing epidermal growth factor receptor (EGFR) mutation or anaplastic lymphoma kinase (ALK) genomic tumor aberrations.
  • NSCLC metastatic non-small cell lung cancer
  • EGFR epidermal growth factor receptor
  • ALK anaplastic lymphoma kinase
  • the method comprising the administration of tremilumumab to a patient diagnosed with NSCLC in an intravenous infusion over 60 minutes after dilution, as follows: 75 mg to a patient weighing 30 kg or greater every 3 weeks in combination with durvalumab 1,500 mg and platinum-based chemotherapy for 4 cycles, and then administer durvalumab 1,500 mg every 4 weeks as a single agent with histology-based pemetrexed therapy every 4 weeks, and a fifth dose of tremilumumab 75 mg in combination with durvalumab dose 6 at week 16; or 1 mg/kg to patient weighing less than 30 kg every 3 weeks in combination with durvalumab 20 mg/kg and platinum-based chemotherapy for 4 cycles, and then administer durvalumab 20 mg/kg every 4 weeks as a single agent with histology-based pemetrexed therapy every 4 weeks, and a fifth dose of tremilumumab 1 mg/kg in combination with durvalumab dose 6 at week 16.
  • the CTLA-4 blocking therapy is ipilimumab.
  • the CTLA-4 blocking therapy comprises the administration of ipilimumab in combination with the compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV).
  • Ipilimumab is a human cytotoxic T-lymphocyte antigen 4 (CTLA-4)-blocking antibody.
  • Ipilimumab is a recombinant IgG1 kappa immunoglobulin with an approximate molecular weight of 148 kDa.
  • Ipilimumab is produced in mammalian (Chinese hamster ovary) cell culture. Ipilimumab injection, for intravenous use is a sterile, preservative-free, clear to slightly opalescent, colorless to pale-yellow solution, which may contain a small amount of visible translucent-to-white, amorphous ipilimumab particulates. It is supplied in single-dose vials of 50 mg/10 mL or 200 mg/40 mL.
  • ipilimumab contains 5 mg of ipilimumab and the following inactive ingredients: diethylene triamine pentaacetic acid (DTPA) (0.04 mg), mannitol (10 mg), polysorbate 80 (vegetable origin) (0.1 mg), sodium chloride (5.85 mg), tris hydrochloride (3.15 mg), and Water for Injection, USP at a pH of 7.
  • DTPA diethylene triamine pentaacetic acid
  • mannitol (10 mg)
  • polysorbate 80 vegetable origin
  • sodium chloride 5.85 mg
  • tris hydrochloride 3.15 mg
  • Water for Injection USP at a pH of 7.
  • ipilumumab is administered by intravenous infusion after dilution based upon recommended infusion rate for each indication.
  • the ipilumumab can be provided as a50 mg/10 mL (5 mg/mL) or a 200 mg/40 mL (5 mg/mL) in a single-dose
  • a method of treating melanoma comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with a ipilumumab.
  • the treatment of melanoma is treating unresectable or metastatic melanoma in adults and pediatric patients.
  • a method of treating unresectable or metastatic melanoma in adults and pediatric patients 12 years and older comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab, either as a single agent or in combination with nivolumab.
  • the ipilumumab is administered for treating Unresectable or Metastatic Melanoma at an infusion dose of 3 mg/kg every 3 weeks for a maximum of 4 doses, or at a dose of 3 mg/kg immediately following nivolumab 1 mg/kg on the same day, every 3 weeks for 4 doses.
  • the nivolumab is administered as a single agent as recommended in the Full Prescribing Information for nivolumab.
  • the ipilumumab is administered as Adjuvant Treatment of Melanoma at a dose of 10 mg/kg every 3 weeks for 4 doses, followed by 10 mg/kg every 12 weeks for up to 3 years.
  • a method of adjuvant treatment of adult patients with cutaneous melanoma with pathologic involvement of regional lymph nodes of more than 1 mm who have undergone complete resection, including total lymphadenectomy comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab.
  • a method of treating Renal Cell Carcinoma comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab.
  • a method of treating adult patients with intermediate or poor risk advanced renal cell carcinoma, as first-line treatment in combination with nivolumab comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab.
  • the ipilumumab is administered for treating Advanced Renal Cell Carcinoma at a dose of 1 mg/kg immediately following nivolumab 3 mg/kg on the same day, every 3 weeks for 4 doses; after completing 4 doses of the combination, nivolumab is administered as a single agent as recommended in Full Prescribing Information for nivolumab.
  • a method of treating colorectal cancer comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab.
  • a method of treating adult and pediatric patients 12 years and older with microsatellite instability-high (MSI-H) or mismatch repair deficient (dMMR) metastatic colorectal cancer that has progressed following treatment with a fluoropyrimidine, oxaliplatin, and irinotecan, in combination with nivolumab comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab.
  • MSI-H microsatellite instability-high
  • a method of treating Microsatellite Instability-High (MSI-H) or Mismatch Repair Deficient (dMMR) Metastatic Colorectal Cancer comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab at a dose of 1 mg/kg intravenously over 30 minutes immediately following nivolumab 3 mg/kg intravenously over 30 minutes on the same day, every 3 weeks for 4 doses; after completing 4 doses of the combination, nivolumab is administered as a single agent as recommended in Full Prescribing Information for nivolumab.
  • a method of treating Hepatocellular Carcinoma comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab.
  • a method of treating adult patients with hepatocellular carcinoma who have been previously treated with sorafenib, in combination with nivolumab comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab.
  • a method of treating Hepatocellular Carcinoma comprises the administration of ipilumumab at a dise if 3 mg/kg intravenously over 30 minutes immediately following nivolumab 1 mg/kg intravenously over 30 minutes on the same day, every 3 weeks for 4 doses; after completing 4 doses of the combination, nivolumab is administered as a single agent as recommended in Full Prescribing Information for nivolumab.
  • a method of treating Non-Small Cell Lung Cancer comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab.
  • a method of treating adult patients with metastatic non-small cell lung cancer expressing PD-L1 ( ⁇ 1%) as determined by an FDA-approved test, with no EGFR or ALK genomic tumor aberrations, as first-line treatment in combination with nivolumab comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab.
  • a method of treating adult patients with metastatic or recurrent non-small cell lung cancer with no EGFR or ALK genomic tumor aberrations as first- line treatment, in combination with nivolumab and 2 cycles of platinum-doublet chemotherapy comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab.
  • a method of treating metastatic non-small cell lung cancer comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab at a dose of 1 mg/kg every 6 weeks with nivolumab 360 mg every 3 weeks.
  • a method of treating metastatic non-small cell lung cancer comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab at a dose of ipilumumab 1 mg/kg every 6 weeks with nivolumab 360 mg every 3 weeks and 2 cycles of platinum-doublet chemotherapy.
  • a method of treating Malignant Pleural Mesothelioma comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab.
  • a method of treating adult patients with unresectable malignant pleural mesothelioma, as first-line treatment in combination with nivolumab comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab.
  • a method of treating Malignant Pleural Mesothelioma comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab at a dise if 1 mg/kg every 6 weeks with nivolumab 360 mg every 3 weeks.
  • a method of treating Esophageal Cancer comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab.
  • a method of treating adult patients with unresectable advanced or metastatic esophageal squamous cell carcinoma, as first line treatment in combination with nivolumab comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab.
  • a method of treating esophageal squamous cell carcinoma comprises the administration of a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab at a dose of 1 mg/kg every 6 weeks with nivolumab 3 mg/kg every 2 weeks or 360 mg every 3 weeks.
  • a method of adjuvant treatment of adult patients with cutaneous melanoma with pathologic involvement of regional lymph nodes of more than 1 mm who have undergone complete resection, including total lymphadenectomy comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with ipilumumab.
  • a method of treating unresectable or metastatic melanoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with 3 mg/kg ipilimumab every 3 weeks for a maximum of 4 doses.
  • a method of treating unresectable or metastatic melanoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with 3 mg/kg ipilimumab immediately following nivolumab 1 mg/kg on the same day, every 3 weeks for 4 doses. After completing 4 doses of the combination, administer nivolumab as a single agent as recommended in the Full Prescribing Information for nivolumab.
  • the method comprises the treatment of a patient diagnosed with cancer by administration of a compound disclosed herein of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with a programmed death receptor-1 (PD 1)-blocking therapy.
  • PD 1 programmed death receptor-1
  • Binding of the PD-1 ligands, PD-L1 and PD-L2 to the PD-1 receptor found on T cells inhibits T cell proliferation and cytokine production. Upregulation of PD-1 ligands occurs in some tumors and signaling through this pathway can contribute to inhibition of active T-cell immune surveillance of tumors.
  • Pembrolizumab is a monoclonal antibody that binds to the PD-1 receptor and blocks its interaction with PD-L1 and PD-L2, releasing PD-1 pathway-mediated inhibition of the immune response, including the anti-tumor immune response.
  • blocking PD-1 activity resulted in decreased tumor growth.
  • combination treatment of a PD-1 blocking antibody and kinase inhibitor lenvatinib decreased tumor-associated macrophages, increased activated cytotoxic T cells, and reduced tumor growth compared to either treatment alone.
  • Nivolumab is a human immunoglobulin G4 (IgG4) monoclonal antibody that binds to the PD-1 receptor and blocks its interaction with PD-L1 and PD-L2, releasing PD-1 pathway-mediated inhibition of the immune response, including the anti-tumor immune response.
  • IgG4 human immunoglobulin G4
  • the PD-1 blocking therapy comprises pembrolizumab.
  • Pembrolizumab is a humanized monoclonal IgG4 kappa antibody with an approximate molecular weight of 149 kDa.
  • Pembrolizumab is produced in recombinant Chinese hamster ovary (CHO) cells.
  • Pembrolizumab injection is a sterile, preservative-free, clear to slightly opalescent, colorless to slightly yellow solution for intravenous use. Each vial contains 100 mg of pembrolizumab in 4 mL of solution.
  • Each 1 mL of solution contains 25 mg of pembrolizumab and is formulated in: L-histidine (1.55 mg), polysorbate 80 (0.2 mg), sucrose (70 mg), and Water for Injection, USP.
  • Pembrolizumab is administered as an intravenous infusion over 30 minutes after dilution.
  • a method of treating melanoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with pembrolizumab at a dose of 200 mg every 3 weeks or 400 mg every 6 weeks; 2 mg/kg (up to 200 mg) every 3 weeks for pediatrics.
  • a method of treating non-small cell lung cancer (NSCLC) comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with pembrolizumab, at a dose of 200 mg every 3 weeks or 400 mg every 6 weeks.
  • a method of treating Head and Neck Squamous Cell Cancer (HNSCC) comprises the administration
  • a method of treating Classical Hodgkin Lymphoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with pembrolizumab at a dose of 200 mg every 3 weeks or 400 mg every 6 weeks for adults; 2 mg/kg (up to 200 mg) every 3 weeks for pediatrics.
  • a method of treating Primary Mediastinal Large B-Cell Lymphoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with pembrolizumab at a dose of 200 mg every 3 weeks or 400 mg every 6 weeks for adults; 2 mg/kg (up to 200 mg) every 3 weeks for pediatrics.
  • a method of treating Urothelial Carcinoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with pembrolizumab, at a dose of 200 mg every 3 weeks or 400 mg every 6 weeks.
  • a method of treating Microsatellite Instability-High or Mismatch Repair Deficient Cancer comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with pembrolizumab at a dose of 200 mg every 3 weeks or 400 mg every 6 weeks for adults; 2 mg/kg (up to 200 mg) every 3 weeks for pediatrics.
  • a method of treating Microsatellite Instability- High or Mismatch Repair Deficient Colorectal Cancer comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with pembrolizumab at a dose of 200 mg every 3 weeks or 400 mg every 6 weeks.
  • a method of treating Gastric Cancer comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with pembrolizumab at a dose of 200 mg every 3 weeks or 400 mg every 6 weeks.
  • a method of treating Esophageal Cancer comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with pembrolizumab at a dose of 200 mg every 3 weeks or 400 mg every 6 weeks.
  • a method of treating Cervical Cancer comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with pembrolizumab at a dose of 200 mg every 3 weeks or 400 mg every 6 weeks.
  • a method of treating Hepatocellular Carcinoma (HCC) comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with pembrolizumab at a dose of 200 mg every 3 weeks or 400 mg every 6 weeks.
  • a method of treating Merkel Cell Carcinoma (MCC) comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (
  • a method of treating Renal Cell Carcinoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with pembrolizumab at a dose of 200 mg every 3 weeks or 400 mg every 6 weeks as a single agent in the adjuvant setting, or in the advanced setting with either: axitinib 5 mg orally twice daily or lenvatinib 20 mg orally once daily.
  • a method of treating Endometrial Carcinoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with pembrolizumab at a dose of 200 mg every 3 weeks or 400 mg every 6 weeks.
  • a method of treating Endometrial Carcinoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with pembrolizumab at a dose of 200 mg every 3 weeks or 400 mg every 6 weeks with lenvatinib 20 mg orally once daily.
  • a method of treating Tumor Mutational Burden-High (TMB-H) Cancer comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with pembrolizumab at a dose of 200 mg every 3 weeks or 400 mg every 6 weeks for adults; or 2 mg/kg (up to 200 mg) every 3 weeks for pediatrics.
  • a method of treating Cutaneous Squamous Cell Carcinoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with pembrolizumab at a dose of 200 mg every 3 weeks or 400 mg every 6 weeks.
  • a method of treating Triple- Negative Breast Cancer (TNBC) comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with pembrolizumab at a dose of 200 mg every 3 weeks or 400 mg every 6 weeks.
  • a method of treating Adult Classical Hodgkin Lymphoma and Adult Primary Mediastinal Large B-Cell Lymphoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with pembrolizumab.
  • the PD-1 blocking therapy comprises nivolumab.
  • Nivolumab is a programmed death receptor-1 (PD-1) blocking antibody.
  • Nivolumab is an IgG4 kappa immunoglobulin that has a calculated molecular mass of 146 kDa.
  • Nivolumab is a sterile, preservative- free, non-pyrogenic, clear to opalescent, colorless to pale-yellow liquid that may contain light (few) particles.
  • Nivolumab injection for intravenous use is supplied in single-dose vials. Each mL of nivolumab solution contains nivolumab 10 mg, mannitol (30 mg), pentetic acid (0.008 mg), polysorbate 80 (0.2 mg), sodium chloride (2.92 mg), sodium citrate dihydrate (5.88 mg), and Water for Injection, USP.
  • Nivolumab may contain hydrochloric acid and/or sodium hydroxide to adjust pH to 6.
  • Nivolumab for Injection can be provided as 40 mg/4 mL (10 mg/mL), 100 mg/10 mL (10 mg/mL), 120 mg/12 mL (10 mg/mL), and 240 mg/24 mL (10 mg/mL) solution in a single-dose vial.
  • nivolumab (anti-PD-1) and ipilimumab (anti-CTLA-4) mediated inhibition results in enhanced T-cell function that is greater than the effects of either antibody alone, and results in improved anti-tumor responses in metastatic melanoma and advanced RCC.
  • a method of treating melanoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with nivolumab.
  • a method of treating unresected or metastatic melanoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with nivolumab as follows: (a) for adult and pediatric patients weighing 40 kg or greater: 240 mg every 2 weeks or 480 mg every 4 weeks; (b) for pediatric patients weighing less than 40 kg: 3 mg/kg every 2 weeks or 6 mg/kg every 4 weeks; (c) for adult and pediatric patients weighing 40kg or greater:1mg/kg followed by ipilimumab 3 mg/kg on the same day every 3 weeks for 4 doses, then 240 mg every 2 weeks or 480 mg every 4 weeks; or (d) for pediatric patients weighing less than 40 kg: 1 mg/kg followed by ipilimumab 3 mg/kg on the same day every 3 weeks for 4 dose
  • a method of providing adjuvant treatment of melanoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with nivolumab as follows: (a) adult and pediatric patients weighing 40 kg or greater: 240 mg every 2 weeks or 480 mg every 4 weeks; or (b) for pediatric patients weighing less than 40 kg: 3 mg/kg every 2 weeks or 6 mg/kg every 4 weeks.
  • a method of providing neoadjuvant treatment of resectable (tumors ⁇ 4 cm or node positive) non- small cell lung cancer comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with nivolumab at a dose of 360 mg with platinum-doublet chemotherapy on the same day every 3 weeks for 3 cycles.
  • a method of treating metastatic non-small cell lung cancer comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with nivolumab as follows: (a) 360 mg every 3 weeks with ipilimumab 1 mg/kg every 6 weeks; (b) 360 mg every 3 weeks with ipilimumab 1 mg/kg every 6 weeks and 2 cycles of platinum-doublet chemotherapy; or (c) 240 mg every 2 weeks or 480 mg every 4 weeks.
  • nivolumab as follows: (a) 360 mg every 3 weeks with ipilimumab 1 mg/kg every 6 weeks; (b) 360 mg every 3 weeks with ipilimumab 1 mg/kg every 6 weeks and 2 cycles of platinum-doublet chemotherapy; or (c) 240 mg every 2 weeks or 480 mg every 4 weeks.
  • a method of treating malignant pleural mesothelioma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with nivolumab at a dose of 360 mg every 3 weeks with ipilimumab 1 mg/kg every 6 weeks.
  • a method of treating advanced renal cell carcinoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with nivolumab as follows: (a) 3 mg/kg followed by ipilimumab 1 mg/kg on the same day every 3 weeks for 4 doses, then 240 mg every 2 weeks or 480 mg every 4 weeks; (b) 240 mg every 2 weeks or 480 mg every 4 weeks administered in combination with cabozantinib 40 mg once daily without food; or (c) 240 mg every 2 weeks or 480 mg every 4 weeks.
  • a method of treating Classical Hodgkin lymphoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with nivolumab at a dose of 240 mg every 2 weeks or 480 mg every 4 weeks.
  • a method of treating Recurrent or metastatic squamous cell carcinoma of the head and neck comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with nivolumab at a dose of 240 mg every 2 weeks or 480 mg every 4 weeks.
  • a method of providing adjuvant treatment of urothelial carcinoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with nivolumab at a dose of 240 mg every 2 weeks or 480 mg every 4 weeks.
  • a method of treating locally advanced or metastatic urothelial carcinoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with nivolumab at a dose of 240 mg every 2 weeks or 480 mg every 4 weeks.
  • a method of treating microsatellite instability-high (MSI-H) or mismatch repair deficient (dMMR) metastatic colorectal cancer comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with nivolumab as follows: (a) adult and pediatric patients weighing 40 kg or greater: 240 mg every 2 weeks or 480 mg every 4 weeks; (b) pediatric patients weighing less than 40 kg: 3 mg/kg every 2 weeks; or (c) adult and pediatric patients weighing 40 kg or greater: 3 mg/kg followed by ipilimumab 1 mg/kg on the same day every 3 weeks for 4 doses, then 240 mg every 2 weeks or 480 mg every 4 weeks.
  • MSI-H microsatellite instability-high
  • dMMR mismatch repair deficient
  • a method of treating hepatocellular carcinoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with nivolumab at a dose of 1 mg/kg followed by ipilimumab 3 mg/kg on the same day every 3 weeks for 4 doses, then 240 mg every 2 weeks or 480 mg every 4 weeks.
  • a method of providing adjuvant treatment of resected esophageal or gastroesophageal cancer comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with nivolumab at a dose of 240 mg every 2 weeks or 480 mg every 4 weeks for total treatment duration of 1 year.
  • a method of treating esophageal squamous cell carcinoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with nivolumab as follows: (a) 240 mg every 2 weeks or 480 mg every 4 weeks in combination with chemotherapy regimen of fluoropyrimidine- and platinum-containing chemotherapy; (b) 3 mg/kg every 2 weeks or 360 mg every 3 weeks with ipilimumab 1 mg/kg every 6 weeks; or (c) 240 mg every 2 weeks or 480 mg every 4 weeks.
  • nivolumab as follows: (a) 240 mg every 2 weeks or 480 mg every 4 weeks in combination with chemotherapy regimen of fluoropyrimidine- and platinum-containing chemotherapy; (b) 3 mg/kg every 2 weeks or 360 mg every 3 weeks with ipilimumab 1 mg/kg every 6
  • a method of treating gastric cancer, gastroesophageal junction cancer, and esophageal adenocarcinoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with nivolumab as follows: (a) 360 mg every 3 weeks with fluoropyrimidine- and platinum-containing chemotherapy every 3 weeks; or (b) 240 mg every 2 weeks with fluoropyrimidine- and platinum-containing chemotherapy every 2 weeks.
  • the PD-1 blocking therapy comprises cemiplimab.
  • Cemiplimab is a human programmed death receptor-1 (PD-1) blocking antibody.
  • Cemiplimab is a recombinant human IgG4 monoclonal antibody that binds to PD-1 and blocks its interaction with PD-L1 and PD-L2.
  • Cemiplimab-rwlc is a recombinant human immunoglobulin G4 (IgG4) monoclonal antibody that binds to PD-1 and blocks its interaction with PD-L1 and PD-L2, releasing PD-1 pathway- mediated inhibition of the immune response, including the anti-tumor immune response.
  • IgG4 immunoglobulin G4
  • Cemiplimab is produced by recombinant DNA technology in Chinese hamster ovary (CHO) cell suspension culture. Cemiplimab-rwlc has an approximate molecular weight of 146 kDa.
  • a cemiplimab injection for intravenous use is a sterile, preservative-free, clear to slightly opalescent, colorless to pale yellow solution with a pH of 6. The solution may contain trace amounts of translucent to white particles.
  • Each vial contains 350 mg of cemiplimab.
  • cemiplimab 50 mg, L-histidine (0.74 mg), L-histidine monohydrochloride monohydrate (1.1 mg), sucrose (50 mg), L-proline (15 mg), Polysorbate 80 (2 mg), and Water for Injection, USP.
  • cemiplimab is cemiplimab-rwlc.
  • Cemiplimab for injection can be provided as 350 mg cemiplimab-rwlc/7 mL (50 mg/mL) solution in a single-dose vial.
  • a method of treating Cutaneous Squamous Cell Carcinoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with cemiplimab at a dose of 350 mg every 3 weeks until disease progression, unacceptable toxicity, or up to 24 months.
  • a method of treating patients with metastatic cutaneous squamous cell carcinoma (mCSCC) or locally advanced CSCC (laCSCC) who are not candidates for curative surgery or curative radiation comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with cemiplimab at a dose of 350 mg every 3 weeks until disease progression, unacceptable toxicity, or up to 24 months.
  • a method of treating Basal Cell Carcinoma comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with cemiplimab at a dose of 350 mg every 3 weeks until disease progression, unacceptable toxicity, or up to 24 months.
  • a method of treating patients with locally advanced or metastatic BCC comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with cemiplimab at a dose of 350 mg every 3 weeks until disease progression, unacceptable toxicity, or up to 24 months.
  • a method of treating non-small cell lung cancer comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with cemiplimab at a dose of 350 mg every 3 weeks until disease progression or unacceptable toxicity.
  • a method of treating non-small cell lung cancer comprises the administration of a compound of Formula (I), (Ia), (Ib), (Ic), (Id) (II), (IIa), (IIb), (IIc), (IId), (III), or (IV), in combination with cemiplimab as follows: (a) in combination with platinum ⁇ based chemotherapy for the first ⁇ line treatment of adult patients with non-small cell lung cancer (NSCLC) with no EGFR, ALK or ROS1 aberrations and is: (i) locally advanced where patients are not candidates for surgical resection or definitive chemoradiation or (ii) metastatic; or (b) as single agent for the first-line treatment of adult patients with NSCLC whose tumors have high PD-L1 expression [Tumor Proportion Score (TPS) ⁇ 50%] as determined by an FDA-approved test, with no EGFR, ALK or ROS1 aberrations, and is: (i) locally advanced where patients are
  • aspects disclosed herein relate to administering to a subject in need an effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof), or a pharmaceutical composition that includes one or more compounds as described herein (such as one or more compounds of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof).
  • some embodiments pertain to treating a disease or condition, such as cancer, through administration of a compound or composition as disclosed herein.
  • a subject in neeed of receiving a compound or composition as disclosed herein to improve the subject’s health need not always be identified prior to receiving a first treatment with the compound or composition.
  • a subject may be predetermined that they will develop a disease or condition, such as cancer, prior to showing any signs of the disease or condition.
  • the subject may receive treatment prophylactically if he or she is at risk or not developing a disease or condition, such as cancer, (e.g., once a patient shows symptoms of another disease or condition associated with a cancer).
  • the compound or composition may be adminsiterd to the subject after the subject receives an early stage diagnosis.
  • not every subject is a candidate for such administration and identification of treatment subjects may be desirable. It is understood that patient selection depends upon a number of factors within the skill of the ordinarily skilled physician.
  • some embodiments disclosed herein further comprise identifying a subject as one that will benefit from administering an effective amount of at least one compound or composition to increase longevity, increase survival time or increase life span.
  • the present disclosure is directed to a method for the treatment, prevention or prophylaxis of cancer can include administering to a subject in need thereof an effective amount of one or more compound described herein (such as a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof), or a pharmaceutical composition that includes a compound described herein (such as a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof).
  • a compound described herein such as a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or
  • the cancer may be selected from brain cancer, breast cancer, lung cancer, ovarian cancer, pancreatic cancer, stomach cancer, prostate cancer, renal cancer, colorectal cancer, melanoma, or leukemia.
  • the cancer is brain cancer or ach-enocortical carcinoma.
  • the cancer is breast cancer.
  • the cancer is ovarian cancer.
  • the cancer is pancreatic cancer.
  • the cancer is stomach cancer.
  • the cancer is renal cancer.
  • the cancer is colorectal cancer.
  • the cancer is myeloid leukemia. In further or additional embodiments, the cancer is glioblastoma. In further or additional embodiments, the cancer is follicular lymphona. In further or additional embodiments, the cancer is pre-B acute leukemia. In further or additional embodiments, the cancer is chronic lymphocytic B-leukemia. In further or additional embodiments, the cancer is mesothelioma. In further or additional embodiments, the cancer is small cell line cancer. In further or additional embodiments, the cancer is melanoma.
  • Some embodiments relate to a method of inhibiting proliferation of a cell having a RAS mutation, comprising administering a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof).
  • the cancer has associated with a RAS mutation.
  • Some embodiments relate to a method of inducing apoptosis in a cell having a RAS mutation, comprising administering a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof).
  • Some embodiments relate to a method of inhibiting proliferation of a cell having a KRAS mutation, comprising administering a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof).
  • the cancer has associated with a KRAS mutation.
  • Some embodiments relate to a method of inducing apoptosis in a cell having a KRAS mutation, comprising administering a compound of Formula (I)), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof).
  • Some embodiments relate to a method of inhibiting proliferation of a cell having a NRAS mutation, comprising administering a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof).
  • the cancer has associated with a NRAS mutation.
  • Some embodiments relate to a method of inducing apoptosis in a cell in a cell having a RAS mutation, comprising administering a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof).
  • the cancer has associated with a HRAS mutation.
  • Some embodiments relate to a method of inducing apoptosis in a cell having a RAS mutation, comprising administering a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof).
  • the KRAS mutation is at codons 12, 13, 59, 61 and/or 146.
  • the mutant form of the KRAS protein has one or more amino acid substitutions selected from the group consisting of G12C, G12S, G12R, G12F, G12L, G12N, G12A, G12D, G12V, G13C, G13S, G13D, G13V, G13P, S17G, P34S, A59E, A59G, A59T, Q61K, Q61L, Q61R, and Q61H.
  • the mutant form of the KRAS protein has one or more amino acid substitutions selected from the group consisting of G12C, G12R, G12S, G12A, G12D, G12V, G13C, G13R, G13S, G13A, G13D, G13V, A59E, A59G, A59T, Q61K, Q61L, Q61R, Q61H, K117N, K117R, K117E, A146P, A146T and A146V.
  • the cancer is resistant to treatment of a MEK protein kinase inhibitor.
  • the cancer is resistant to treatment of a RAF protein kinase inhibitor.
  • the resistance is acquired resistance. In other embodiments, the resistance is de novo resistance. In further or additional embodiments, the cancer is resistant to an anticancer agent.
  • a compounds or pharamceutical compositions and methods for treating cancer comprising a therapeutically effective amount of a dual-MEK protein kinase inhibitor.
  • the administration of the dual- MEK protein kinase inhibitor provides an increase in the area under the serum concentration time curve (AUC) of the dual-MEK protein kinase inhibitor.
  • the cancer is resistant to treatment of a RAF protein kinase inhibitor.
  • the cancer is resistant to a RAF protein kinase inhibitor and the RAF protein kinase inhibitor comprises an A-RAF inhibitor, a B-RAF inhibitor, or a C-RAF inhibitor. In further embodiments, the cancer is resistant to a RAF protein kinase inhibitor, and the RAF protein kinase inhibitor comprises a B-RAF inhibitor.
  • the resistant cancer is pancreatic, melanoma, colon, lung, or stomach cancer. In further embodiments, the resistant cancer is pancreatic. In additional embodiments, the resistant cancer is stomach.
  • a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof is co- administered with a CTLA-4 receptor inhibitor compound.
  • a compound of Formula (I) is co-administered a PD-1 or PD-L1 receptor inhibitor compound.
  • the method comprises treating a subject by co- administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and a LAG-3 receptor inhibitor compound.
  • the method comprises treating a subject by co-administering a therapeutically effective amount of a compound of Formula (I) and a TIM-3 receptor inhibitor compound.
  • the method comprises treating a subject by co-administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and a BTLA receptor inhibitor compound.
  • the method comprises treating a subject by co-administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and a KIR receptor inhibitor compound.
  • the method comprises treating a subject by co-administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and a PD-L1 inhibitor compound.
  • the method comprises treating a subject by co-administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and a PD- L2 inhibitor compound.
  • the method comprises treating a subject by co-administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and a blocking antibody of an immune checkpoint pathway.
  • the method comprises treating a subject by co-administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and an anti-CTLA-4 receptor antibody.
  • the method comprises treating a subject by co-administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and an anti-PD-1 receptor antibody.
  • the method comprises co-administering to a subject having a tumor a therapeutically effective amount of the compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and an anti-LAG-3 receptor antibody.
  • the method comprises co- administering to a subject having a tumor a therapeutically effective amount of the compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and an anti-TIM-3 receptor antibody.
  • the method comprises co-administering to a subject having a tumor a therapeutically effective amount of the compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and an anti-BTLA receptor antibody.
  • the method comprises co-administering to a subject having a tumor a therapeutically effective amount of the compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and an anti-KIR receptor antibody.
  • the anti-KIR receptor antibody is lirilumab.
  • the method comprises co-administering to a subject having a tumor a therapeutically effective amount of the compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and an anti-PD-1 antibody.
  • the anti-PD-1 antibody is lambrolizumab, pidilizumab, or nivolumab.
  • the method comprises co-administering to a subject having a tumor a therapeutically effective amount of the compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and an anti-PD-L1 antibody.
  • the method comprises co-administering to a subject having a tumor a therapeutically effective amount of the compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and an anti-PD-L2 antibody.
  • the method comprises co- administering to a subject having a tumor a therapeutically effective amount of the compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and an anti-CTLA-4 antibody.
  • the anti-CTLA- 4 antibody is ipilimumab or tremelimumab.
  • a method comprises co-administering a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and an immune checkpoint inhibitor for treating, preventing, or ameliorating a cancer or tumor in a subject by administering a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and an immune checkpoint inhibitor.
  • the subject was resistant to prior treatment with only an immune checkpoint inhibitor.
  • a method for treating a subject with a cancer or tumor comprises administering a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and a PD-1 inhibitor.
  • a method for treating a subject with a cancer or tumor comprises administering a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and a PD-L1 inhibitor.
  • a method for treating a subject with a cancer or tumor comprises administering a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and a PD-L2 inhibitor.
  • a method for treating a subject with a cancer or tumor comprises administering a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and a CTLA-4 inhibitor.
  • a method for treating a subject with a cancer or tumor comprises administering a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, a PD-1 inhibitor and a CTLA-4 inhibitor.
  • a method for treating a subject with a cancer or tumor comprises administering a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and a LAG-3 inhibitor.
  • a method for treating a subject with a cancer or tumor comprises administering a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and a KIR inhibitor.
  • a method for treating a subject with a cancer or tumor comprises administering a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and a TIM-3 inhibitor.
  • a method for treating a subject with a cancer or tumor comprises administering a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, and a BTLA inhibitor.
  • a method for treating a subject comprises treating a subject having exhibited resistance to a PD-1 inhibitor by administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof.
  • a method comprises treating a subject having exhibited resistance to a PD-L1 inhibitor by administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof.
  • a method comprises treating a subject having exhibited resistance to a PD-L2 inhibitor by administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof.
  • a method comprises treating a subject having exhibited resistance to a CTLA-4 inhibitor by administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof.
  • the method includes administering the compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof when the subject has exhibited resistance to two different immune checkpoint inhibitors.
  • a method comprises treating a subject having exhibited resistance to a PD-1 inhibitor and CTLA-4 inhibitor by administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof.
  • a method comprises treating a subject having exhibited resistance to a PD-L1 inhibitor and a CTLA-4 inhibitor by administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof.
  • a method comprises treating a subject having exhibited resistance to a PD- 1 inhibitor, a PD-L1 inhibitor, and a CTLA-4 inhibitor by administering a therapeutically effective amount of a compound of Formula (I) (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof.
  • Some embodiments disclosed herein relate to a method of treating a mammal having a disease that can include administering to a subject in need thereof an effective amount of one or more compound described herein (such as a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof), or a pharmaceutical composition that includes a compound described herein (such as a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof).
  • a compound described herein such as a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV),
  • inventions disclosed herein relate to a method of treating a subject with cancer cachexia that can include administering to a subject an effective amount of one or more compounds described herein (such as a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof), or a pharmaceutically acceptable salt of any of the foregoing), or a pharmaceutical composition that includes a compound described herein such as a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof).
  • compounds described herein such as a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId),
  • Some embodiments described herein relate to using one or more compounds described herein (such as a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof), in the manufacture of a medicament for ameliorating and/or treating cancer or conditions of cancer, such as cancer cachexia, that can include administering to a subject an effective amount of one or more compounds described herein (such as a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (Iib), (Iic), (Iid), (III), (IV), or a pharmaceutically acceptable salt thereof).
  • Still other embodiments described herein relate to one or more compounds described herein (such as a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof) that can be used for ameliorating and/or treating cancer or conditions of cancer, such as cancer cachexia, by administering to a subject an effective amount of one or more compounds described herein, or a pharmaceutically acceptable salt thereof.
  • compounds described herein such as a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof.
  • Some embodiments disclosed herein relate to methods of ameliorating and/or treating cancer that can include contacting a cancerous cell an effective amount of one or more compounds described herein (such as a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (Iib), (Iic), (Iid), (III), (IV), or a pharmaceutically acceptable salt thereof), or a pharmaceutical composition that includes one or more compounds described herein (such as a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof).
  • compounds described herein such as a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof).
  • a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof can act as an inhibitor of MEK.
  • a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof can act as an inhibitor of ERK.
  • a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (Iib), (Iic), (Iid), (III), (IV), or a pharmaceutically acceptable salt thereof may act as a STAT3 (pSER-727) inhibitor.
  • a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof may reduce inflammatory cachexia and muscle wasting.
  • the compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof may be administered in a single dose, once daily.
  • the compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof may be administered in multiple doses, more than once per day.
  • the compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof may be administered once a day.
  • the compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof may be administered administered administered twice a day.
  • the compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof may be administered administered trice a day.
  • the compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof may be administered administered four times a day.
  • a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof may inhibit abnormal cell growth.
  • the abnormal cell growth occurs in a mammal.
  • Methods for inhibiting abnormal cell growth may comprise administering an effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (Iib), (Iic), (Iid), (III), (IV), or a pharmaceutically acceptable salt thereof, wherein abnormal cell growth is inhibited.
  • Methods for inhibiting abnormal cell growth in a mammal may comprise administering to the mammal a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, wherein the amounts of the compound is effective in inhibiting abnormal cell growth in the mammal.
  • the present invention is directed to a method for degrading, inhibiting the growth of or killing a cancer cell comprising contacting said cell with a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, effective to degrade, inhibit the growth of or to kill said cell.
  • the cancer cells comprise brain, breast, lung, ovarian, pancreatic, stomach, prostate, renal, melanoma or colorectal cancer cells.
  • the cancer cells are degraded.
  • 1% of the cancer cells are degraded. In further or additional embodiments, 2% of the cancer cells are degraded. In further or additional embodiments, 3% of the cancer cells are degraded. In further or additional embodiments, 4% of the cancer cells are degraded. In further or additional embodiments, 5% of the cancer cells are degraded. In further or additional embodiments, 10% of the cancer cells are degraded. In further or additional embodiments, 20% of the cancer cells are degraded. In further or additional embodiments, 25% of the cancer cells are degraded. In further or additional embodiments, 30% of the cancer cells are degraded. In further or additional embodiments, 40% of the cancer cells are degraded. In further or additional embodiments, 50% of the cancer cells are degraded.
  • the cancer cells are killed. In further or additional embodiments, 1% of the cancer cells are killed. In further or additional embodiments, 2% of the cancer cells are killed. In further or additional embodiments, 3% of the cancer cells are killed.
  • 4% of the cancer cells are killed. In further or additional embodiments, 5% of the cancer cells are killed. In further or additional embodiments, 1.0% of the cancer cells are killed. In further or additional embodiments, 20% of the cancer cells are killed. In further or additional embodiments, 25% of the cancer cells are killed. In further or additional embodiments, 30% of the cancer cells are killed. In additional embodiments, 40% of the cancer cells are killed. In further or additional embodiments, 50% of the cancer cells are killed. In further or additional embodiments, 60% of the cancer cells are killed. In further or additional embodiments, 70% of the cancer cells are killed. In further or additional embodiments, 75% of the cancer cells are killed. In further or additional embodiments, 80% of the cancer cells are killed.
  • the growth of the cancer cells is inhibited. In further or additional embodiments, the growth of the cancer cells is about 1% inhibited. In further or additional embodiments, the growth of the cancer cells is about 2% inhibited. In further or additional embodiments, the growth of the cancer cells is about 3% inhibited. In further or additional embodiments, the growth of the cancer cells is about 4% inhibited. In further or additional embodiments, the growth of the cancer cells is about 5% inhibited.
  • the growth of the cancer cells is about 10% inhibited. In further or additional embodiments, the growth of the cancer cells is about 20% inhibited. In further or additional embodiments, the growth of the cancer cells is about 25% inhibited. In further or additional embodiments, the growth of the cancer cells is about 30% inhibited, hi further or additional embodiments, the growth of the cancer cells is about 40% inhibited. In further or additional embodiments, the growth of the cancer cells is about 50% inhibited. In further or additional embodiments, the growth of the cancer cells is about 60% inhibited. In further or additional embodiments, the growth of the cancer cells is about 70% inhibited. In further or additional embodiments, the growth of the cancer cells is about 75% inhibited.
  • the growth of the cancer cells is about 80% inhibited. In further or additional embodiments, the growth of the cancer cells is about 90% inhibited. In further or additional embodiments, the growth of the cancer cells is about 100% inhibited. [0284] In some embodiments, the size of a tumor is reduced by administering a therapeutically effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof. In further or additional embodiments, the size of a tumor is reduced by at least 1%.
  • the size of a tumor is reduced by at least 2%. In further or additional embodiments, the size of a tumor is reduced by at least 3%. In further or additional embodiments, the size of a tumor is reduced by at least 4%. In further or additional embodiments, the size of a tumor is reduced by at least 5%. In further or additional embodiments, the size of a tumor is reduced by at least 10%. In further or additional embodiments, the size of a tumor is reduced by at least 20%. In further or additional embodiments, the size of a tumor is reduced by at least 25%. In further or additional embodiments, the size of a tumor is reduced by at least 30%. In further or additional embodiments, the size of a tumor is reduced by at least 40%.
  • the size of a tumor is reduced by at least 50%. In further or additional embodiments, the size of a tumor is reduced by at least 60%. In further or additional embodiments, the size of a tumor is reduced by at least 70%. In further or additional embodiments, the size of a tumor is reduced by at least 75%. In further or additional embodiments, the size of a tumor is reduced by at least 80%. In further or additional embodiments, the size of a tumor is reduced by at least 85%. In further or additional embodiments, the size of a tumor is reduced by at least 90%. In further or additional embodiments, the size of a tumor is reduced by at least 95%. In further or additional embodiments, the tumor is eradicated. In some embodiments, the size of a tumor does not increase.
  • tumor proliferation is reduced by administering a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof.
  • tumor proliferation is reduced by at least 1 %.
  • tumor proliferation is reduced by at least 2 %.
  • tumor proliferation is reduced by at least 3 %.
  • tumor proliferation is reduced by at least 4 %.
  • tumor proliferation is reduced by at least 5 %.
  • tumor proliferation is reduced by at least 10 %.
  • tumor proliferation is reduced by at least 20 %.
  • tumor proliferation is reduced by at least 25 %. In some embodiments, tumor proliferation is reduced by at least 30 %. In some embodiments, tumor proliferation is reduced by at least 40 %. In some embodiments, tumor proliferation is reduced by at least 50 %. In some embodiments, tumor proliferation is reduced by at least 60 %. In some embodiments, tumor proliferation is reduced by at least 70 %. In some embodiments, tumor proliferation is reduced by at least 75 %. In some embodiments, tumor proliferation is reduced by at least 75 %. In some embodiments, tumor proliferation is reduced by at least 80 %. In some embodiments, tumor proliferation is reduced by at least 90 %. In some embodiments, tumor proliferation is reduced by at least 95 %. In some embodiments, tumor proliferation is prevented.
  • the compounds or pharmaceutical compositions may be administered to the patient by any suitable means.
  • methods of administration include, among others, (a) administration though oral pathways, which administration includes administration in capsule, tablet, granule, spray, syrup, or other such forms; (b) administration through non-oral pathways such as rectal, vaginal, intraurethral, intraocular, intranasal, or intraauricular, which administration includes administration as an aqueous suspension, an oily preparation or the like or as a drip, spray, suppository, salve, ointment or the like; (c) administration via injection, subcutaneously, intraperitoneally, intravenously, intramuscularly, intradermally, intraorbitally, intracapsularly, intraspinally, intrasternally, or the like, including infusion pump delivery; (d) administration locally such as by injection directly in the renal or cardiac area, e.g., by depot implantation; as well as administration topically; as deemed appropriate by those of skill in the
  • compositions suitable for administration include compositions where the active ingredients are contained in an amount effective to achieve its intended purpose.
  • the therapeutically effective amount of the compounds disclosed herein required as a dose will depend on the route of administration, the type of animal, including human, being treated, and the physical characteristics of the specific animal under consideration. The dose can be tailored to achieve a desired effect, but will depend on such factors as weight, diet, concurrent medication, and other factors which those skilled in the medical arts will recognize. More specifically, a therapeutically effective amount means an amount of compound effective to prevent, alleviate or ameliorate symptoms of disease or prolong the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • the useful in vivo dosage to be administered and the particular mode of administration will vary depending upon the age, weight and mammalian species treated, the particular compounds employed, and the specific use for which these compounds are employed.
  • the determination of effective dosage levels can be accomplished by one skilled in the art using routine pharmacological methods. Typically, human clinical applications of products are commenced at lower dosage levels, with dosage level being increased until the desired effect is achieved. Alternatively, acceptable in vitro studies can be used to establish useful doses and routes of administration of the compositions identified by the present methods using established pharmacological methods.
  • dosages may range broadly, depending upon the desired effects and the therapeutic indication. Typically, dosages may be between about 1 microgram/kg and 200 mg/kg body weight, preferably between about 180 microgram/kg and 10 mg/kg body weight. Alternatively, dosages may be based and calculated upon the surface area of the patient, as understood by those of skill in the art. [0290] The exact formulation, route of administration and dosage for the pharmaceutical compositions of the present invention can be chosen by the individual physician in view of the patient’s condition. (See e.g., Fingl et al.
  • the dose range of the composition administered to the patient can be from about 0.5 to 1000 mg/kg of the patient’s body weight.
  • the dosage may be a single one or a series of two or more given in the course of one or more days, as is needed by the patient.
  • the present invention will use those same dosages, or dosages that are between about 0.1% and 500%, more preferably between about 25% and 250% of the established human dosage.
  • a suitable human dosage can be inferred from ED50 or TD50 values, or other appropriate values derived from in vitro or in vivo studies, as qualified by toxicity studies and efficacy studies in animals.
  • the attending physician would know how to and when to terminate, interrupt, or adjust administration due to toxicity or organ dysfunctions. Conversely, the attending physician would also know to adjust treatment to higher levels if the clinical response were not adequate (precluding toxicity).
  • the magnitude of an administrated dose in the management of the disorder of interest will vary with the severity of the condition to be treated and to the route of administration.
  • the severity of the condition may, for example, be evaluated, in part, by standard prognostic evaluation methods. Further, the dose and perhaps dose frequency, will also vary according to the age, body weight, and response of the individual patient. A program comparable to that discussed above may be used in veterinary medicine. [0292] Although the exact dosage will be determined on a drug-by-drug basis, in most cases, some generalizations regarding the dosage can be made.
  • the daily dosage regimen for an adult human patient may be, for example, an oral dose of between 0.1 mg and 2000 mg of each active ingredient, preferably between 1 mg and 500 mg, e.g. 5 to 200 mg.
  • an intravenous, subcutaneous, or intramuscular dose of each active ingredient of between 0.01 mg and 100 mg, preferably between 0.1 mg and 60 mg, e.g. 1 to 40 mg is used.
  • dosages may be calculated as the free base.
  • the composition is administered 1 to 4 times per day.
  • the compositions of the invention may be administered by continuous intravenous infusion, preferably at a dose of each active ingredient up to 1000 mg per day.
  • the compounds will be administered for a period of continuous therapy, for example for a week or more, or for months or years.
  • the amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof may be administered in a range from about 0.001 to about 1000 mg/kg body weight/day.
  • the amount a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof may be administered the range of about 0.5 to about 50 mg/kg/day. In further or additional embodiments, the amount a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (Iib), (Iic), (Iid), (III), (IV), or a pharmaceutically acceptable salt thereof, may be administered from about 0.001 to about 7 g/day.
  • the amount a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof may be administered from about 0.002 to about 6 g/day. In further or additional embodiments, the amount a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, may be administered from about 0.005 to about 5 g/day.
  • the amount a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof may be administered from about 0.01 to about 5 g/day. In further or additional embodiments, the amount a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, may be administered from about 0.02 to about 5 g/day.
  • the amount a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof may be administered from about 0.05 to about 2.5 g/day. In further or additional embodiments, the amount a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, may be administered from about 0.1 to about 1 g/day.
  • dosage levels below the lower limit of the aforesaid range may be more than adequate.
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the modulating effects, or minimal effective concentration (MEC).
  • MEC minimal effective concentration
  • the MEC will vary for each compound but can be estimated from in vitro data. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations.
  • Dosage intervals can also be determined using MEC value. Compositions should be administered using a regimen which maintains plasma levels above the MEC for 10- 90% of the time, preferably between 30-90% and most preferably between 50-90%.
  • the effective local concentration of the drug may not be related to plasma concentration.
  • the amount of composition administered may be dependent on the subject being treated, on the subject’s weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician.
  • Compounds disclosed herein can be evaluated for efficacy and toxicity using known methods. For example, the toxicology of a particular compound, or of a subset of the compounds, sharing certain chemical moieties, may be established by determining in vitro toxicity towards a cell line, such as a mammalian, and preferably human, cell line. The results of such studies are often predictive of toxicity in animals, such as mammals, or more specifically, humans.
  • the toxicity of particular compounds in an animal model may be determined using known methods.
  • the efficacy of a particular compound may be established using several recognized methods, such as in vitro methods, animal models, or human clinical trials. Recognized in vitro models exist for nearly every class of condition, including but not limited to cancer, cardiovascular disease, and various immune dysfunction.
  • acceptable animal models may be used to establish efficacy of chemicals to treat such conditions.
  • selecting a model to determine efficacy the skilled artisan can be guided by the state of the art to choose an appropriate model, dose, and route of administration, and regime.
  • human clinical trials can also be used to determine the efficacy of a compound in humans.
  • compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient.
  • the pack may for example comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • the pack or dispenser may also be accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration.
  • Such notice for example, may be the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert.
  • compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
  • Administration and Pharmaceutical Compositions [0300] The compounds are administered at a therapeutically effective dosage. While human dosage levels have yet to be specifically identified for the compounds described herein, generally, a daily dose may be from about 0.25 mg/kg to about 120 mg/kg or more of body weight, from about 0.5 mg/kg or less to about 70 mg/kg, from about 1.0 mg/kg to about 50 mg/kg of body weight, or from about 1.5 mg/kg to about 10 mg/kg of body weight.
  • the dosage range would be from about 17 mg per day to about 8000 mg per day, from about 35 mg per day or less to about 7000 mg per day or more, from about 70 mg per day to about 6000 mg per day, from about 100 mg per day to about 5000 mg per day, or from about 200 mg to about 3000 mg per day.
  • the amount of active compound administered will, of course, be dependent on the subject and disease state being treated, the severity of the affliction, the manner and schedule of administration and the judgment of the prescribing physician.
  • Administration of the compounds disclosed herein, or the pharmaceutically acceptable salts thereof can be via any of the accepted modes of administration for agents that serve similar utilities including, but not limited to, orally, subcutaneously, intravenously, intranasally, topically, transdermally, intraperitoneally, intramuscularly, intrapulmonarilly, vaginally, rectally, or intraocularly. Oral and parenteral administrations are customary in treating the indications that are the subject of the preferred embodiments. [0302] The compounds useful as described above can be formulated into pharmaceutical compositions for use in treatment of these conditions.
  • compositions comprising: (a) a safe and therapeutically effective amount of a compound described herein (including enantiomers, diastereoisomers, tautomers, polymorphs, and solvates thereof), or pharmaceutically acceptable salts thereof; and (b) a pharmaceutically acceptable carrier, diluent, excipient or combination thereof.
  • a pharmaceutically-acceptable carrier for example, a pharmaceutically-acceptable carrier.
  • pharmaceutically acceptable carrier or “pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated.
  • various adjuvants such as are commonly used in the art may be included. Considerations for the inclusion of various components in pharmaceutical compositions are described, e.g., in Gilman et al.
  • substances which can serve as pharmaceutically- acceptable carriers or components thereof, are sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and methyl cellulose; powdered tragacanth; malt; gelatin; talc; solid lubricants, such as stearic acid and magnesium stearate; calcium sulfate; vegetable oils, such as peanut oil, cottonseed oil, sesame oil, olive oil, corn oil and oil of theobroma; polyols such as propylene glycol, glycerine, sorbitol, mannitol, and polyethylene glycol; alginic acid; emulsifiers
  • compositions described herein are preferably provided in unit dosage form.
  • a "unit dosage form" is a composition containing an amount of a compound that is suitable for administration to an animal, preferably mammal subject, in a single dose, according to good medical practice. The preparation of a single or unit dosage form however, does not imply that the dosage form is administered once per day or once per course of therapy.
  • Such dosage forms are contemplated to be administered once, twice, thrice or more per day and may be administered as infusion over a period of time (e.g., from about 30 minutes to about 2-6 hours), or administered as a continuous infusion, and may be given more than once during a course of therapy, though a single administration is not specifically excluded.
  • a single administration is not specifically excluded.
  • the skilled artisan will recognize that the formulation does not specifically contemplate the entire course of therapy and such decisions are left for those skilled in the art of treatment rather than formulation.
  • compositions useful as described above may be in any of a variety of suitable forms for a variety of routes for administration, for example, for oral, nasal, rectal, topical (including transdermal), ocular, intracerebral, intracranial, intrathecal, intra-arterial, intravenous, intramuscular, or other parental routes of administration.
  • routes for administration for example, for oral, nasal, rectal, topical (including transdermal), ocular, intracerebral, intracranial, intrathecal, intra-arterial, intravenous, intramuscular, or other parental routes of administration.
  • oral and nasal compositions comprise compositions that are administered by inhalation, and made using available methodologies.
  • a variety of pharmaceutically-acceptable carriers well-known in the art may be used.
  • Pharmaceutically-acceptable carriers include, for example, solid or liquid fillers, diluents, hydrotropies, surface-active agents, and encapsulating substances.
  • Optional pharmaceutically-active materials may be included, which do not substantially interfere with the inhibitory activity of the compound.
  • the amount of carrier employed in conjunction with the compound is sufficient to provide a practical quantity of material for administration per unit dose of the compound.
  • Techniques and compositions for making dosage forms useful in the methods described herein are described in the following references, all incorporated by reference herein: Modern Pharmaceutics, 4th Ed., Chapters 9 and 10 (Banker & Rhodes, editors, 2002); Lieberman et al., Pharmaceutical Dosage Forms: Tablets (1989); and Ansel, Introduction to Pharmaceutical Dosage Forms 8th Edition (2004).
  • Various oral dosage forms can be used, including such solid forms as tablets, capsules, granules and bulk powders.
  • Tablets can be compressed, tablet triturates, enteric-coated, sugar-coated, film-coated, or multiple-compressed, containing suitable binders, lubricants, diluents, disintegrating agents, coloring agents, flavoring agents, flow-inducing agents, and melting agents.
  • Liquid oral dosage forms include aqueous solutions, emulsions, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules, and effervescent preparations reconstituted from effervescent granules, containing suitable solvents, preservatives, emulsifying agents, suspending agents, diluents, sweeteners, melting agents, coloring agents and flavoring agents.
  • the pharmaceutically-acceptable carrier suitable for the preparation of unit dosage forms for peroral administration is well-known in the art.
  • Tablets typically comprise conventional pharmaceutically-compatible adjuvants as inert diluents, such as calcium carbonate, sodium carbonate, mannitol, lactose and cellulose; binders such as starch, gelatin and sucrose; disintegrants such as starch, alginic acid and croscarmelose; lubricants such as magnesium stearate, stearic acid and talc.
  • Glidants such as silicon dioxide can be used to improve flow characteristics of the powder mixture.
  • Coloring agents such as the FD&C dyes, can be added for appearance.
  • Sweeteners and flavoring agents such as aspartame, saccharin, menthol, peppermint, and fruit flavors, are useful adjuvants for chewable tablets.
  • Capsules typically comprise one or more solid diluents disclosed above.
  • the selection of carrier components depends on secondary considerations like taste, cost, and shelf stability, which are not critical, and can be readily made by a person skilled in the art.
  • Peroral compositions also include liquid solutions, emulsions, suspensions, and the like.
  • the pharmaceutically-acceptable carriers suitable for preparation of such compositions are well known in the art.
  • Typical components of carriers for syrups, elixirs, emulsions and suspensions include ethanol, glycerol, propylene glycol, polyethylene glycol, liquid sucrose, sorbitol and water.
  • typical suspending agents include methyl cellulose, sodium carboxymethyl cellulose, AVICEL RC-591, tragacanth and sodium alginate;
  • typical wetting agents include lecithin and polysorbate 80;
  • typical preservatives include methyl paraben and sodium benzoate.
  • Peroral liquid compositions may also contain one or more components such as sweeteners, flavoring agents and colorants disclosed above.
  • compositions may also be coated by conventional methods, typically with pH or time-dependent coatings, such that the subject compound is released in the gastrointestinal tract in the vicinity of the desired topical application, or at various times to extend the desired action.
  • dosage forms typically include, but are not limited to, one or more of cellulose acetate phthalate, polyvinylacetate phthalate, hydroxypropyl methyl cellulose phthalate, ethyl cellulose, Eudragit coatings, waxes and shellac.
  • Compositions described herein may optionally include other drug actives.
  • Other compositions useful for attaining systemic delivery of the subject compounds include sublingual, buccal and nasal dosage forms.
  • compositions typically comprise one or more of soluble filler substances such as sucrose, sorbitol and mannitol; and binders such as acacia, microcrystalline cellulose, carboxymethyl cellulose and hydroxypropyl methyl cellulose. Glidants, lubricants, sweeteners, colorants, antioxidants and flavoring agents disclosed above may also be included.
  • a liquid composition which is formulated for topical ophthalmic use, is formulated such that it can be administered topically to the eye. The comfort should be maximized as much as possible, although sometimes formulation considerations (e.g. drug stability) may necessitate less than optimal comfort.
  • the liquid should be formulated such that the liquid is tolerable to the patient for topical ophthalmic use. Additionally, an ophthalmically acceptable liquid should either be packaged for single use, or contain a preservative to prevent contamination over multiple uses.
  • solutions or medicaments are often prepared using a physiological saline solution as a major vehicle. Ophthalmic solutions should preferably be maintained at a comfortable pH with an appropriate buffer system.
  • the formulations may also contain conventional, pharmaceutically acceptable preservatives, stabilizers and surfactants.
  • Preservatives that may be used in the pharmaceutical compositions disclosed herein include, but are not limited to, benzalkonium chloride, PHMB, chlorobutanol, thimerosal, phenylmercuric, acetate and phenylmercuric nitrate.
  • a useful surfactant is, for example, Tween 80.
  • various useful vehicles may be used in the ophthalmic preparations disclosed herein. These vehicles include, but are not limited to, polyvinyl alcohol, povidone, hydroxypropyl methyl cellulose, poloxamers, carboxymethyl cellulose, hydroxyethyl cellulose and purified water.
  • Tonicity adjustors may be added as needed or convenient.
  • buffers include, but are not limited to, salts, particularly sodium chloride, potassium chloride, mannitol and glycerin, or any other suitable ophthalmically acceptable tonicity adjustor.
  • Various buffers and means for adjusting pH may be used so long as the resulting preparation is ophthalmically acceptable.
  • the pH will be between 4 and 9.
  • buffers include acetate buffers, citrate buffers, phosphate buffers and borate buffers. Acids or bases may be used to adjust the pH of these formulations as needed.
  • an ophthalmically acceptable antioxidant includes, but is not limited to, sodium metabisulfite, sodium thiosulfate, acetylcysteine, butylated hydroxyanisole and butylated hydroxytoluene.
  • Other excipient components which may be included in the ophthalmic preparations, are chelating agents.
  • a useful chelating agent is edetate disodium, although other chelating agents may also be used in place or in conjunction with it.
  • creams, ointments, gels, solutions or suspensions, etc., containing the compound disclosed herein are employed.
  • Topical formulations may generally be comprised of a pharmaceutical carrier, co-solvent, emulsifier, penetration enhancer, preservative system, and emollient.
  • a pharmaceutically acceptable diluent such as a saline or dextrose solution.
  • Suitable excipients may be included to achieve the desired pH, including but not limited to NaOH, sodium carbonate, sodium acetate, HCl, and citric acid.
  • the pH of the final composition ranges from 2 to 8, or preferably from 4 to 7.
  • Antioxidant excipients may include sodium bisulfite, acetone sodium bisulfite, sodium formaldehyde, sulfoxylate, thiourea, and EDTA.
  • Other non-limiting examples of suitable excipients found in the final intravenous composition may include sodium or potassium phosphates, citric acid, tartaric acid, gelatin, and carbohydrates such as dextrose, mannitol, and dextran.
  • Antimicrobial agents may also be included to achieve a bacteriostatic or fungistatic solution, including but not limited to phenylmercuric nitrate, thimerosal, benzethonium chloride, benzalkonium chloride, phenol, cresol, and chlorobutanol.
  • compositions for intravenous administration may be provided to caregivers in the form of one more solids that are reconstituted with a suitable diluent such as sterile water, saline or dextrose in water shortly prior to administration.
  • a suitable diluent such as sterile water, saline or dextrose in water shortly prior to administration.
  • the compositions are provided in solution ready to administer parenterally.
  • the compositions are provided in a solution that is further diluted prior to administration.
  • the combination may be provided to caregivers as a mixture, or the caregivers may mix the two agents prior to administration, or the two agents may be administered separately.
  • the second therapeutic agent is anti-inflammatory agent.
  • the second therapeutic agent is a non-steroidal anti-inflammatory agent.
  • the second therapeutic agent is anti-cancer agent.
  • the methods comprise administering an effective amount of a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof, in combination with an amount of a chemotherapeutic, wherein the amounts of the combination and the chemotherapeutic are together effective in inhibiting abnormal cell growth.
  • chemotherapeutics are presently known in the art and can be used in combination.
  • the chemotherapeutic is selected from the group consisting of mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, anti-hormones, angiogenesis inhibitors, and anti-androgens.
  • Also described are methods for inhibiting abnormal cell growth in a mammal comprising administering to the mammal an amount of a MEK protein kinase inhibitor and/or Raf protein kinase inhibitor in combination with radiation therapy, wherein the amounts of the MEK protein kinase inhibitor and/or Raf protein kinase inhibitor in combination with the radiation therapy effective in inhibiting abnormal cell growth or treating the hyperproliferative disorder in the mammal.
  • Techniques for administering radiation therapy are known in the art, and these techniques can be used in the combination therapy described herein.
  • the disclosure also relates to a method of inhibiting abnormal cell growth in a mammal which may comprises a compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), (IV), or a pharmaceutically acceptable salt thereof, and an amount of one or more substances selected from anti-angiogenesis agents, signal transduction inhibitors, and antiproliferative agents.
  • a mammal which may comprises a compound of Formula (I), (Ia), (Ib), (Ic), (II), (IIa), (IIb), (IIc), (III), (IV), or a pharmaceutically acceptable salt thereof, and an amount of one or more substances selected from anti-angiogenesis agents, signal transduction inhibitors, and antiproliferative agents.
  • Anti-angiogenesis agents such as MMP- 2 (matrix-metalloprotienase 2) inhibitors, MMP-9 (matrix-metalloprotienase 9) inhibitors, and COX-11 (cyclooxygenase 11) inhibitors, can be used in conjunction with a compound of the present invention and pharmaceutical compositions described herein.
  • MMP- 2 matrix-metalloprotienase 2
  • MMP-9 matrix-metalloprotienase 9 inhibitors
  • COX-11 cyclooxygenase 11
  • useful COX-II inhibitors include CELEBREXTM (alecoxib), valdecoxib, and rofecoxib.
  • Examples of useful matrix metalloproteinase inhibitors are described in WO 96/33172 (published October 24,1996), WO 96/27583 (published March 7,1996), European Patent Application No. 97304971.1 (filed July 8,1997), European Patent Application No.
  • MMP-2 and MMP-9 inhibitors have little or no activity inhibiting MMP-1, while some selectively inhibit MMP-2 and/or AMP-9 relative to the other matrix-motalloproteinases (L e., MAP-1, NEMP-3, MMP-4, M7v1P-5, MMP-6, MMP- 7, MMP-8, MMP-10, MMP-11, and MMP-13).
  • M1v1P inhibitors useful in the present invention are AG-3340, RU 32-3555, and RS 13-0830.
  • a compound of Formula (I), (Ia), (Ib), (Ic), (Id), (II), (IIa), (IIb), (IIc), (IId), (III), (IV), or a pharmaceutically acceptable salt thereof is administered with at least one additional therapeutic agent.
  • the therapeutic agent is a taxol, bortezornib or both.
  • the therapeutic agent is selected from the group consisting of cytotoxic agents, anti-angiogenesis agents and anti- neoplastic agents.
  • the anti-neoplastic agents selected from the group of consisting of alkylating agents, anti-metabolites, epiclophyllotoxims; antineoplastic enzymes, topoisomerase inhibitors, procarbazine, mitoxantrone, platinum coordination complexes, biological response modifiers and growth inhibitors, hormonal/anti- hormonal therapeutic agents, and baematopoietic growth factors.
  • alkylating agents anti-metabolites, epiclophyllotoxims
  • antineoplastic enzymes topoisomerase inhibitors
  • procarbazine mitoxantrone
  • platinum coordination complexes platinum coordination complexes
  • biological response modifiers and growth inhibitors hormonal/anti- hormonal therapeutic agents
  • baematopoietic growth factors baematopoietic growth factors
  • the chemotherapeutic is selected from the group consisting of mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, anti-hormones, angiogenesis inhibitors, and anti-androgens.
  • the combination is administered in combination with an additional therapy.
  • the additional therapy is radiation therapy, chemotherapy, surgery or any combination thereof.
  • the combination is administered in combination with at least one additional therapeutic agent.
  • the therapeutic agent is selected from the group of cytotoxic agents, anti-angiogenesis agents and anti-neopiastic agents.
  • the anti-neoplastic agent is selected from the group of consisting of alkylating agents, anti-metabolites, epidophyllotoxins; antineoplastic enzymes, topoisomerase inhibitors, procarbazines, mitoxantrones, platinum coordination complexes, biological response modifiers and growth inhibitors, hormonal/anti-hormonal therapeutic agents, and haematopoietic growth factors.
  • the second therapeutic is an agent for co-regulating RAF pathways.
  • the second therapeutic agent is a RAF inhibitor.
  • the RAF inhibitor is vemurafenib, dabrafenib, encorafenib, XL-281, LGX-818, CEP-32496, and ARQ-736.
  • the second therapeutic agent is selected from aspirin; diflunisal; salsalate; acetaminophen; ibuprofen; dexibuprofen; naproxen; fenoprofen; ketoprofen; dexketoprofen; flurbiprofen; oxaprozin; loxoprofen; indomethacin; tolmetin; sulindac; etodolac; ketorolac; diclofenac; aceclofenac; nabumetone; enolic acid; piroxicam; meloxicam; tenoxicam; droxicam; lornoxicam; isoxicam; mefenamic acid; meclofena
  • the second therapeutic agent is selected from mechlorethamine; cyclophosphamide; melphalan; chlorambucil; ifosfamide; busulfan; N- nitroso-N-methylurea (MNU); carmustine (BCNU); lomustine (CCNU); semustine (MeCCNU); fotemustine; streptozotocin; dacarbazine; mitozolomide; temozolomide; thiotepa; mytomycin; diaziquone (AZQ); cisplatin; carboplatin; and oxaliplatin.
  • MNU N- nitroso-N-methylurea
  • BCNU carmustine
  • CCNU lomustine
  • Semustine MeCCNU
  • fotemustine streptozotocin
  • dacarbazine mitozolomide
  • temozolomide temozolomide
  • thiotepa mytomycin
  • the second therapeutic agent is selected from vincristine; vinblastine; vinorelbine; vindesine; vinflunine; paclitaxel; docetaxel; etoposide; teniposide; tofacitinib; ixabepilone; irinotecan; topotecan; camptothecin; doxorubicin; mitoxantrone; and teniposide.
  • the second therapeutic agent is selected from actinomycin; bleomycin; plicamycin; mitomycin; daunorubicin; epirubicin; idarubicin; pirarubicin; aclarubicin; mitoxantrone; cyclophosphamide; methotrexate; 5-fluorouracil; prednisolone; folinic acid; methotrexate; melphalan; capecitabine; mechlorethamine; uramustine; melphalan; chlorambucil; ifosfamide; bendamustine; 6-mercaptopurine; and procarbazine.
  • the second therapeutic agent is selected from cladribine; pemetrexed; fludarabine; gemcitabine; hydroxyurea; nelarabine; cladribine; clofarabine; ytarabine; decitabine; cytarabine; cytarabine liposomal; pralatrexate; floxuridine; fludarabine; colchicine; thioguanine; cabazitaxel; larotaxel; ortataxel; tesetaxel; aminopterin; pemetrexed; pralatrexate; raltitrexed; pemetrexed; carmofur; and floxuridine.
  • the second therapeutic agent is selected from azacitidine; decitabine; hydroxycarbamide; topotecan; irinotecan; belotecan; teniposide; aclarubicin; epirubicin; idarubicin; amrubicin; pirarubicin; valrubicin; zorubicin; mitoxantrone; pixantrone; mechlorethamine; chlorambucil; prednimustine; uramustine; estramustine; carmustine; lomustine; fotemustine; nimustine; ranimustine; carboquone; thioTEPA; triaziquone; and triethylenemelamine.
  • the second therapeutic agent is selected from nedaplatin; satraplatin; procarbazine; dacarbazine; temozolomide; altretamine; mitobronitol; pipobroman; actinomycin; bleomycin; plicamycin; aminolevulinic acid; methyl aminolevulinate; efaproxiral; talaporfin; temoporfin; verteporfin; alvocidib; seliciclib; palbociclib; bortezomib; carfilzomib; anagrelide; masoprocol; olaparib; belinostat; panobinostat; romidepsin; vorinosta; idelalisib; atrasentan; bexarotene; testolactone; amsacrine; trabectedin; alitretinoin; tretinoin; demecol
  • the second therapeutic agent is selected from azathioprine; Mycophenolic acid; leflunomide; teriflunomide; tacrolimus; cyclosporin; pimecrolimus; abetimus; gusperimus; lenalidomide; pomalidomide; thalidomide; anakinra; sirolimus; everolimus; ridaforolimus; temsirolimus; umirolimus; zotarolimus; eculizumab; adalimumab; afelimomab; certolizumab pegol; golimumab; infliximab; nerelimomab; mepolizumab; omalizumab; faralimomab; elsilimomab; lebrikizumab; ustekinumab; etanercept; otelixizumab; teplizumab; visili
  • the second therapeutic agent is selected from pascolizumab; gomiliximab; lumiliximab; teneliximab; toralizumab; aselizumab; galiximab; gavilimomab; ruplizumab; belimumab; blisibimod; ipilimumab; tremelimumab; bertilimumab; lerdelimumab; metelimumab; natalizumab; tocilizumab; odulimomab; basiliximab; daclizumab; inolimomab; zolimoma; atorolimumab; cedelizumab; fontolizumab; maslimomab; morolimumab; pexelizumab; reslizumab; rovelizumab; siplizumab; talizumab; tel
  • Z 1 is –CH 2 –
  • Z 2 is selected from the group consisting of –NR 5 R 5’ , -NHCH 2 CO-, C 3 to C 8 cycloalkyl, optionally substituted C 3 to C8 heterocyclyl, optionally substituted C3 to C8 heteroaryl and Z3 is selected from the group consisting of H, deuterium, halo, optionally substituted C1 to C6 alkyl, optionally substituted C 3 to C 8 cycloalkyl, optionally substituted C 6 to C 10 aryl, or -CH 2 -(optionally substituted aryl).
  • R 3 is chloro.
  • R 2 , R 6 , R 7 , and R 13 are each independently selected from the group consisting of H, deuterium, hydroxyl, halogen, cyano, nitro, optionally substituted amino, optionally substituted C-amido, optionally substituted N-amido, optionally substituted ester, optionally substituted sulfonyl, optionally substituted S-sulfonamido, optionally substituted N-sulfonamido, optionally substituted sulfonate, optionally substituted O-thiocarbamyl, optionally substituted N-thiocarbamyl, optionally substituted N-carbamyl, optionally substituted O-carbamyl, optionally substituted urea, optionally substituted C1 to C6 alkoxy, optionally substituted C 1 to C 6 alkyl, optionally substituted C 2 to C 6 alkenyl, optionally substituted C2 to C6 alkynyl, optionally substituted
  • Z 1 is –CH 2 –
  • Z2 is selected from the group consisting of –NR 5 R 5’ , -NHCH2CO-, C3 to C8 cycloalkyl, optionally substituted C3 to C8 heterocyclyl, optionally substituted C3 to C8 heteroaryl and Z3 is selected from the group consisting of H, deuterium, halo, optionally substituted C 1 to C 6 alkyl, optionally substituted C3 to C8 cycloalkyl, optionally substituted C6 to C10 aryl, or -CH2- (optionally substituted aryl).
  • R 3 is chloro.
  • composition of alternative 20, wherein the compound is selected from a compound of Table A, B, C, D, or E.
  • 39. The pharmaceutical composition of any one of any one of alternatives 20 to 38, further comprising one or more immune checkpoint inhibitors.
  • the pharmaceutical composition of alternative 40, wherein the immune checkpoint inhibitor is an inhibitor of PD-1, PD-Ll, PD-L2, PD-L3, PD-L4, CTLA-4, LAG3, B7-H3, B7-H4, KIR or TIM3.
  • 41. The pharmaceutical composition of alternative 39 or 40, wherein the immune checkpoint inhibitor is a PD-1 inhibitor.
  • the pharmaceutical composition of alternative 39 or 40, wherein the immune checkpoint inhibitor is a PD-Ll inhibitor.
  • the pharmaceutical composition of alternative 39 or 40, wherein the first and the second immune checkpoint inhibitor is independently an inhibitor of PD-1 , PD- Ll, PD-L2, PD-L3, PD-L4, CTLA-4, LAG3, B7-H3, B7-H4, KIR or TIM3.
  • the first immune checkpoint inhibitor is a PD-1 inhibitor
  • the second immune checkpoint inhibitor is a CTLA-4 inhibitor.
  • the pharmaceutical composition of alternative 39 or 40, wherein the first immune checkpoint inhibitor is a PD-Ll inhibitor
  • the second immune checkpoint inhibitor is a CTLA-4 inhibitor.
  • a method of treating a disease or disorder comprising administering to a subject suffering from said disease or disorder an effective amount of a compound of any alternatives 1 to 19 or a pharmaceutical composition of any one of alternatives 20 to 55.
  • a method of treating a disease comprising administering to a subject suffering from said disease an effective amount of a compound of any one alternatives 1 to 19 or a pharmaceutical composition of any one of alternatives 20 to 55.
  • a method of treating cancer cachexia in a mammal with cancer comprising administering an effective amount of a compound of any one alternatives 1 to 19 or a pharmaceutical composition of any one of alternatives 20 to 55.
  • the cancer is selected from the group consisting of brain cancer, breast cancer, lung cancer, non-small cell lung cancer, ovarian cancer, pancreatic cancer, stomach cancer, prostate cancer, renal cancer, colorectal cancer or leukemia.
  • the fibrogenetic disorder is scleroderma, polymyositis, systemic lupus, rheumatoid arthritis, liver cirrhosis, keloid formation, interstitial nephritis or pulmonary fibrosis.
  • the method alternative 65 wherein the RAS mutation is a KRAS mutation selected from the group consisting of G12C, G12S, G12R, G12F, G12L, G12N, G12A, G12D, G12V, G13C, G13S, G13D, G13V, G13P, S17G, P34S, A59E, A59G, A59T, Q61K, Q61L, Q61R, and Q61H. [0408] 67.
  • R 6 is hydrogen, fluoro or chloro; R13 is ethyl or -NRARB wherein RA is hydrogen and RB is methyl; Z 2 is - ; R 5 is C1 to R 5’ is C 1 to C 6 alkyl.
  • R 6 is hydrogen, fluoro or chloro; R13 is ethyl or -NRARB wherein RA is hydrogen and RB is methyl; Z 2 is - ; R 5 is C1 to R 5’ is C 1 to C 6 alkyl.
  • 68 The compound of alternative 67, wherein R 5 is methyl.
  • 69 The compound of alternative 68, wherein R 5’ is methyl.
  • 70 The compound of alternative 68, wherein R 5’ is ethyl.
  • 71 The compound of any one of alternatives 67 to 70, wherein Z 2 is - NR 5 R 5’ .
  • R 2 is L;
  • R 6 is selected from the group consisting of H or fluoro, chloro or bromo;
  • R 7 is H;
  • R 13 is selected from the group consisting of optionally substituted optionally substituted amin, C 1 to C 6 alkyl, H, deuterium, hydroxyl, halogen, cyano, nitro, optionally substituted amino, optionally substituted C-amido, optionally substituted N-amido, optionally substituted ester, optionally substituted sulfonyl, optionally substituted S-sulfonamido, optionally substituted N-sulfonamido, optionally substituted sulfonate, optionally substituted O-thiocarbamyl, optionally substituted N-thiocarbamyl, optionally substituted N-carbamyl, optionally substituted O-carbamyl, optionally substituted urea, optionally substituted C1 to C6 alkoxy, optionally substituted C 2 to
  • R 6 is H.
  • a compound having the structure of Formula (III): including R 2 is selected from the group consisting of halogen, H, deuterium, hydroxyl, cyano, nitro, optionally substituted amino, optionally substituted C-amido, optionally substituted N- amido, optionally substituted ester, optionally substituted sulfonyl, optionally substituted S-sulfonamido, optionally substituted N-sulfonamido, optionally substituted sulfonate, optionally substituted O-thiocarbamyl, optionally substituted N-thiocarbamyl, optionally substituted N-carbamyl, optionally substituted O-carbamyl, optionally substituted urea, optionally substituted C 1 to C 6 alkoxy, optionally substituted C 1 to C 6 alkyl, optionally substituted C2 to C6 alkenyl, optionally substituted C2 to C6 alkynyl, optionally substituted C3 to C8 cycloalkyl, optional
  • the compound of alternative 120 wherein the compound is 4- ((dimethylamino)methyl)-3-(2-fluoro-3-((N-methylsulfamoyl)amino)benzyl)-5-methoxy-2- oxo-2H-chromen-7-yl dimethylcarbamate.
  • a pharmaceutical composition comprising a compound of alternatives 1 and a pharmaceutically acceptable salt.
  • the compound of alternative 126 wherein the compound is 3-(2- chloro-3-(ethylsulfonamido)benzyl)-4-((dimethylamino)methyl)-2-oxo-2H-chromen-7-yl dimethylcarbamate.
  • 129. The compound of alternative 126, wherein the compound is 3-(2- chloro-3-((N-methylsulfamoyl)amino)benzyl)-6-fluoro-2-oxo-4-(piperazin-1-ylmethyl)-2H- chromen-7-yl dimethylcarbamate.
  • the compound of alternative 126 wherein the compound is 3-(2- chloro-3-(ethylsulfonamido)benzyl)-6-fluoro-2-oxo-4-(piperazin-1-ylmethyl)-2H-chromen-7- yl dimethylcarbamate.
  • 131 The compound of alternative 126, wherein the compound is 6-chloro- 3-(2-chloro-3-((N-methylsulfamoyl)amino)benzyl)-2-oxo-4-(piperazin-1-ylmethyl)-2H- chromen-7-yl dimethylcarbamate.
  • 132 132.
  • the compound of alternative 126 wherein the compound is 6-chloro- 3-(2-chloro-3-(ethylsulfonamido)benzyl)-2-oxo-4-(piperazin-1-ylmethyl)-2H-chromen-7-yl dimethylcarbamate.
  • 133 The compound of alternative 126, wherein the compound is 3-(2- chloro-3-(ethylsulfonamido)benzyl)-2-oxo-4-(piperazin-1-ylmethyl)-2H-chromen-7-yl dimethylcarbamate.
  • 134 134.
  • a pharmaceutical composition comprising a compound of any one of alternatives 126 to 134 and a pharmaceutically acceptable salt [0476] 136.
  • the compound of alternative 136 wherein the compound is 3-(2- chloro-3-((N-methylsulfamoyl)amino)benzyl)-4-((ethyl(methyl)amino)methyl)-6-fluoro-2- oxo-2H-chromen-7-yl dimethylcarbamate.
  • 140 The compound of alternative 136, wherein the compound is 4- (azetidin-1-ylmethyl)-3-(2-chloro-3-((N-methylsulfamoyl)amino)benzyl)-6-fluoro-2-oxo-2H- chromen-7-yl dimethylcarbamate.
  • 141 141.
  • the compound of alternative 136 wherein the compound is 6-chloro- 3-(2-chloro-3-((N-methylsulfamoyl)amino)benzyl)-4-((dimethylamino)methyl)-2-oxo-2H- chromen-7-yl dimethylcarbamate.
  • 142 The compound of alternative 136, wherein the compound is 6-chloro- 3-(2-chloro-3-((N-methylsulfamoyl)amino)benzyl)-4-((ethyl(methyl)amino)methyl)-2-oxo- 2H-chromen-7-yl dimethylcarbamate.
  • 143 The compound of alternative 136, wherein the compound is 6-chloro- 3-(2-chloro-3-((N-methylsulfamoyl)amino)benzyl)-4-((ethyl(methyl)amino)methyl)-2-oxo- 2H-chromen-7-yl dimethylcarbamate.
  • a pharmaceutical composition comprising a compound of any one of alternatives 136 to 142 and a pharmaceutically acceptable salt.
  • [0484] 144. A compound selected from the group consisting of: 3-(2-fluoro-3-((N-methylsulfamoyl)amino)benzyl)-4-(((2- fluoroethyl)(methyl)amino)methyl)-2-oxo-2H-chromen-7-yl dimethylcarbamate; 3-(3-(ethylsulfonamido)-2-fluorobenzyl)-4-(((2- fluoroethyl)(methyl)amino)methyl)-2-oxo-2H-chromen-7-yl dimethylcarbamate; 3-(2-fluoro-3-((N-methylsulfamoyl)amino)benzyl)-4-((methyl(prop-2-yn-1- yl)amino)methyl)-2-oxo-2H-chromen-7-yl dimethyl
  • the compound of alternative 144 wherein the compound is 3-(2- fluoro-3-((N-methylsulfamoyl)amino)benzyl)-4-((methyl(prop-2-yn-1-yl)amino)methyl)-2- oxo-2H-chromen-7-yl dimethylcarbamate.
  • the compound of alternative 144 wherein the compound is 4-(((2,2- difluoroethyl)(methyl)amino)methyl)-3-(2-fluoro-3-((N-methylsulfamoyl)amino)benzyl)-2- oxo-2H-chromen-7-yl dimethylcarbamate.
  • 149 The compound of alternative 144, wherein the compound is 3-(2- fluoro-3-((N-methylsulfamoyl)amino)benzyl)-4-((methyl(prop-2-yn-1-yl)amino)methyl)-2- oxo-2H-chromen-7-yl di
  • the compound of alternative 144, wherein the compound is 4- (((cyanomethyl)(methyl)amino)methyl)-3-(2-fluoro-3-((N-methylsulfamoyl)amino)benzyl)- 2-oxo-2H-chromen-7-yl dimethylcarbamate.
  • 150 The compound of alternative 144, wherein the compound is 4- ((dimethylamino)methyl)-3-(2-fluoro-3-(methyl(sulfamoyl)amino)benzyl)-2-oxo-2H- chromen-7-yl dimethylcarbamate.
  • the compound of alternative 144, wherein the compound is 4- ((dimethylamino)methyl)-3-(2-fluoro-3-(hydroxymethyl)benzyl)-2-oxo-2H-chromen-7-yl dimethylcarbamate.
  • 152. The compound of alternative 144, wherein the compound is 3-(3-((tert- butylsulfinyl)amino)-2-fluorobenzyl)-4-((dimethylamino)methyl)-2-oxo-2H-chromen-7-yl dimethylcarbamate.
  • a pharmaceutical composition comprising a compound of any one of alternatives 144 to 152 and a pharmaceutically acceptable salt.
  • 154 A pharmaceutical composition comprising a compound of any one of alternatives 144 to 152 and a pharmaceutically acceptable salt.
  • R 2 , R 6 , R 7 , and R 13 are each independently selected from the group consisting of H, deuterium, hydroxyl, halogen, cyano, nitro, optionally substituted amino, optionally substituted C-amido, optionally substituted N-amido, optionally substituted ester, optionally substituted sulfonyl, optionally substituted S-sulfonamido, optionally substituted N-sulfonamido, optionally substituted sulfonate, optionally substituted O-thiocarbamyl, optionally substituted N-thiocarbamyl, optionally substituted N-carbamyl, optionally substituted O-carbamyl, optionally substituted urea, optionally substituted C1 to C6 alkoxy, optionally substituted C 1 to C 6 alkyl, optionally substituted C 2 to C 6 alkenyl, optionally substituted C 2 to
  • the compound or pharmaceutically acceptable salt according to altnernative 154 wherein the compound is selected from the group consisting of: ; ; ; ; ; ; and . [0507] 167.
  • the compound or pharmaceutically acceptable salt according to alternative 154 wherein the compound is: . or pharmaceutically acceptable salt according to alternative 154, wherein the compound is: . compound or pharmaceutically acceptable salt according to alternative 154, wherein the compound is: . compound or pharmaceutically acceptable salt according to alternative 154, wherein the compound is: . [0511] 171.
  • a pharmaceutical composition comprising a therapeutically effective amount of at least one compound or pharmaceutically acceptable salt thereof as defined in any one of alternatives 154 to 175.
  • R 2 , R 6 , R 7 , and R 13 are each independently selected from the group consisting of H, deuterium, hydroxyl, halogen, cyano, nitro, optionally substituted amino, optionally substituted C-amido, optionally substituted N-amido, optionally substituted ester, optionally substituted sulfonyl, optionally substituted S-sulfonamido, optionally substituted N-sulfonamido, optionally substituted sulfonate, optionally substituted O-thiocarbamyl, optionally substituted N-thiocarbamyl, optionally substituted N-carbamyl, optionally substituted O-carbamyl, optionally substituted urea, optionally substituted C 1 to C 6 alkoxy, optionally substituted C1 to C6 alkyl, optionally substituted C2 to C6 alkenyl, optionally substituted C 2 to C 6 alkynyl
  • Z1 is –CH2–
  • Z2 is selected from the group consisting of –NR 5 R 5’ , -NHCH2CO-, C3 to C8 cycloalkyl, optionally substituted C3 to C 8 heterocyclyl, optionally substituted C 3 to C 8 heteroaryl
  • Z 3 is selected from the group consisting of H, deuterium, halo, optionally substituted C1 to C6 alkyl, optionally substituted C3 to C8 cycloalkyl, optionally substituted C6 to C10 aryl, or -CH2-(optionally substituted aryl).
  • R 3 is chloro.
  • R 2 , R 6 , R 7 , and R 13 are each independently selected from the group consisting of H, deuterium, hydroxyl, halogen, cyano, nitro, optionally substituted amino, optionally substituted C-amido, optionally substituted N-amido, optionally substituted ester, optionally substituted sulfonyl, optionally substituted S-sulfonamido, optionally substituted N-sulfonamido, optionally substituted sulfonate, optionally substituted O-thiocarbamyl, optionally substituted N-thiocarbamyl, optionally substituted N-carbamyl, optionally substituted O-carbamyl, optionally substituted urea, optionally substituted C 1 to C 6 alkoxy, optionally substituted C1 to C6 alkyl, optionally substituted C2 to C6 alkenyl, optionally substituted C2 to C6 alkynyl, optionally substituted
  • composition of Alternative 20 wherein the compound is selected from a compound of Table A.
  • 39 The pharmaceutical composition of any one of any one of Alternatives 20 to 38, further comprising one or more immune checkpoint inhibitors.
  • 40. The pharmaceutical composition of Alternative 39, wherein the immune checkpoint inhibitor is an inhibitor of PD-1, PD-Ll, PD-L2, PD-L3, PD-L4, CTLA-4, LAG3, B7-H3, B7-H4, KIR or TIM3.
  • 41 The pharmaceutical composition of alternative 39 or 40, wherein the immune checkpoint inhibitor is a PD-1 inhibitor.
  • 42 The pharmaceutical composition of alternative 39 or 40, wherein the immune checkpoint inhibitor is a PD-Ll inhibitor.
  • the pharmaceutical composition of alternative 39 or 40, wherein the first and the second immune checkpoint inhibitor is independently an inhibitor of PD-1 , PD- Ll, PD-L2, PD-L3, PD-L4, CTLA-4, LAG3, B7-H3, B7-H4, KIR or TIM3.
  • the pharmaceutical composition of alternative 39 or 40, wherein the first immune checkpoint inhibitor is a PD-1 inhibitor, and the second immune checkpoint inhibitor is a CTLA-4 inhibitor.
  • the first immune checkpoint inhibitor is a PD-Ll inhibitor
  • the second immune checkpoint inhibitor is a CTLA-4 inhibitor.
  • a method of treating a disease or disorder comprising administering to a subject suffering from said disease or disorder an effective amount of a compound of any one alternatives 1 to 19 or a pharmaceutical composition of any one of alternatives 20 to 55.
  • a method of treating a disease comprising administering to a subject suffering from said disease an effective amount of a compound of any one alternatives 1 to 19 or a pharmaceutical composition of any one of alternatives 20 to 55.
  • 59. The method of any one of alternatives 56 to 58, wherein the disease is cancer.
  • a method of treating cancer cachexia in a mammal with cancer comprising administering an effective amount of a compound of any one alternatives 1 to 19 or a pharmaceutical composition of any one of alternatives 20 to 55.
  • the cancer is selected from the group consisting of brain cancer, breast cancer, lung cancer, non-small cell lung cancer, ovarian cancer, pancreatic cancer, stomach cancer, prostate cancer, renal cancer, colorectal cancer or leukemia.
  • the fibrogenetic disorder is scleroderma, polymyositis, systemic lupus, rheumatoid arthritis, liver cirrhosis, keloid formation, interstitial nephritis or pulmonary fibrosis.
  • RAS mutation is a KRAS mutation selected from the group consisting of G12C, G12S, G12R, G12F, G12L, G12N, G12A, G12D, G12V, G13C, G13S, G13D, G13V, G13P, S17G, P34S, A59E, A59G, A59T, Q61K, Q61L, Q61R, and Q61H.
  • Compound 3 A mixture of Compound 2 (1.0 eq.) and sodium iodide (1.0 eq.) was stirred in THF (dry) for 30 min. In another flask, ethyl 3-oxobutanoate (1.10 eq.) was dissolved in THF (dry) and lithium tert-butoxide (1.10 eq.) was slowly added. The reaction mixture was stirred for 30 min. and was then slowly added to the bromide suspension.
  • the amine (1 – 10 eq.) was added.
  • 2-5 eq. of Et 3 N were added and the reaction mixture was stirred for 2 – 16 h at rt.
  • the reaction mixture was filtered and purified by preparative HPLC (method: prep acid or prep base) to obtain the desired amine B.5 after freeze drying or GenevacTM as a solid.
  • Step 1 Starting with 4-(chloromethyl)-5-fluoro-3-(2-fluoro-3-((N- methylsulfamoyl)amino)benzyl)-2-oxo-2H-chromen-7-yl dimethylcarbamate and dimethylamine 2.0 M in MeOH, following the geneal synthesis of Compound B.5. The product was combined with another batch and purified by column chromatography to obtain the title compound after freeze drying as a white solid.
  • Step 1 Starting with 4-(bromomethyl)-3-(2-chloro-3-((N- methylsulfamoyl)amino)benzyl)-2-oxo-2H-chromen-7-yl dimethylcarbamate and N-Boc piperazine, following the general synthesis of Compound B.5.. The product was purified by prep basic. Desired fractions were combined and concentrated under reduced pressure to obtain the amine as a colorless oil.
  • Step 2 The amine was dissolved in 1,4-dioxane (3 mL) and HCl in dioxane (4M, 16.7 eq,) was added and stirred for 1 hour at rt. The reaction mixture was concentrated under reduced pressure and the twice co-evaporated with DCMDCM. The residue was dissolved in MeCN/water and lyophilized to obtain the title compound as a white solid.
  • Step 1 Starting with 4-(bromomethyl)-6-fluoro-3-(2-fluoro-3-((N- methylsulfamoyl)amino)benzyl)-2-oxo-2H-chromen-7-yl dimethylcarbamate and N- Methylethylamine, following the geneal synthesis of Compound B.5, The product was purified by column chromatography to obtain the title compound after freeze drying as a white solid.
  • Step 2 The product was purified by prep basic. Desired fractions were combined and concentrated under reduced pressure to obtain the amine (as a colorless oil.
  • Step 2 The amine was dissolved in 1,4-dioxane (3 mL) and HCl in dioxane (4M, 16.7 eq.) was added and stirred for 1 hour at rt. The reaction mixture was concentrated under reduced pressure and the twice co-evaporated with DCMDCM. The residue was dissolved in MeCN/water and lyophilized to obtain the title compound as a white solid.
  • Step 1 Starting with 4-(bromomethyl)-3-(2-fluoro-3-((N- methylsulfamoyl)amino)benzyl)-2-oxo-2H-chromen-7-yl dimethylcarbamate and N- methylprop-2-yn-1-amine, following the geneal synthesis of Compound B.5.. The product was purified by column chromatography to obtain the title compound after freeze drying as a white solid.
  • Step 1 Starting with 4-(bromomethyl)-3-(2-fluoro-3-((N- methylsulfamoyl)amino)benzyl)-2-oxo-2H-chromen-7-yl dimethylcarbamate and 2,2- difluoro-N-methylethan-1-amine, following the geneal synthesis of Compound B.5. The product was purified by column chromatography to obtain the title compound after freeze drying as a white solid.
  • Step 1 Starting with 4-(bromomethyl)-3-(2-fluoro-3-((N- methylsulfamoyl)amino)benzyl)-2-oxo-2H-chromen-7-yl dimethylcarbamate (45 mg, 0.075 mmol, py:40%) and 2-(methylamino)acetonitrile, following the geneal synthesis of Compound B.2, with addition of Net3 3.0 eq. The product was purified by prep basic followed by prep acid to obtain the title compound (3.8 mg, 0.007 mmol, y: 21%) after freeze drying as a white solid.
  • reaction mixture was stirred for 18 hours at 100 °C.
  • Reaction mixture was filtered and washed with MeCN.
  • 40 mg of the still impure product was purified by prep basic to obtain the title compound (29 mg, 0.067 mmol, yield: 1.5%) as an off-white solid.
  • reaction mixture was stirred for 18 hours at 100 °C.
  • Reaction mixture was filtered and washed with MeCN.
  • Step 2 To a solution of 4-((dimethylamino)methyl)-3-(2-fluoro-3- (hydroxymethyl)benzyl)-2-oxo-2H-chromen-7-yl dimethylcarbamate (1.41 g, 3.29 mmol, 1.0 eq.) in DCM (0.23 M) at 0 °C was slowly added a solution of thionyl chloride (0.48 ml, 6.58 mmol, 2.0 eq.) in DCM (2 ml). The formed reaction mixture was stirred for 1 hour allowing to warm to rt.
  • Step 3 To a mixture of 3-(3-(chloromethyl)-2-fluorobenzyl)-4- ((dimethylamino)methyl)-2-oxo-2H-chromen-7-yl dimethylcarbamate (0.5 g, 1.12 mmol, 1.0 eq.) in ethanol (Abs) (0.1 M) under N2 atmosphere was added thiourea (0.102 g, 1.343 mmol, 1.2 eq.). The reaction mixture was stirred for 2 hours at 80 °C and then for 2 days at rt. NaOH 2 N (1.68 ml, 3.36 mmol, 3.0 eq.) was added and stirred for 2 hours at 80 °C.
  • the reaction mixture was acidified with 1M HCl. Some extra water was added followed by DCM. The layers were separated by a phase separator and the organic layer was concentrated under reduced pressure to obtain 4-((dimethylamino)methyl)-3-(2-fluoro-3- (mercaptomethyl)benzyl)-2-oxo-2H-chromen-7-yl dimethylcarbamate and its dimer ((((disulfanediylbis(methylene))bis(2-fluoro-3,1-phenylene))bis(methylene))bis(4- ((dimethylamino)methyl)-2-oxo-2H-chromene-3,7-diyl) bis(dimethylcarbamate)) (570 mg, 0.67 mmol, yield: 60%) as a yellow solid.
  • Step 4 (((disulfanediylbis(methylene))bis(2-fluoro-3,1- phenylene))bis(methylene))bis(4-((dimethylamino)methyl)-2-oxo-2H-chromene-3,7-diyl) bis(dimethylcarbamate) (0.57 g, 0.257 mmol, 1.0 eq.) was dissolved in THF (0.11 M)/water (0.11 M) and cooled to 0 °C. tri-n-butylphosphine (0.071 ml, 0.283 mmol, 1.1 eq.) was slowly added and stirred for 1 hour at rt.
  • Step 5 4-((dimethylamino)methyl)-3-(2-fluoro-3- (mercaptomethyl)benzyl)-2-oxo-2H-chromen-7-yl dimethylcarbamate (50 mg, 0.112 mmol, 1.0 eq.) was dissolved in DMF (dry) (0.23 M), Cs 2 CO 3 (36.6 mg, 0.112 mmol, 1.0 eq.) and iodoethane (10.91 ⁇ l, 0.135 mmol, 1.2 eq.) were added and stirred for 1 hour at rt.
  • Step 6 4-((dimethylamino)methyl)-3-(3-((ethylthio)methyl)-2- fluorobenzyl)-2-oxo-2H-chromen-7-yl dimethylcarbamate (43 mg, 0.091 mmol, 1.0 eq.) was dissolved in MeOH (0.06 M)/Water (0.06 M), oxone, monopersulfate compound (55.9 mg, 0.091 mmol, 1.0 eq.) was added and stirred for 1 hour at rt. Water was added to the reaction mixture and the product was extracted with DCM. Combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure.
  • the reaction mixture was flushed with argon and Pd 2 (dba) 3 (4.80 mg, 5.24 ⁇ mol, 0.05 eq.) and t-BuXPhos (8.90 mg, 0.021 mmol, 0.2 eq.) were added.
  • the reaction mixture was stirred for 48 hours at 50 °C.
  • the impure product was purified by prep. basic to obtain the title compound (40 mg, 0.076 mmol, yield: 72.7%) as a white solid. Yield: The title compound was isolated as a white solid (73% over 1 step).
  • Step 1 Starting with 6-chloro-3-(2-chloro-3-((N- methylsulfamoyl)amino)benzyl)-4-(chloromethyl)-2-oxo-2H-chromen-7-yl dimethylcarbamate and N-ethylmethylamine, following the geneal synthesis of Compound B.5. The impure product was purified by column chromatography to obtain the title compound (after freeze drying as a white solid.
  • EXAMPLE 35 Materials & Methods [0703] Media components, reagents and buffers for Western Blot: All cell culture media components were obtained from ThermoFisher Scientific. Cell lysis/Protein Extraction Reagent (Cell Signal Technology, Cat No: 9803).
  • MOPS/SDS electrophoresis running buffer GenScript, Cat No. M00138.
  • Tris-buffered saline with Tween 20 (TBST buffer): 20 mM Tris-HCl (pH 7.5), 150 mM NaCl, 0.1% Tween 20 NuPAGE gels, 4-12% (ThermoFisher Scientific, Cat No. NP0322BOX).
  • iBLOT nitrocellulose transfer kit (ThermoFisher Scientific Cat No. IB301002). Blocking Buffer (LICOR Cat No. 927-50000).
  • Antibodies Phospho-STAT3 (S727), mouse polyclonal antibodies were obtained from BD Biosciences (Cat No.
  • Anti-STAT3 rabbit monoclonal antibodies (Cat No. 12640), Anti- phospho-MEK1/2 (S218/S222), rabbit polyclonal antibodies (Cat No: 9121), Anti MEK- 1/2, rabbit monoclonal antibodies (Cat No: 9122), Anti-ERK, mouse monoclonal antibodies (Cat No: 9107), and Anti-phospho-ERK, rabbit monoclonal antibodies (Cat No. 4377).
  • Secondary antibodies IRDye 800CW goat anti-rabbit antibodies (LICOR Cat No. 926-32211), IRDye 680RD goat anti-rabbit antibodies (LICOR Cat No.
  • Tumor Cell Lines Cell Lines and Tissue Culture conditions: The A549 (Cat No. CCL-185) cell line was obtained from American Type Culture Collection (ATCC) and grown in T75 flasks in DMEM containing 10% FBS and Pen-Strep at 37C in a humidified, 5% CO incubator. [0707] The Colon26 syngeneic adenocarcinoma cell line was obtained from the National Cancer Institute.
  • Colon26 tumor cells were maintained as exponentially growing cultures in RPMI-1640 medium containing 10% fetal bovine serum, 2 mM glutamine, 100 units/mL penicillin G sodium, 100 ⁇ g/mL streptomycin sulfate, 25 ⁇ g/mL gentamicin, 10 mM HEPES, and 0.075% sodium bicarbonate.
  • the tumor cells were grown in tissue culture flasks in a humidified incubator at 37 °C, in an atmosphere of 5% CO2 and 95% air.
  • Subculture conditions Adherent cells were grown to approximately 90% confluency, culture medium was aspirated and the cell layer was rinsed with PBS.
  • Cell lysis and protein estimation Cells were washed with PBS, and scraped in 50 uL of lysis buffer containing protease and phosphatase inhibitors. Cell lysates were stored at -20C. Cell lysates were thawed and spun at 12,000 rpm for one minute, 3 ul of the supernatant was added to 500 uL of Coomassie blue reagent following by 500 uL of water. Absorbance was read at 595 nm after 10 minutes of incubation.
  • Protein standards were used (0 – 20 mg/mL) to calculate protein concentrations of test samples.
  • Western Blotting For electrophoresis 20 ug of protein was mixed with 5 ul of 4X Laemmle’s sample buffer and 1 ul of 0.4 M DTT in a volume of 20 ul made up with lysis buffer. All samples were heated at 95C for 5 minutes, cooled to room temperature and spun down. Protein samples were loaded onto 4-12% polyacrylamide gels and run at 100V for approximately 1.5 hours till the blue dye reached the bottom. After the run, gel was removed and protein transfer was done using iBlot for 7 minutes, as per manufacturer’s recommendations.
  • nitrocellulose membrane was incubated on a shaker in 5 mL of blocking buffer at room temperature for 1hr. The blot was then incubated overnight on a shaker in 5 ml of blocking buffer containing 0.2% Tween-20 and primary antibody, at room temperature.
  • Anti-phospho-STAT3 antibody was used at a dilution of 1:500, the other 3 primary antibodies were used at a dilution of 1:1000.
  • the blot was washed 3 times for 10 min each with 10 mL of TBST followed by incubation on a shaker in 5 ml of blocking buffer containing 0.2% Tween-20 and 0.5 ul of the IRDye labeled secondary antibodies, diluted 1:10000, at room temperature for 1hr. The blot was then washed 3 times for 10 min each with 10 mL of TBST and dried between sheets of paper towels. Imaging was done using LICOR’s Odyssey imaging system, quantitation was done using their software, Image Studio version 3.1.
  • mice Female BALB/c mice (BALB/cAnNCrl, Charles River) were eleven weeks old on Day 1 of the study and had a body weight (BW) range of 15.8 to 21.4 g. The animals were fed ad libitum water (reverse osmosis, 1 ppm Cl) and NIH 31 Modified and Irradiated Lab Diet® consisting of 18.0% crude protein, 5.0% crude fat, and 5.0% crude fiber. The mice were housed on irradiated Enrich-o’cobsTM bedding in static microisolators on a 12-hour light cycle at 20–22 °C (68–72 °F) and 40–60% humidity.
  • BW body weight
  • CR Discovery Services specifically complies with the recommendations of the Guide for Care and Use of Laboratory Animals with respect to restraint, husbandry, surgical procedures, feed and fluid regulation, and veterinary care.
  • the animal care and use program at CR Discovery Services is accredited by the Association for Assessment and Accreditation of Laboratory Animal Care International (AAALAC), which assures compliance with accepted standards for the care and use of laboratory animals.
  • AALAC Laboratory Animal Care International
  • EXAMPLE 36 Pharmacokinetic Properties [0718] Physicochemical Properties: Absorption & Efflux [0719] Materials and Methods: Stock solutions of Test Article (TA) was prepared at 50 mM in DMSO and was further diluted to 10 mM using DMSO. Control inhibitor stock solutions were prepared at concentrations that were 1000X the final assay concentration. Individual stocks for controls ranitidine, warfarin and talinolol were prepared at 10 mM in DMSO and verapamil stock solution was prepared at 25 mM in DMSO.
  • TA Test Article
  • Caco-2 cells obtained from ATCC, clone C2BBe1, were grown in 96-well Transwell plates, cultured for 27 days in Dulbecco’s modified Eagle’s medium supplemented with 10% fetal bovine serum, 1% L -glutamax,1% penicillin-streptomycin (pen-strep) and 10 mM HEPES (2-[4-(2-hydroxyethyl) piperazin-1- yl] ethane sulfonic acid) and incubated at 37°C, 5% CO2, and humidified.
  • Dulbecco’s modified Eagle’s medium supplemented with 10% fetal bovine serum, 1% L -glutamax,1% penicillin-streptomycin (pen-strep) and 10 mM HEPES (2-[4-(2-hydroxyethyl) piperazin-1- yl] ethane sulfonic acid
  • test article 10 mM for conditions to be tested for test article as inhibitor
  • control inhibitor for conditions to be tested for test article and control articles as substrates
  • stock solutions were diluted 500-fold to get 2X the final assay concentrations).
  • blank DMSO was added to normalize DMSO percentage in the assay system.
  • the basal assay plates were prepared by filling apical-to-basolateral wells of a 96-well sterile plate with 250 ⁇ L of Transport Buffer and basolateral-to-apical wells with 130 ⁇ L of 2X assay solutions prepared above and 130 ⁇ L of HBSS. Samples (10 ⁇ L) were collected each from basal compartment of basolateral-to-apical wells for time zero (T0) samples and were diluted with 4 volumes of transport buffer (HBSS). Monolayer integrity was assessed for the assay plate before and after the assay by taking measurements of Transepithelial Electrical Resistance (TEER) of cell monolayer in each assay well.
  • TEER Transepithelial Electrical Resistance
  • Caco- 2 cell plates were prepared for the bidirectional assay by exchanging the cell culture medium (Dulbecco’s modified Eagle’s medium with supplements) in the apical wells of the plate three times with 85 ⁇ L transport buffer (HBSS). After the final wash, 52.5 ⁇ L of the buffer was removed from the apical wells (leaving 62.5 ⁇ L buffer in the wells) and replaced with 62.5 ⁇ L of 2X assay solutions prepared above for apical-to-basolateral wells or 52.5 ⁇ L of fresh Transport Buffer for basolateral-to-apical wells.
  • the cell culture medium Dulbecco’s modified Eagle’s medium with supplements
  • HBSS transport buffer
  • 52.5 ⁇ L of the buffer was removed from the apical wells (leaving 62.5 ⁇ L buffer in the wells) and replaced with 62.5 ⁇ L of 2X assay solutions prepared above for apical-to-basolateral wells or 52.5 ⁇ L of fresh Transport Buffer
  • the total volume in the apical wells at this point is assumed to be 125 ⁇ L for apical-to-basolateral wells and 115 ⁇ L for basolateral-to- apical wells.
  • Samples (10 ⁇ L) were collected from each apical compartment of apical-to- basolateral wells for time zero (T0) samples and were diluted with 4 volumes of transport buffer (HBSS).
  • the final nominal concentrations for the assay were 10 ⁇ M for TA as substrateand as inhibitor, 10 ⁇ M for all control substrates, 10 ⁇ M for valspodar, Ko143, ranitidine, talinolol and warfarin and 25 ⁇ M for verapamil.
  • the apical section of the Caco-2 plate was transferred to the basal plate and incubated at 37°C for 2 hours; the assay was performed in triplicate. Following the 2-hour incubation period, samples (10 ⁇ L each) were collected from all apical compartments, samples (50 ⁇ L each) were collected from apical-to- basolateral basal compartments, and samples (10 ⁇ L each) were collected from basolateral-to-apical basal compartments.
  • the 10- ⁇ L samples from both the apical and basolateral sides of the cell monolayer were diluted with 4 volumes of transport buffer (HBSS). The samples were quenched with 100 ⁇ L ice-cold acetonitrile containing internal standards at 250 ng/mL concentration.
  • Cisapride (hERG positive control): eight (8) concentrations ranging from 0.003 - 3 ⁇ M were used to determine the dose response of the block of hERG current.
  • hERG current was measured using stimulus voltage patterns with fixed amplitudes: activation pre-pulse (TP1) to +40 mV for 2 s and test pulse (TP2) to -40 mV for 2 s from a holding potential of -80 mV.
  • hERG current was measured as the outward peak current at TP2 (tail current). The stimulation was repeated with 0.1 Hz frequency during 2 min as baseline and 5 min after TA application.
  • Data acquisition and analyses were performed using the SP384PE system operation software.
  • %Block 100%- ((%Block - %PC)*(100% / (%VC - %PC)), [0737] where %VC and %PC were the mean values of the current block with the vehicle and positive controls, respectively.
  • the liver microsomes (20 mg protein/mL) were diluted in 0.1 M potassium phosphate buffer, pH 7.4, (warmed to 37°C) to a concentration of 1 mg protein/mL (2X final assay concentration).
  • 75 ⁇ L of diluted 2X microsomes were added to an equal volume of 2X compound/NADPH solution in a polypropylene 96-well microtiter plate. The plate was incubated with gentle shaking at 37°C. Duplicate 30 ⁇ L aliquots were removed immediately after compound addition (T 0 : time zero) and at 60 minutes. At each timepoint, control and test article samples were quenched with 180 ⁇ L of ice-cold acetonitrile containing internal standards.
  • the quenched samples were gently mixed and were stored in a -20°C freezer for at least 30 minutes. After the final time point (60 minutes), the quenched samples were vortexed (10 minutes) and then centrifuged at 3100 rpm for 10 minutes at 4°C. Supernatant (50 ⁇ L) was removed, transferred to a new 96- well plate, and diluted with 100 ⁇ L of water. The sample plate was sealed, mixed, and then stored refrigerated until analysis.
  • Bioanalysis [0743] For the analysis of all test articles and verapamil (control), a Waters XSELECT HSS T32.5 ⁇ m, 30 ⁇ 2.1 mm column was used with a gradient (0.9 mL/min flow rate) starting at 99% mobile phase A (0.1% formic acid in water) to 95% mobile phase B (0.1% formic acid in acetonitrile). The column oven was set to a temperature of 55°C. [0744] Analytes and internal standards were detected using an Applied Biosystems Sciex API 5500 triple quadrupole mass spectrometer with Sound Analytics ADDA Autosampler. The instruments were equipped with an electrospray ionization source (600°C) operated in the positive ion.
  • Clint is intrinsic clearance calculated in the units of mL/min/kg of protein CD-1 mouse CD-1 mouse: 45 mg microsomes/g liver; 87.5 g liver/kg body weight Sprague-Dawley rat: 45 mg microsomes/g liver; 45 g liver/kg body weight Beagle dog: 45 mg microsomes/g liver; 25 g liver/kg body weight Human: 45 mg microsomes/g liver; 20 g liver/kg body weight [0749] Physicochemical Properties: Plasma Stability [0750] Materials and Methods: Stock solutions of Test Article (TA) were prepared at a concentration of 50 mM in DMSO and was further diluted to 2 mM with DMSO.
  • Frozen matrices (Balb/c mouse, Wistar Han rat, Beagle dog, and human plasma containing K2EDTA as anti-coagulant) were thawed and centrifuged at 3100 rpm for 10 minutes at 4°C to remove particulates. The lipid layer was then removed, and the supernatant was transferred to a new tube without disturbing the pellet. After the matrices were incubated at 37°C for at least 10 minutes, the pH of each matrix was adjusted to pH 7.4, using 10% phosphoric acid or 1N sodium hydroxide as necessary.
  • Test and control article stock solutions were spiked (2 ⁇ L) into matrix (1.998 mL) and mixed to final assay concentration of 2 ⁇ M.
  • Duplicate 30- ⁇ L aliquots of spiked plasma were transferred to matrix tubes in 96-well plate immediately after spiking for timepoints of 0, 30, 60, 120, 240, and 360 minutes. These tubes were then incubated at 37°C with shaking and at each timepoint, the corresponding tubes containing samples were quenched with 180 ⁇ L of cold acetonitrile containing the internal standards. The quenched samples were vortex-mixed briefly and stored refrigerated.
  • the column was set to a temperature of 55°C.
  • a Waters XSELECT HSS T3 2.5 ⁇ m, 30 ⁇ 2.1 mm column was used with a gradient (0.9 mL/min flow rate) starting at 70% mobile phase A (0.1% formic acid in water) to 95% mobile phase B (0.1% formic acid in acetonitrile).
  • the column was set to a temperature of 55°C.
  • All analytes and internal standards were detected using an Applied Biosystems Sciex API-5500 triple quadrupole mass spectrometer with Agilent 1260 Infinity Binary Pump and Apricot Designs ADDA High-Speed Dual Arm Autosampling System.
  • T 1/2 -0.693 / Slope
  • TA Test Article
  • Positive control inhibitors (fluvoxamine, ticlopidine, quercetin, sulfaphenazole, omeprazole, paroxetine and mifepristone) were dissolved at 50 mM in DMSO followed by eight serial dilutions in DMSO down to 0.00545 mM. Each inhibitor solution and DMSO alone were diluted 3.33-fold in acetonitrile followed by a further 100-fold dilution into 100 mM potassium phosphate buffer, pH 7.4 with and without 3 mM NADPH (to make 3X compound solutions). Pooled human liver microsomes were submerged in a 37°C water bath until just thawed and then placed on ice.
  • the human liver microsomes were diluted in 100 mM potassium phosphate buffer (warmed), pH 7.4 to 0.3 mg/mL (3X concentration) directly before use.
  • the substrates were used at approximately their Km concentration for the respective isoenzyme, which were expected to fall within the linear range of CYP450-mediated metabolism.
  • the substrates in DMSO 40 mM phenacetin, 25 mM bupropion, 1 mM amodiaquine, 10 mM diclofenac, 40 mM mephenytoin, 10 mM dextromethorphan, and 125 mM testosterone
  • methanol 3.06 mM midazolam
  • 3X 3X buffer/cofactor/substrate
  • the 3X substrate solutions with 3 mM NADPH in 100 mM KPhos buffer, pH 7.4 were added to the plate containing compound solutions with no NADPH in 100 mM KPhos buffer, pH 7.4 and vice versa; the final concentrations of DMSO and ACN were less than 1%.
  • final concentrations were 0.00, 0.00545, 0.0218, 0.0870, 0.348, 1.39, 5.55, 22.2, 33.3 and 50.0 ⁇ M.
  • Supernatants from 1A2, 2B6 and 2C8 assays were pooled (50 ⁇ L per sample).
  • Supernatants from 2D6, 3A4 (midazolam substrate) and 3A4 (testosterone substrate) assays were pooled (50 ⁇ L per sample). Pooled samples were diluted with 300 ⁇ L water prior to analysis. For unpooled 2C9 and 2C19 assay samples, supernatant (50 ⁇ L) was diluted with 100 ⁇ L water prior to analysis.
  • Bioanalysis For TA, a Waters XSELECT HSS T3 2.5 ⁇ m, 30 ⁇ 2.1 mm column was used with a gradient (1.0 mL/min flow rate) starting at 99% mobile phase A (0.1% formic acid in water) to 95% mobile phase B (0.1% formic acid in acetonitrile). The column was set to a temperature of 55°C.
  • a Waters XSELECT HSS T3 2.5 ⁇ m, 50 ⁇ 2.1 mm column was used with a gradient (0.8 mL/min flow rate) starting at 100% mobile phase A (0.1% formic acid in water) to 95% mobile phase B (0.1% formic acid in acetonitrile).
  • Cells were lysed on ice, with 50 uL 1X CST lysis buffer +1mM PMSF for 5 minutes. After 5 minutes, cells were removed using a scraper and transferred to cold 1.5 mL tubes, and centrifuged for 10 min, 4°C, 14,000 x g. Supernatant was gently removed and snap frozen in liquid nitrogen. Protein concentration of the lysate was determined using Bradford Reagent (analyzed using SpectraMax M2E) and diluted to 1 mg/mL or pERK or 1.5 mg/mL for pMEK analysis.
  • Test compounds were dissolved in DMSO to a concentration of 10 mM and further diluted to 100 ⁇ M using acetonitrile. Liver microsomes from selected species were incubated in duplicate with the test compound at a final concentration of 1 ⁇ M in 0.1 M potassium phosphate buffer (pH 7.4) containing 3.3 mM MgCl2, 0.5 mg/ml microsomal protein, in the presence or absence of NADPH (1 mM). Incubations were performed at 37°C in a total volume of 500 ⁇ l. Control incubations with reference substances were included for each experiment.
  • Capacity factors data (k’ _ (tr – _t0)/t0) obtained at various amounts of methanol were extrapolated to 0% methanol and k’w values are determined using a linear procedure. ElogDoct (7.4) is calculated using a series of reference standards with known LogD values. Each experiment was performed in triplicate. [0780] All sample analysis was performed using an Agilent HPLC-system equipped with an autosampler, a binary pump, a column compartment and a diode array detector.
  • PVDF polyvinylidene fluoride
  • GIT-0 gastrointestinal tract lipid formulation
  • pION acceptor sink buffer
  • Table 7 illustrates a A549 (KRAS G12S) pERK Dose Resposne study.
  • Table 7 Attribute Reference 1 pERK IC50 (A549) [12-point dose] 72 nM
  • the results of a phospo ERK Screen in A549 (KRAS G12S) study is described in FIG. 1, together with the Mouse and Human Microsomal stability and the phosphorylated-ERK IC50 values in A549 cells.
  • EXAMPLE 38 Drug Profiles [0785] Table 8 describes the attributes of two compounds and a reference compound.
  • A549 (Cat No. CCL-185) cell line was obtained from American Type Culture Collection (ATCC). They were grown in T75 flasks in DMEM containing 10% FBS and Pen-Strep. at 37 o C in a humidified, 5% CO2 incubator. The adherent cells were grown to about 90% confluency, culture medium was aspirated, and the cell layer was rinsed with PBS.
  • Protein standards were used (0 – 20 mg/mL) to calculate protein concentrations of test samples.
  • 20 ⁇ g of protein was mixed with 5 ⁇ l of 4X Laemmle’s sample buffer and 1 ⁇ l of 0.4 M DTT in a volume of 20 ⁇ l made up with lysis buffer. All samples were heated at 95 o C for 5 minutes, cooled to room temperature and spun down. Protein samples were loaded onto 4-12% polyacrylamide gels and run at 100V for about 1.5 hours till the blue dye reached the bottom. After the run, gel was removed and protein transfer was done using iBlot for 7 minutes, as per manufacturer’s recommendations. After the transfer, nitrocellulose membrane was incubated on a shaker in 5 mL of blocking buffer at room temperature for 1hr.
  • the blot was then incubated overnight on a shaker in 5 ml of blocking buffer containing 0.2% Tween-20 and primary antibody, at room temperature.
  • Anti- phospho-STAT3 antibody was used at a dilution of 1:500, the other 3 primary antibodies were used at a dilution of 1:1000.
  • the blot was washed 3 times for 10 min each with 10 mL of TBST followed by incubation on a shaker in 5 ml of blocking buffer containing 0.2% Tween-20 and 0.5 ⁇ l of the IRDye labeled secondary antibodies, diluted 1:10000, at room temperature for 1hr.
  • Antibodies Phospho-STAT3 (S727), mouse polyclonal antibodies were obtained from BD Biosciences (Cat No. 612542), following 3 antibodies were obtained from Cell Signaling Technologies. Anti-STAT3, rabbit monoclonal antibodies (Cat No. 12640), Anti-ERK, mouse monoclonal antibodies (Cat No: 9107), and Anti-phospho-ERK, rabbit monoclonal antibodies (Cat No. 4377).
  • Secondary antibodies IRDye 800CW goat anti-rabbit antibodies (LICOR Cat No. 926-32211), IRDye 680RD goat anti-rabbit antibodies (LICOR Cat No.
  • Imaging was done using LICOR’s Odyssey imaging system, quantitation was done using their software, Image Studio version 3.1.
  • A549 or 375 cells were treated in duplicate for two hours with compounds at varying concentrations. After two hours, the cells were lysed, snap frozen, and stored at -80 °C. After storage, the lysate was quantified with the Bradford assay.
  • ERK and MEK phosphorylation levels were estimated by taking the ratio of phospho-protein to total protein and normalizing to the DMSO control. The results of this study are described in Tables 9-11. Table 9.
  • the cells were lysed, snap frozen, and stored at -80 °C. After storage, the lysate was quantified with the Bradford assay. Before the lysates were prepared to run on the Jess, the lysates were diluted to 1 mg/mL for pERK analysis and 1.5 mg/mL pMEK analysis using quantitative Western blotting. ERK and MEK phosphorylation levels were estimated by taking the ratio of phospho-protein to total protein and normalizing to the DMSO control. The results of this study are described in Tables 12-13. Table 12.
  • the results of a phospho-ERK Screen in A549 (KRAS G12S) study is described in Table 15 below. Table 15.
  • A549, A375 or SK-MEL-2 Melanoma Model cells were treated in duplicate for two hours with compounds at varying concentrations.
  • the cells were lysed, snap frozen, and stored at -80 °C. After storage, the lysate was quantified with the Bradford assay. Before the lysates were prepared to run on the Jess, the lysates were diluted to 1 mg/mL for pERK analysis and 1.5 mg/mL pMEK analysis using quantitative Western blotting. ERK and MEK phosphorylation levels were estimated by taking the ratio of phospho-protein to total protein and normalizing to the DMSO control. The results of this study are described in FIG. 2A-2C.
  • EXAMPLE 43 pERK:total ERK Ratio and pMEK:total MEK Ratio [0795] Compound 274 was evaluated at 100 nM doses for impact on pERK:tERK and pMEK:tMEK levels across a panel of 9 melaoma tumor models compared to Selumetinib and Binimetinib. The nine models are described in Table 16. Materials and methods are described for in EXAMPLE 35 with the tumor models detailed in Table 16. Table 16 Model HRAS NRAS BRAF NF1 MM415 p.Q61L MEL-JUSO p.G13D p.Q61L p .
  • Compound 274 is a dual-MEK inhibitor due to the reductions in both pERK and pMEK obvserveed across RAS mutant and NF-1 loss of function (LoF) tumor models.
  • Table 17. pERK/tERK ratio normalized to DMSO Cell line Binimetinib Selumetinib Compound 274
  • Table 18. pM EK/tMEK ratio normalized to DMSO Cell line Binimetinib Selumetinib Compound 274 MEL-JUSO 3.291 2.988 0.548 SK-MEL-30 2.649 2.177 0.609 [079 q , nd DSMZ.
  • 3D- Tumor Growth Assay 3D-TGA sensitivity (green) defined as IC 50 ⁇ 10uM in 72-hour ECM- based assay with %EdU readout, and IC50 ⁇ 10uM considered resistant.
  • mice Female mice (BALC/c) received a single dose (3 mice/treatment) of vehicle (10% 1N HCl : 90% [20% Captisol in saline pH adjusted to 5.0 ⁇ 0.1] adjusted to pH 3.0 ⁇ 0.1) containing 5 x compounds at 10 mg/kg by gavage (p.o.) and at 0.5, 1, 2, 4, 8, 12 and 24- hour post dose humanely euthanized.
  • vehicle 10% 1N HCl : 90% [20% Captisol in saline pH adjusted to 5.0 ⁇ 0.1] adjusted to pH 3.0 ⁇ 0.1
  • mice received either vehicle (10% 1N HCl : 90% [20% Captisol in saline pH adjusted to 5.0 ⁇ 0.1] adjusted to pH 3.0 ⁇ 0.1), or vehicle containing compounds at 25, 50,75, 100, 125, 150, 175 and 200 mg/kg by gavage (p.o.) either QD or BID for 8 days.
  • Tumor growth inhibition (TGI) was calculate and represented in FIG. 3 and 4 obtained with Compoudn 274.
  • FIG. 6 illustrates a graph of a colon-26 syngeneic CRC tumor mouse model (BID) study with Compound 274.
  • FIG. 7 illustrates a graph of a colon-26 syngeneic CRC tumor mouse model (QD) study with Compound 274.
  • EXAMPLE 46 Human ether-a-go-go related gene (hERG) channel [0802]
  • hERG Ion channel block of IC50 values were deteremined. The values are described in Table 21.
  • CHO cells were cultured in Ham’s F-12 supplemented with 10% fetal bovine serum, 100 U/mL penicillin G sodium, 100 ⁇ g/mL streptomycin sulfate and 400 ug/mL Zeocin. Before testing, cells in culture dishes were rinsed with Hank’s Balanced Salt Solution, detached with accutase.
  • hERG current was measured using stimulus voltage patterns with fixed amplitudes: activation pre-pulse (TP1) to +40 mV for 2 s and test pulse (TP2) to -40 mV for 2 s from a holding potential of -80 mV.
  • hERG current was measured as the outward peak current at TP2 (tail current).
  • the stimulation was repeated with 0.1 Hz frequency during 2 min as baseline and 5 min after TA application.
  • Data acquisition and analyses were performed using the SP384PE system operation software. The decrease in current amplitude after TA application was used to calculate the percent block relative to control. Results for each TA concentration (n ⁇ 2) were averaged; the mean and standard error values were calculated, and used to generate dose-response curves.
  • Table 21 Compound ID IC50 ( ⁇ M) 255 18.1 256 216 Membrane Permeability (Caco-2) [0803] In this study, membrane permeability (Caco-2) was determined. The results of this study are described in Table 22. Table 22 A-B Mean Papp A-B Mean Papp B-A Mean Efflux Talinolol VERA 0.940 2.39 2.55 Lower Warfarin 40.5 20.8 0.513 Higher Hepatocyte Stability [0804] In this study, human, cynomolgus monkey, beagle dog, wistar hannover rat, and balb/c mouse hepatocyle stability was determined.
  • Table 23 describes the results of the human hepatocyte stability at 2 ⁇ M concentration and 0.5 million cells/ml for 120 min.
  • Table 24 describes the results of the cynomolgus monkey hepatocyte stability at 2 ⁇ M concentration and 0.5 million cells/ml.
  • Table 25 describes the results of the beagle dog hepatocyte stability at 2 ⁇ M concentration and 0.5 million cells/ml.
  • Table 26 describes the results of the wistar hannover rat hepatocyte stability at 2 ⁇ M concentration and 0.5 million cells/ml.
  • Table 27 describes the results of the balb/c mouse hepatocyte stability at 2 ⁇ M concentration and 0.5 million cells/ml.
  • Table 31 describes the results of the wistar hannover rat plasma stability at 2 ⁇ M concentration and anti-coagulant K2-EDTA.
  • Table 32 describes the results of the balb/c plasma stability at 2 ⁇ M concentration and anti-coagulant K2-EDTA.
  • Table 28 Compound ID T1/2 (min) % Remaining at T120
  • Table 29 Compound ID T1/2 (min) % Remaining at T120 120 > 120 107.3 124 1279 95.9
  • Table 30 Compound ID T1/2 (min) % Remaining at T120 274 > 120 100.5 Propantheline 292 71.0
  • Table 32 Compound ID T1/2 (min) % Remaining at T120 146 747 89.8 161 317 74.9 50 Human Liver Microsomes (MLM) CYP Inhibition [0806] In this study, human liver microsomes as a mean percentage CYP inhibition at 10 ⁇ M were detere
  • KRAS is the most frequently altered RAS gene ( ⁇ 85%) and is often mutated in pancreatic ductal adenocarcinoma (PDAC; 95%), non-small cell lung cancer (NSCLC; 40%) and colorectal cancer (CRC; 45%).
  • PDAC pancreatic ductal adenocarcinoma
  • NSCLC non-small cell lung cancer
  • CRC colorectal cancer
  • MEK inhibitors could broaden the potential for immune therapy in RAS-mutant tumors, but they have been largely ineffective in this setting as monotherapy.
  • the short-lived Dual-MEK inhibitor, compound 274 is active across multiple MAPK-driven tumor models both as a single agent and in combination with checkpoint inhibitors (CPI).
  • CPI checkpoint inhibitors
  • Compound 274 was evaluated in a series of preclinical in vitro and in vivo models enriched for activation mutations that increase MAPK pathway signaling.
  • Cell-based 2D biochemical and 3D pharmacologic assays were performed along with multiple in vivo studies in RAS mutant and wildtype models: [0810] (1.) Colon 26, a KRAS G12D CRC syngeneic model, [0811] (2.) A549, a KRAS G12S NSCLC xenograft model, [0812] (3.) CT-26, a KRAS G12D syngeneic model and [0813] (4.) MC38, a RAS wild-type syngeneic model.
  • FIG. 8A illustrates graphs depicting compound 274 with a short plasma and tumor PK half-life in vivo.
  • FIG. 8B illustrates graphs depicting compound 274 with a short plasma and tumor PK half-life in vivo.
  • Colon 26 tumor-bearing syngeneic BALB/c mice Pharmacokinetics (PK) of compound 274 in Colon 26 tumor bearing syngeneic BALB/c mice (timepoints: 0.083, 0.5, 1, 2, 8 hours after 10 mg/kg p.o.
  • Table 34 provides data from this portion of the study.
  • Table 34 Cmpd 274 Cmpd 274 Cmpd 274 Cmpd 274 Time 2 k 1 k 2 k i 1 k [0815]
  • Table 35 illustrates differentiating characteristics of 1 st , 2 nd , and 3 rd generation MEK inhibitors.
  • Table 35 Cmax or MEKi MEK R gs Cmax or MEKi pMEK Response Drug Chronic or Cyclic Example Drugs 4 .
  • FIGs 9A-B Colon 26 (KRAS G12D ) syngeneic colorectal tumor model in immune competent BALB/c mice;
  • FIG. 10A-10B illustrate graphs depicting MEKi, ⁇ -PD-1, ⁇ -CTLA-4 alone and combinations in CT-26/MC38.
  • FIG.10A CT-26 (KRAS G12D ) syngeneic colorectal tumor model in immune competent BALB/c mice (note: monotherapy and combinations were inactive in athymic nude CT-26 model – data not shown).
  • FIG. 10B MC38 (RAS wild-type ) syngeneic colorectal tumor model in immune competent C57BL/6 mice (note: immune compromised model not evaluated).
  • Tables 37 and 38 provides data from this portion of the study.
  • Table 37 – Cmpd 274 checkpoint inhibitor in KRAS G12D CT-26 Dose ⁇ PD-1 ⁇ CTLA- Cmpd Cmpd 274 Cmpd 274 Schedule Veh k BI 4 BI 274 PD 1 TLA 4 han
  • Table 38 – Cmpd 274 checkpoint inhibitor in RAS wild-type MC38 Dose ⁇ PD-1 ⁇ CTLA- Cmpd Cmpd 274 Cmpd 274 S h d l 4 QD 60 0/12 0/12* 4/12 2/12 QD 30 0/12 0/12* 5/12 2/12 an ase on e a a rom ese s u es, compoun re uce p and pMEK across all RAS mutant models tested.
  • Compound 274 displayed activity across multiple RAS and RAF-mutant tumor models, and when combined with PD-1 or CTLA-4 checkpoint inhibitors at well tolerated, sub-MED dose levels, significant survival benefit was observed (p-values: ⁇ 0.05 to ⁇ 0.0001; CT-26). These data suggest that moderated, cyclic inhibition of MEK in combination with CPIs may improve survival times versus monotherapy in MAPK-activated tumors. Antitumor responses with compound 274 +/- CPIs in an immune compromised CT-26 model at the same doses, combinations and schedules were not observed, suggesting that moderated, cyclic disruption of the MAPK pathway can enhance CPI-dependent adaptive antitumor immunity and improve overall MEKio combination tolerability.

Landscapes

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

Abstract

La présente divulgation concerne des méthodes de traitement d'un patient atteint d'un cancer à mutation RAS ou RAF, comprenant l'administration d'un composé divulgué, comprenant des méthodes de traitement comprenant l'administration d'un composé de formule (IV) ou d'un sel pharmaceutiquement acceptable de celui-ci, en association avec un inhibiteur de point de contrôle immunitaire
PCT/US2023/079156 2022-11-09 2023-11-08 Inhibiteurs oncologiques immunitaires de mek et utilisations thérapeutiques associées WO2024102859A1 (fr)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US202263383067P 2022-11-09 2022-11-09
US63/383,067 2022-11-09
PCT/US2023/060181 WO2023133472A1 (fr) 2022-01-06 2023-01-05 Inhibiteurs oncologiques immunitaires de mek et leurs utilisations thérapeutiques
USPCT/US2023/060181 2023-01-05
US202363511789P 2023-07-03 2023-07-03
US63/511,789 2023-07-03

Publications (1)

Publication Number Publication Date
WO2024102859A1 true WO2024102859A1 (fr) 2024-05-16

Family

ID=91033585

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/079156 WO2024102859A1 (fr) 2022-11-09 2023-11-08 Inhibiteurs oncologiques immunitaires de mek et utilisations thérapeutiques associées

Country Status (1)

Country Link
WO (1) WO2024102859A1 (fr)

Similar Documents

Publication Publication Date Title
US20230119327A1 (en) Mek inhibitors and therapeutic uses thereof
KR102477960B1 (ko) 항증식성 화합물 및 이의 사용 방법
CA3144859A1 (fr) Derives de quinazoline-4-one utiles pour le traitement de maladies et de troubles associes a braf
CN105339008A (zh) 用于治疗癌症的包括tor激酶抑制剂和n-(3-(5-氟-2-(4-(2-甲氧基乙氧基)苯基氨基)嘧啶-4-基氨基)苯基)丙烯酰胺的组合疗法
TW201534305A (zh) 使用組合療法治療癌症之方法
JP6147246B2 (ja) Akt及びmek阻害剤化合物の組み合わせ、及び使用方法
TW202045480A (zh) 化合物及使用方法
TW201938149A (zh) 用於治療癌症的選擇性組蛋白去乙醯酶3 (hdac3)抑制劑及免疫治療劑之組合
US20240076287A1 (en) Solid forms of a cdk2 inhibitor
ES2895419T3 (es) Formas sólidas de (1S,4S)-4-(2-(((3S,4R)-3-fluorotetrahidro-2H-piran-4-il)amino)-8-((2,4,6-triclorofenil)amino)-9H-purin-9-il)-1-metilciclohexan-1-carboxamida y métodos para su uso
JP2024505680A (ja) 異常な細胞成長を処置するための併用療法
JP7291839B2 (ja) 固体形態のcdk4阻害薬
BR112017010849B1 (pt) Triazolopiridazina deuterada como um modulador quinase, seu processo de preparação, composição farmacêutica, usos e combinação
WO2024102859A1 (fr) Inhibiteurs oncologiques immunitaires de mek et utilisations thérapeutiques associées
WO2023133472A1 (fr) Inhibiteurs oncologiques immunitaires de mek et leurs utilisations thérapeutiques
WO2019126568A1 (fr) Analogues de nifuroxazide et leurs utilisations thérapeutiques
RU2812929C1 (ru) Ингибиторы mek и их терапевтическое применение
TWI837478B (zh) 用於治療braf相關的疾病和病症之化合物
RU2814662C1 (ru) Соединения 4-оксо-3,4-дигидрохиназолинона для лечения braf-ассоциированных заболеваний и нарушений
TW202214577A (zh) 用於治療braf相關的疾病和病症之化合物
CN117561255A (zh) 作为mek抑制剂的3,4-二氢-2,7-萘啶-1,6(2h,7h)-二酮化合物
CN117729923A (zh) 治疗异常细胞生长的联合疗法
NZ629859B (en) Methods for treating cancer using tor kinase inhibitor combination therapy