WO2020206034A1 - Cell cycle inhibiting compounds for the treatment of medical disorders - Google Patents

Cell cycle inhibiting compounds for the treatment of medical disorders Download PDF

Info

Publication number
WO2020206034A1
WO2020206034A1 PCT/US2020/026262 US2020026262W WO2020206034A1 WO 2020206034 A1 WO2020206034 A1 WO 2020206034A1 US 2020026262 W US2020026262 W US 2020026262W WO 2020206034 A1 WO2020206034 A1 WO 2020206034A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
alkyl
pharmaceutically acceptable
aryl
heteroaryl
Prior art date
Application number
PCT/US2020/026262
Other languages
French (fr)
Inventor
Jay Copeland Strum
Original Assignee
G1 Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by G1 Therapeutics, Inc. filed Critical G1 Therapeutics, Inc.
Publication of WO2020206034A1 publication Critical patent/WO2020206034A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains three hetero rings
    • C07D487/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains three hetero rings
    • C07D487/20Spiro-condensed systems

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

This invention is in the area of cell cycle inhibiting compounds for the treatment of disorders involving abnormal cellular proliferation, and include CDK2 and CDK9 inhibitors for medical therapy.

Description

CELL CYCLE INHIBITING COMPOUNDS LOR THE TREATMENT
OF MEDICAL DISORDERS
CROSS REFERNCE TO RELATED APPLICATIONS
This application claims the benefit of priority of U.S. Provisional Application 62/827,703, filed on April 1, 2019, U.S. Provisional Application 62/827,692, filed on April 1, 2019, U.S. Provisional Application 62/870,357, filed on July 3, 2019, and U.S. Provisional Application 62/870,351, filed on July 3, 2019, the entirety of each of which are hereby incorporated by reference for all purposes.
FIELD OF THE INVENTION
This invention is in the area of pyrimidine-based compounds for the treatment of disorders involving abnormal cellular proliferation, including but not limited to the treatment of cancers and tumors.
BACKGROUND
In normal tissue, cellular proliferation is generally restricted to cells that are required to replenish the tissue. Once cells have terminally differentiated, they have a specialized function and no longer divide. Most tissues are made of non-dividing cells. Thus, normal cell proliferation is tightly controlled to ensure that only the necessary cells divide. There is also a careful balance between cell division and programmed cell death (apoptosis).
Cell division, sometimes referred to as the cell cycle, has four phases: Gi phase (synthesis of various enzymes required for DNA replication), S phase (DNA replication producing two identical sets of chromosomes), G2 (significant protein synthesis, including production of microtubules) and M phase (nuclear division, cytoplasmic division and formation of new cell membrane). Cell division also includes a complex system of cell signaling networks that allow cells to interpret information from numerous extracellular signals, including through receptor proteins, inflammatory factors and pro-apoptotic and anti-apoptotic signals. Dysfunctional signals include those from genetic mutation, infection, exposure to environmental factors including toxins, system stress, autoimmune disorders, and inflammation.
A range of disorders can occur when the process of cell proliferation becomes dysfunctional, including benign grow ths neoplasms, tumorigenesis, cancerogenesis, autoimmune disorders, inflammatory disorders graft- versus-host rejection, and fibrotic disorders.
A number of broad-spectrum anti -neoplastic agents have been developed. Cytoskeletal drugs like paclitaxel target tubulin to arrest mitotic cell division and are used to treat a variety of cancers including ovarian, breast, lung, pancreatic, and testicular tumors (See e.g., Jordan, Wilson, Nature Reviews Cancer (2004) 4: 253-265). Organometallic-based drugs such as cisplatin have been used to treat lymphomas, sarcomas, germ cell tumors, and some carcinomas including bladder, small cell lung cancer, and ovarian cancer. Cisplatin has the ability to bind nitrogenous bases and cause extensive DNA cross-linking that ultimately leads to apoptosis (See e.g., Siddick, Oncogene (2003) 22: 7265-7279). Intercalating and alkylating agents have also been extensive use in the clinic for the treatment of various neoplasms, however, the global toxicity associated with these drugs presents a critical concern for patients requiring long-term therapy.
Palbociclib (PD-033299; Ibrance) is sold by Pfizer for the treatment of estrogen- positive, HER2 -negative breast cancer in combination with letrozole. The compound inhibits
CDK4 and CDK6. The structure of palbociclib is:
Figure imgf000004_0001
Abemaciclib (LY2835219) is a CDK 4/6 inhibitor currently in human clinical trials for the treatment of various types of cancers. It is in a phase III trial for stage IY non-small cell lung carcinoma; in combination with Fulvestrant for women with breast cancer; and with either anastrozole or letrozole for first line treatment of breast cancer. The structure of abemaciclib is:
Figure imgf000004_0002
Ribociclib (LeeOl l ; Kisqali), is a CDK 4/6 inhibitor approved for use in combination with an aromatase inhibitor to treat some metastatic breast cancers, and is in clinical trials for the treatment of certain other tumors. The structure of ribociclib is:
Figure imgf000005_0001
Lerociclib is an oral, selective CDK4/6 inhibitor in clinical development by G1
Therapeutics for use in combination with other targeted therapies in multiple oncology indications. Lerociclib is currently being evaluated in two Phase 1/2 clinical trials: a trial in combination with fulvestrant (Faslodex®) for patients with estrogen receptor-positive, HER2- negative (ER+, HER2-) breast cancer (NCT02983071) and a trial in combination with osmirtinib (Tagrisso®) in EGFRm non-small cell lung cancer. Lerociclib has the structure:
Figure imgf000005_0002
Trilaciclib is a selective CDK4/6 inhibitor in clinical development by G1 Therapeutics for use as a first-in-class myelopreservation therapy designed to improve outcomes of patients who receive chemotherapy by preserving hematopoietic stem and progenitor cell (HSPC) and immune system function. Trilaciclib is a short-acting intravenous CDK4/6 inhibitor administered prior to chemotherapy and is currently being evaluated in four randomized Phase 2 clinical trials, including in first-line SCLC trials in combination with a chemotherapy regimen of etoposide and carboplatin (NCT02499770); and in first-line SCLC trial in combination with the same chemotherapy regimen and the checkpoint inhibitor Tecentriq® (atezolizumab). Trilaciclib has the structure:
Figure imgf000005_0003
SHR 6390 is a selective CDK4/6 inhibitor in clinical development by Jiangsu HengRui Medicine Co., Ltd. SHR6390 is currently being investigated in in combination with letrozole or anastrozole or fulvestrant in patients with HR-positive and HER2-negative advanced breast cancer.
Various other pyrimidine-based agents have been developed for the treatment of hyperproliferative diseases. U.S. Patent Nos. 8,822,683; 8,598,197; 8,598,186; 8,691,830; 8,829,102; 8,822,683; 9,102,682; 9,260,442; 9,481,691; 9,499,564; 9,957,276; 10,189,849;
10,189,850; and 10,189,851; filed by Tavares and Strum and assigned to G1 Therapeutics describe a class of N-(heteroaryl)-pyrrolo[3,2-d]pyrimidin-2-amine cyclin dependent kinase inhibitors including those of the formula (with variables as defined therein):
Figure imgf000006_0001
U.S. Patent Nos. 9,464,092; 9,487,530; 9,527,857; 10,076,523; 10,085,992; and 10,434,104 which are also assigned to G1 Therapeutics describe the use of the above pyrimidine-based agents in the treatment of cancer.
WO 2013/148748 (U.S.S.N. 61/617,657) titled“Lactam Kinase Inhibitors”, WO 2013/163239 (U.S.S.N. 61/638,491) titled“Synthesis of Lactams” and WO 2015/061407 filed by Tavares and also assigned to G1 Therapeutics describes the synthesis of N-(heteroaryl)- pyrrolo[3,2-d]pyrimidin-2-amines and their use as lactam kinase inhibitors.
Other publications include the following. WO 2014/144326 filed by Strum et al. and assigned to G1 Therapeutics describes compounds and methods for protection of normal cells during chemotherapy using pyrimidine-based CDK4/6 inhibitors. WO 2014/144596 filed by Strum et al. and assigned to G1 Therapeutics describes compounds and methods for protection of hematopoietic stem and progenitor cells against ionizing radiation using pyrimidine-based CDK4/6 inhibitors. WO 2014/144847 filed by Strum et al. and assigned to G1 Therapeutics describes HSPC-sparing treatments of abnormal cellular proliferation using pyrimidine-based CDK4/6 inhibitors. WO 2014/144740 filed by Strum et al. and assigned to G1 Therapeutics describes highly active anti-neoplastic and anti-proliferative pyrimidine-based CDK 4/6 inhibitors. WO 2015/161285 filed by Strum et al. and assigned to G1 Therapeutics describes tricyclic pyrimidine-based CDK inhibitors for use in radioprotection. WO 2015/161287 filed by Strum et al. and assigned to G1 Therapeutics describes tricyclic pyrimidine-based CDK inhibitors for the protection of cells during chemotherapy. WO 2015/161283 filed by Strum et al. and assigned to G1 Therapeutics describes tricyclic pyrimidine-based CDK inhibitors for use in HSPC-sparing treatments of RB-positive abnormal cellular proliferation. WO 2015/161288 filed by Strum et al. and assigned to G1 Therapeutics describes tricyclic pyrimidme-based CDK inhibitors for use as anti-neoplastic and anti-proliferative agents. WO 2016/040858 filed by Strum et al. and assigned to G1 Therapeutics describes the use of combinations of pyrimidine-based CDK4/6 inhibitors with other anti-neoplastic agents. WO 2016/040848 filed by Strum et al. and assigned to G1 Therapeutics describes compounds and methods for treating certain Rb-negative cancers with CDK4/6 inhibitors and topoisomerase inhibitors. WO 2018/005860, WO 2018/005533, and WO 2018/005863 filed by Strum and assigned to G1 Therapeutics describes various CDK inhibitors. WO 2018/106739 filed by Sorrentino et al., and assigned to G1 Therapeutics describes the use of CDK4/6 inhibitors with specific dosage regimens. WO 2018/156812 filed by Strum et al., and assigned to G1 Therapeutics describes the use of CDK4/6 inhibitors to treat EGFR-driven cancer. WO 2019/199883 filed by Strum et al. and assigned to G1 Therapeutics describes compounds and methods for treating chemotherapy resistant cancer. WO 2019/136451 filed by Beelen et al. and assigned to G1 Therapeutics describes dosage regimes for the administration of G1T38. WO 2019/136244 filed by Strum et al. and assigned to G1 Therapeutics describes additional compounds for inhibiting CDKs.
Despite research in the area of cell cycle inhibiting compounds to treat abnormal cellular proliferation in a host, for example, a human, given the seriousness of these diseases, there remains a need to identify new compounds that can meet this medical need. It is another goal to identify cell cycle inhibiting compounds that can be use in combination or alternation (for example, for periodic dosage switching) with already approved cell cycle inhibiting compounds.
Therefore, it is an object of the present invention to provide new compounds, methods, compositions and processes of manufacture to inhibit undesired cell cycling in a host, for example, a human, wherein the compounds can be used to treat abnormal cellular proliferation. It is yet another aspect of the invention to provide compounds, methods and compositions that can be used to treat cell cycle disorders in cells that are naturally or have become resistant to other therapies.
SUMMARY
The invention provides a therapeutically active compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula
XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, or Formula XXIV or a pharmaceutically acceptable salt or composition thereof. In one embodiment, the active compound or its salt, composition, isotopic derivative or prodrug thereof is used in an effective amount to treat a medical disorder involving abnormal cellular proliferation, including a tumor or cancer, in a host, typically a human, in need thereof.
Based on this discovery, compounds and methods are presented for the treatment of a patient with a proliferative disorder including a tumor or cancer that includes administering an effective amount of one or a combination of the compounds described herein to a patient in need thereof, optionally in a pharmaceutically acceptable carrier. In certain embodiments, the antiproliferative disorder is selected from a benign growth, neoplasm, tumor, cancer, autoimmune disorder, inflammatory disorder, graft-versus-host rejection and a fibrotic disorder. In a typical embodiment, the patient is a human.
The present invention also provides advantageous methods to treat a patient with a selective CDK4/6 inhibitor resistant proliferative order, for example a tumor or cancer, which include administering an effective amount of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula
XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, or Formula XXIV, or a pharmaceutically acceptable composition, salt, isotopic derivative, or prodrug thereof. Despite the development of selective CDK4/6 inhibitors, MYC- dnven tumor types with retinoblastoma (Rb) protein loss or high expression levels of cyclin E, such as triple negative breast cancer (TNBC) and small cell lung cancer (SCLC) are difficult to treat due to an intrinsic or primary resistance to existing selective CDK4/6 inhibitors. In addition, certain cancers, despite being Rb-positive, are intrinsically resistant to the effects of selective CDK4/6 inhibitors. In addition, certain cancers that have an intact Rb-pathway may otherwise be intrinsically resistant to selective CDK4/6 inhibitor due to the presence of other genetic or phenotypical abnormalities. For example, it is estimated that 40% of uterine, 20% of ovarian, 15% of bladder, 20% or prostate, and 15% of breast cancers may be intrinsically resistant to selective CDK4/6 inhibition due to the up regulation of Cyclin E, despite intact Rb. See, e.g., Knudsen et al., The Strange Case of CDK4/6 Inhibitors: Mechanisms, Resistance, and Combination Strategies. Trends Cancer. 2017 Jan; 3(1): 39-55. Furthermore, certain cancers, for example ER+ breast cancers, are capable of acquiring resistance to selective CDK4/6 inhibitors during the course of selective CDK4/6 inhibitor therapy, for example by upregulation of cyclin E, which allows G1 to S cell cycle progression through CDK2. The compounds described herein effectively inhibit cell-cycle progression in cancer cells that are intrinsically resistant to, susceptible to acquiring resistance to, or have become resistant to selective CDK4/6 inhibitors.
The active compounds described herein act as inhibitors of a cyclin-dependent kinase (CDK), for example through inhibition of CDK2 and/or CDK4 and/or CDK6, or a combination thereof providing for cell-cycle inhibition in a replicating cell. Unlike selective CDK4/6 inhibitors, however, certain of the active compounds herein are capable of inhibiting cells that are or have become selective CDK4/6 inhibitor resistant by their ability to also inhibit CDK2, thus providing additional cell-cycle inhibition mechanisms. This characteristic is especially useful in inhibiting the cell-cycle progression of cancers or other proliferative disorders that are or have become Rb-negative, this escaping CDK4/6 cell-cycle control.
In one aspect of the present invention a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, or Formula VI is provided:
Figure imgf000009_0001
Figure imgf000010_0001
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof;
wherein:
y is 0, 1, 2, 3, or 4;
x is 0, 1, 2, 3, or 4;
L is selected from -S(O)-, -S(O)2-, -C(O)-, and -C(S)-;
X1, X2, X3, X4, and X5 are independently selected from N, CH, CR2, and CR4; wherein at least one of X1, X2, X3, X4, and X5 is CR2; and wherein no more than 2 of X1, X2, X3, X4, and X5 are selected to be N;
X6, X . X8, X9, and X10 are independently selected from N, CH, CR2, and CR4; and wherein no more than 2 of X6, X7, X8, X9, and X10 are selected to be N;
each R1 is independently hydrogen, alkyl, ary l, cycloalkyl, haloalkyl, heteroaryl, alkyl-hydroxyl, or heterocycle, wherein two R1s on adjacent ring atom(s) or on the same ring atom may come together with the ring atom(s) to which they are attached to optionally constitute a 3, 4, 5, 6, 7, or 8-membered cycloalkyl or 4, 5, 6, 7, or 8-membered heterocycle that has 1 , 2, or 3 heteroatoms selected from N, O, and S; wherein the cycloalkyl or heterocycle formed by combining two R1s with the atom(s) to which they are attached can be optionally substituted with 1 or 2 substituents independently selected from R50;
in an alternative embodiment each R1 is independently halogen, -OR7, hydrogen, alkyl, aryl, cycloalkyl, haloalkyl, heteroaryl, alkyl-hydroxyl, or heterocycle, wherein two R1s on adjacent ring atom(s) or on the same ring atom may come together with the nng atom(s) to which they are attached to optionally constitute a 3, 4, 5, 6, 7, or 8-membered cycloalkyl or 4, 5, 6, 7, or 8-membered heterocycle that has 1, 2, or 3 heteroatoms selected from N, O, and S; wherein the cycloalkyl or heterocycle formed by combining two R1s with the atom(s) to which they are attached can be optionally substituted with 1 or 2 substituents independently selected from R50;
each R2 is independently -NR14C(O)R6, -NR14S(O)R6, -NR14S(O)2R6, -NR14C(S)R6, -0C(O)R6, -OS(O)R6, -OS(O)2R6, -OC(S)R6, -C(O)R6; -C(S)R6, -S(O)R6, or -S(O)2R6;
R3 is selected from:
Figure imgf000011_0001
and alkyl;
Figure imgf000011_0003
in an alternative embodiment R3 is
Figure imgf000011_0002
each R4 is independently hydrogen, alky l, aryl, cycloalkyl, haloalkyl, heteroaryl, heterocycle, halogen, cyano, -OR14, -NR14R15, -NR14C(O)R6, -NR14S(O)R6, -NR14S(O)2R6, -NR14C(S)R6, -OC(O)R6, -OS(O)R6, -OS(O)2R6, -OC(S)R6, -C(O)R6; -C(S)R6, -S(O)R6, or -S(O)2R6;
R5 is hydrogen, alkyl, aryl, cycloalkyl, haloalkyl, heteroaryl, heterocycle, -C(O)R6, -C(S)R6, -S(O)R6, -S(O)2R6, -C(O)NR6R6, -C(S)NR6R6, -S(O)NR6R6, -S(O)2NR6R6, -C(O)OR6, -C(S)OR6, -S(O)OR6, or -S(O)20R6;
or R5 is hydrogen, alkyl, ary l cycloalkyl, haloalkyl, heteroaryl, heterocycle, -C(O)R6, -C(S)R6, -S(O)R6, -S(O)2R6, -C(O)NR7R7, -C(S)NR7R7, -S(O)NR7R7, -S(O)2NR7R7, -C(O)OR7, -C(S)OR7, -S(O)OR7, or -S(O)20R7;
each R6 is selected independently from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocy cle, aryl, heteroaryl, NR R7, and OR7 each of which R6 except hydrogen, NR7R7, and OR7 is optionally substituted with 1, 2, 3, or 4 R8 groups;
in one embodiment each R6 is selected independently from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycle, aryl, and heteroaryl, each of which R6 except hydrogen is optionally substituted with 1, 2, 3, or 4 R8 groups;
each R7 is independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycle, aryl, alkyl-aryl, alkyl-heteroaryl, and heteroaryl each of which R7 except hydrogen is optionally substituted with 1, 2, 3, or 4 R8 groups;
each R8 is independently selected from hydrogen, halogen, haloalkyl, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycle, aryl, heteroaryl, -S(O)2alkyl, NR12R13, alkyl-heteroaryl, alkyl-aryl, and OR12;
R11 is selected from hydrogen, alkyl, -NR12R13, -S(O)alkyl, -SO2alkyl, cycloalkyl, heterocycle, aryl, heteroaryl, alkyl-aryl, and alkyl-heteroaryl;
in an alternative embodiment R11 is selected from halogen, haloalkyl, hydrogen, alkyl, -NR12R13, -S(O)alkyl, -SO2alkyl, cycloalkyl, heterocycle, aryl, heteroaryl, alkyl-aryl, and alkyl-heteroaryl;
each R12 and R13 are independently selected from hydrogen, alkyl, alkenyl, alkynyl, -C(O)alkyl, -C(S)alkyl, aryl, -SO2alkyl, -SOalkyl, heteroaryl, alkyl-aryl, cycloalkyl, heterocycle, and alkyl-heteroaryl;
each R14 and R15 are independently selected from hydrogen, alkyl, alkenyl, alkynyl, -C(O)R6, -C(O)alkyl, -C(S)alkyl, aryl, -SO2alkyl, heteroaryl, heterocycle, -alkyl-aryl, and -alkyl-heteroaryl; and
R50 is selected from hydrogen, amino, -NHR14, -NR14R15, hydroxyl, OR14, and R4. In one embodiment, wherein R4 is halogen, cyano, -OR14, -NR14R15, -NR14C(O)R6, -NR14S(O)R6, -NR14S(O)2R6, -NR14C(S)R6, -OC(O)R6, -OS(O)R6, -OS(O)2R6, or -OC(S)R6; the substituent is not alpha to the heteroatom in the heterocycle. For example, in this
embodiment,
Figure imgf000013_0002
could only be .
Figure imgf000013_0003
In another aspect of the present invention a compound of Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, or Formula XV is provided:
Figure imgf000013_0001
Figure imgf000014_0001
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof;
wherein:
R16 is selected from hydrogen, halogen, alkyl, hydroxyl, alkoxy, and haloalkyl; and wherein all other variables are as defined herein.
In another aspect of the present invention a compound of Formula XVI, Formula XVII, Formula XVIII, and Formula XIX is provided:
Figure imgf000014_0002
or a pharmaceutically acceptable salt, N-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof;
wherein:
R17 is selected from cycloalkyl, heterocycle, heteroaryl, and aryl, each of which R17 is optionally substituted with 1, 2, 3, or 4 substituents independently selected from R2, R4 and
R5;
in one embodiment R17 is selected from:
Figure imgf000015_0001
R18 is selected from: cycloalkyl, heterocycle, and aryl, each of which R18 is substituted with at least one R2 group, and wherein each R18 is optionally substituted with 1, 2, 3, or 4 substituents independently selected from R4 and R5; and
wherein all other variables are as defined herein.
In an alternative embodiment the compound of Formula XIX is:
Figure imgf000015_0002
or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof, wherein all variables are as defined herein. In another aspect of the present invention a compound of Formula XX or FormulaXXI is provided:
Figure imgf000016_0001
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof;
wherein:
R19 is selected from ; and
Figure imgf000016_0002
R20 is selected from alkyl, alkenyl, alkynyl, -C(O)R6, -C(O)alkyl, -C(S)alkyl, aryl, -S02alkyl, heteroaryl, heterocycle, -alkyl-aryl, and -alkyl-heteroaryl;
R21 is selected from hydrogen, halogen, alkyl, -S(O)alkyl, -SO2alkyl, cycloalkyl, heterocycle, aryl, heteroaryl, alkyl-aryl, and alkyl-heteroaryl; and
wherein all other variables are as defined herein.
In certain embodiments R21 is hydrogen or alkyl.
In another aspect of the present invention a compound of Formula XXII, Formula XXIII, or Formula XXIV is provided:
and
Figure imgf000016_0003
Figure imgf000017_0001
or a pharmaceutically acceptable salt. N-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof;
y2 is 0, 1, 2, 3, or 4;
each m is independently 0 or 1;
each n is independently 0, 1, or 2;
each R31 is independently alkyl, aryl, cycloalkyl or haloalkyl, wherein each of said alkyl, cycloalkyl and haloalkyl groups optionally includes heteroatoms O, N, or S in place of a carbon in the chain and two R31s on adjacent ring atoms or on the same ring atom together with the ring atom(s) to which they are attached optionally form a 3-8-membered cycle or two R31s on adjacent ring atoms together with the ring atoms to which they are attached optionally form a 6-membered aryl ring;
R32 is -(alkylene)m-heterocyclo, -(alkylene)m-heteroaryl, -(alkylene)m-NR33R34, -(alkylene)m-C(O)-NR33R34; -(alkylene)m-C(O)-O-alkyl; -(alkylene)m-O-R35, -(alkylene)m-S(O)n-R35, or -(alkylene)m-S(O)n-NR33R34 any of which may be optionally independently substituted with one or more Rx groups as allowed by valance, and wherein two Rx groups bound to the same or adjacent atom may optionally combine to form a ring;
R32* is -(alkylene)m-heterocyclo, -(alkylene)m-heteroaryl, -(alkylene)m-NR33R34, -(alkylene)m-C(O)-NR33R34; -(alkylene)m-C(O)-O-alkyl; -(alkylene)m-S(O)n-R35, or -(alkylene)m-S(O)n-NR33R34 any of which may be optionally independently substituted with one or more Rx groups as allowed by valance, and wherein two Rx groups bound to the same or adjacent atom may optionally combine to form a ring;
or R32* is -alkylene-heterocyclo, -(alkyl ene)m-heteroaryl, -alkylene-NR33R34, -(alkylene)m-C(O)-NR33R34; -(alkylene)m-C(O)-O-alkyl; -alkylene-S(O)n-R35, or -(alkylene)m-S(O)n-NR33R34 any of which may be optionally independently substituted with one or more Rx groups as allowed by valance, and wherein two Rx groups bound to the same or adjacent atom may optionally combine to form a ring;
R33 and R34 at each occurrence are independently:
(i) hydrogen or
(ii) alkyl, cycloalkyl, heterocyclo, aryl, heteroaryl, cycloalkylalkyl, heterocycloalkyl, arylalkyl, or heteroarylalkyl; or R33 and R34 together with the nitrogen atom to which they are attached may combine to form a heterocyclo ring;
R35 is independently:
(i) hydrogen or
(ii) alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclo, aryl, heteroaryl, cycloalkylalkyl, heterocycloalkyl, arylalkyl, or heteroarylalkyl;
Rx at each occurrence is independently selected from halo, cyano, nitro, oxo, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclo, aryl, heteroaryl, arylalky l, heteroarylalkyl, cycloalkylalkyl, heterocycloalkyl, -(alkylene)m-OR35, -(alkylene)m-O-alkylene-OR35, -(alkylene)m-S(O)n-R35, -(alkylene)m-NR33R34, -(alkylene)m-CN, -(alkylene)m-C(O)-R35,
-(alkylene)m-C(S)-R35, -(allky lene)m-C (O)-OR35, -(alkylene)m-O-C(O)-R35,
-(alkylene)m-C(S)-OR35, -(alkylene)m-C(O)-(alkylene)m-NR33R34,
-(alkylene)m-C(S)-NR33R34, -(alkylene)m-N(R33)-C(O)-NR33R34,
-(alkylene)m-N(R33)-C(S)-NR33R34, -(alkylene)m-N(R33)-C(O)-R35,
-(alkylene)m-N(R33)-C(S)-R35, -(alkylene)m-O-C(O)-NR33R34,
-(alkylene)m-O-C(S)-NR33R34, -(alkylene)m-SO2-NR33R34, -(alkylene)m-N(R33)-SO2-R35, -(alkylene)m-N(R33)-SO2-NR33R34, -(alkylene)m-N(R33)-C(O)-OR35 ,
-(alkylene)m-N(R33)-C(S)-OR35, or -(alkylene)m-N(R33)-SO2-R35.
In one embodiment R32 is
Figure imgf000018_0001
These compounds can be used to treat conditions of abnormal cellular proliferation in a host in need thereof, typically a human.
In another embodiment, a method for the treatment of a fibrotic disorder in a host is provided that includes the administration of an effective amount of a compound of the present invention or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier. In another embodiment, a method for the treatment of rheumatoid arthritis or psoriasis in a host is provided that includes the administration of an effective amount of a compound of the present invention or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier.
In yet another embodiment, a method for the treatment of an autoimmune disorder in a host is provided that includes the administration of an effective amount of a compound of the present invention or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier.
In a principal embodiment, a method for the treatment of a tumor or cancer in a host is provided that includes the administration of an effective amount of a compound of the present invention or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier. In an aspect of this embodiment, the cancer is an Rb-positive tumor or cancer. In another aspect of this embodiment, the cancer is an Rb-negative tumor or cancer. In certain aspects, the cancer is selected from breast cancer, prostate cancer (including androgen- resistant prostate cancer), colon, including metastatic colon, another cancer of the reproductive system such as endometrial, ovarian or testicular cancer, small cell lung carcinoma, glioblastoma and head and/or neck cancer.
In yet another embodiment, a method for the treatment of a disorder of abnormal cellular proliferation in a host such as a human is provided that includes administering an effective amount of a combination of one or more of the active compounds described herein in combination or alternation with another active compound. In certain aspects of the invention, the second compound is a chemotherapeutic agent. In another aspect of this embodiment, the second active compound is an immune modulator, including but not limited to a checkpoint inhibitor such as an anti-PDl, Ant-PD-Ll, anti-CTLA, anti -LAG-3, anti-Tim, etc. antibody, small molecule, peptide, nucleotide or other inhibitor, including but not limited to ipilimumab (Yervoy), pembrolizumab (Keytruda) nivolumab (Opdivo), cemiplimab (Libtayo), atezolizumab (Tecentriq), avelumab (Bavencio), and durvalumab (Imfinzi).
In yet another embodiment, one of the active compounds described herein is administered in an effective amount for the treatment of abnormal tissue of the female reproductive system such as breast, ovarian, endometrial, or uterine cancer, in combination or alternation with an effective amount of an estrogen inhibitor including but not limited to a SERM (selective estrogen receptor modulator), a SERD (selective estrogen receptor degrader), a complete estrogen receptor degrader, or another form of partial or complete estrogen antagonist. In another embodiment, one of the active compounds described herein is administered in an effective amount for the treatment of abnormal tissue of the male reproductive system such as prostate or testicular cancer, in combination or alternation with an effective amount of an androgen (such as testosterone) inhibitor including but not limited to a selective androgen receptor modulator, a selective androgen receptor degrader, a complete androgen receptor degrader, or another form of partial or complete androgen antagonist. In one embodiment, the prostate or testicular cancer is androgen-resistant.
In one embodiment, the compounds described herein inhibit cyclin dependent kinase (“CDK”). For example, a compound described in the present invention provides a dose- dependent G1 -arresting effect on a subject’s CDK replication dependent healthy cells, for example HSPCs or renal epithelial cells. The methods provided for herein are sufficient to afford chemoprotection to targeted CDK replication dependent healthy cells during chemotherapeutic agent exposure, for example, during the time period that a DNA-damaging chemotherapeutic agent is capable of DNA-damaging effects on CDK replication dependent healthy cells in the subject.
In one embodiment, the administration of a compound using a method described herein is combined with the use of a hematopoietic growth factor including, but not limited to, granulocyte colony stimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), thrombopoietin, interleukin (IL)-12, steel factor, and erythropoietin (EPO), or their derivatives. In one embodiment, the compound is administered prior to administration of the hematopoietic growth factor. In one embodiment, the hematopoietic growth factor administration is timed so that the compound’s effect on HSPCs has dissipated.
In one embodiment, a compound described herein is administered in combination with a BTK inhibitor. In another embodiment, a compound described herein is administered in combination with an EGFR inhibitor.
The present invention also provides advantageous methods to treat a patient with a selective CDK4/6 inhibitor resistant cancer, which include administering an effective amount of a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, or a pharmaceutically acceptable composition, salt, isotopic analog, or prodrug thereof. In one aspect, a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, is used to treat a patient with a cancer intrinsically resistant to selective CDK4/6 inhibition. In one aspect, a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, is used to treat a patient with a cancer that has acquired resistance to one or more selective CDK4/6 inhibitors. In one aspect, a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, is administered in combination with a selective CDK4/6 inhibitor to a patient with a CDK4/6 inhibition responsive cancer in order to extend the therapeutic effectiveness of cell-cycle inhibition in the cancer. In some embodiments, the compound administered is selected from Compound 1, Compound 2, or Compound 3. In some embodiments, the compound administered is selected from Compound 74, Compound 76, or Compound 77.
Likewise, cancers initially susceptible to selective CDK4/6 inhibitor inhibition, such as ER+ breast cancer, may acquire resistance to selective CDK4/6 inhibition by upregulation of cyclin E which allows G1 to S cell cycle progression through CDK2. Thus, a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, can be used in an effective amount to treat patients with a cancer that has developed selective CDK4/6 inhibitor resistance over time, either due to prior exposure to a CDK 4/6 inhibitor or through a natural progression of the tumor. Accordingly, methods are provided herein that include administering an effective amount of a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, to treat a patient with a cancer initially responsive to selective CDK4/6 inhibition or susceptible to selective CDK4/6 inhibition that extend the efficacy of the selective CDK4/6 inhibitor treatment against a CDK4/6 responsive cancer by delaying acquired resistance to the inhibitory effects of the selective CDK4/6 inhibitor. In some embodiments, the compound administered is selected from Compound 1, Compound 2 and Compound 3. In some embodiments, the compound administered is selected from Compound 74, Compound 76 and Compound 77.
In a particular aspect, the present invention provides methods for treating a patient with cancer that has developed acquired resistance to a selective CDK4/6 inhibitor by administering to the patient an effective amount of a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV. In some embodiments, the selective CDK4/6 inhibitor to which the cancer has developed resistance is selected from palbociclib, abemaciclib, lerociclib, trilaciclib, SH6390, or ribociclib. In some embodiments, the compound administered is selected from Compound 1, Compound 2, or Compound 3. In some embodiments, the compound administered is selected from Compound 74, Compound 76, or Compound 77. In one aspect of the invention, provided herein is a method of treating a patient with cancer by administering a therapeutically effective amount of a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, in combination with a selective CDK 4/6 inhibitor, wherein the patient is selective CDK4/6 inhibitor treatment naive. By administering a compound of Formula I, II,
III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, in combination with a selective CDK 4/6 inhibitor, a delay in the onset of selective CDK4/6 inhibitor acquired resistance may be realized. In some embodiments, the selective CDK4/6 inhibitor administered in combination with a compound of Formula I, II, III,
IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, is selected from palbociclib, abemaciclib, ribociclib, trilaciclib, SHR6390, or lerociclib. In some embodiments, the compound administered is selected from Compound 1, Compound 2, or Compound 3. In some embodiments, the compound administered is selected from Compound 74, Compound 76, or Compound 77.
In one aspect of the invention, a method of treating a patient with cancer is provided that includes administering a therapeutically effective amount of a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, wherein the patient has previously received a selective CDK4/6 inhibitor, and the cancer has become selective CDK4/6 inhibitor resistant. By administering a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, following the development of selective CDK 4/6 inhibitor resistance, the current methods allow continued use of cell-cycle inhibition to treat the cancer. In some embodiments, the selective CDK4/6 inhibitor to which the cancer has developed resistance is selected from palbociclib, abemaciclib, ribociclib, trilaciclib, SHR6390, or lerociclib. In some embodiments, the compound administered is selected from Compound 1, Compound 2, or Compound 3. In some embodiments, the compound administered is selected from Compound 74, Compound 76, or Compound 77.
In one alternative aspect, provided herein is a method of treating a patient with an Rb- positive cancer which includes:
a) administering to the patient a selective CDK4/6 inhibitor;
b) monitoring the patient’s cyclin E levels in the cancer; and,
c) administering to the patient a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, upon the detection of an increase in cyclin E levels that confers resistance upon the cancer to the inhibitory effects of the selective CDK4/6 inhibitor. In some embodiments, the selective CDK4/6 inhibitor administered is selected from palbociclib, abemaciclib, ribociclib, trilaciclib, SHR6390, or lerociclib. In some embodiments, the compound administered is selected from Compound 1, Compound 2, or Compound 3. In some embodiments, the compound administered is selected from Compound 74, Compound 76, or Compound 77.
In one alternative aspect, provided herein is a method of treating a patient with cancer which includes:
a) determining the cancer’s Rb-status,
b) if the Rb-status is positive, administering to the patient a selective CDK4/6 inhibitor in combination with a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV;
c) if the Rb-status is negative, administering to the patient a compound of Formula I,
II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI,
XXII, XXIII, or XXIV, without a selective CDK 4/6 inhibitor.
In some embodiments, the selective CDK4/6 inhibitor administered in combination with a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, is selected from palbociclib, abemaciclib, ribociclib, trilaciclib, SHR6390, or lerociclib. In some embodiments, the compound administered is selected from Compound 1, Compound 2, or Compound 3. In some embodiments, the compound administered is selected from Compound 74, Compound 76, or Compound 77.
In one alternative aspect, provided herein is a method of treating a patient with an abnormal cellular proliferations such as cancer which includes:
a) administering to the patient a selective CDK4/6 inhibitor;
b) monitoring the patient’s cancer’s response to the selective CDK4/6 inhibitor;
c) administering to the patient a compound of Formula I, II, III, IV, V, VI, VII, VIII,
IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, upon the detection of the patient’s cancer becoming non-responsive to the selective CDK4/6 inhibitor. In some embodiments, the CDK4/6 inhibitor administered in combination with a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, is selected from palbociclib, abemaciclib, ribociclib, trilaciclib, SHR6390, or lerociclib. In some embodiments, the non-responsiveness is disease progression. In some embodiments, the compound administered is selected from Compound 1, Compound 2, or Compound 3. In some embodiments, the compound administered is selected from Compound 74, Compound 76, or Compound 77.
In one alternative aspect, provided herein is a method of treating a patient with an abnormal cellular proliferation, for example cancer, which includes:
a) administering to the patient a selective CDK4/6 inhibitor;
b) monitoring one or more cellular signals indicating the development of selective CDK4/6 inhibitor resistance in the cancer;
c) administering to the patient a compound of Formula I, II, III, IV, V, VI, VII, VIII,
IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, if one or more cellular signals indicate the development of selective CDK4/6 inhibitor resistance in the cancer. In some embodiments, the selective CDK4/6 inhibitor administered is selected from palbociclib, abemaciclib, ribociclib, trilaciclib, SHR6390, or lerociclib. In some embodiments, one or more cellular signals indicating the development of selective CDK4/6 inhibitor resistance in the cancer is selected from an increase in cyclin E expression, CCNEl/2 amplification, E2F amplification, CDK2 amplification, amplification of CDK6, amplification of CDK4, pl6 amplification, WEE1 overexpression, DM2 overexpression, CDK7 overexpression, loss of FZR1, HD AC activation, activation of the FGFR pathway, activation of the PI3K/AKT/mTOR pathway, loss of ER or PR expression, higher transcriptional activity of AP-1, epithelial-mesenchymal transition, Smad 3 suppression, autophagy activation, Rbl- loss, or inactivating RB 1 mutations. In some embodiments, the compound administered is selected from Compound 1, Compound 2, or Compound 3. In some embodiments, the compound administered is selected from Compound 74, Compound 76, or Compound 77.
In one alternative aspect of the present invention, provided herein is a pharmaceutically acceptable composition comprising a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX,
X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, and a selective CDK4/6 inhibitor, for example but not limited to one selected from palbociclib, abemaciclib, ribociclib, trilaciclib, SHR6390, or lerociclib. In some embodiments, the compound administered is selected from Compound 1, Compound 2, or Compound 3. In some embodiments, the compound administered is selected from Compound 74, Compound 76, or Compound 77.
In yet another embodiment, a method for the treatment of a disorder of abnormal cellular proliferation in a host such as a human is provided that includes administering an effective amount of a combination a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, and a selective CDK4/6 inhibitor in combination or alternation with an additional active compound. In certain aspects of the invention, the additional active compound is a chemotherapeutic agent. In another aspect of this embodiment, the additional active compound is an immune modulator, including but not limited to a checkpoint inhibitor such as an anti-PDl, anti-PD-Ll, anti- CTLA, anti-LAG-3, anti-Tim, etc. antibody, small molecule, peptide, nucleotide or other inhibitor including but not limited to ipilimumab (Yervoy), pembrolizumab (Keytruda) nivolumab (Opdivo), cemiplimab (Libtayo), atezolizumab (Tecentriq), avelumab (Bavencio), and durvalumab (Imfinzi). In some embodiments, the compound administered is selected from Compound 1, Compound 2, or Compound 3. In some embodiments, the compound administered is selected from Compound 74, Compound 76, or Compound 77.
In yet another embodiment, a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX,
X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, in combination with a selective CDK4/6 inhibitor, is administered in an effective amount for the treatment of abnormal tissue of the female reproductive system such as breast, ovarian, endometrial, or uterine cancer, in combination or alternation with an effective amount of an estrogen inhibitor including but not limited to a SERM (selective estrogen receptor modulator), a SERD (selective estrogen receptor degrader), a complete estrogen receptor degrader, or another form of partial or complete estrogen antagonist. In some embodiments, the compound administered is selected from Compound 1, Compound 2, or Compound 3. In some embodiments, the compound administered is selected from Compound 74, Compound 76, or Compound 77.
In another embodiment, a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X,
XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, in combination with a selective CDK4/6 inhibitor, is administered in an effective amount for the treatment of abnormal tissue of the male reproductive system such as prostate or testicular cancer, in combination or alternation with an effective amount of an androgen (such as testosterone) inhibitor including but not limited to a selective androgen receptor modulator, a selective androgen receptor degrader, a complete androgen receptor degrader, or another form of partial or complete androgen antagonist. In some embodiments, the prostate or testicular cancer is androgen-resistant. In some embodiments, the compound administered is selected from Compound 1, Compound 2, or Compound 3. In some embodiments, the compound administered is selected from Compound 74, Compound 76, or Compound 77.
In some embodiments, a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, in combination with a CDK4/6 inhibitor, is administered in an effective amount in combination with a BTK inhibitor. In another embodiment, a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, in combination with a CDK4/6 inhibitor, is administered in an effective amount in combination with an EGFR inhibitor. In some embodiments, the compound administered is selected from Compound 1, Compound 2, or Compound 3. In some embodiments, the compound administered is selected from Compound 74, Compound 76, or Compound 77.
In certain embodiments the compound of the present invention inhibits CDK2, CDK4, CDK6, and/or CDK9. In certain embodiments the compound is a CDK2 inhibitor. In certain embodiments the compound is a CDK4 inhibitor. In certain embodiments the compound is a CDK6 inhibitor. In certain embodiments the compound is a CDK9 inhibitor.
The present invention thus includes at least the following features:
(a) a compound of the present invention as described herein, or a pharmaceutically acceptable salt or prodrug thereof;
(b) a compound of the present invention as described herein, or a pharmaceutically acceptable salt or prodrug thereof that is useful in an effective amount to treat a disorder of abnormal cellular proliferation, including a tumor or cancer;
(c) a compound of the present invention as described herein, or a pharmaceutically acceptable salt or prodrug thereof that is useful in the treatment of cancer that is resistant to treatment with a compound that is a CDK4/6 inhibitor for example treatment with palbociclib, abemaciclib, or ribociclib;
(d) use of a compound of the present invention, or a pharmaceutically acceptable salt or prodrug thereof in the manufacture of a medicament for the treatment of a disorder of abnormal cellular proliferation, such as a tumor or cancer;
(e) use of a compound of the present invention, or a pharmaceutically acceptable salt or prodrug thereof in the manufacture of a medicament for the treatment of a cancer that is resistant to treatment with a compound that is a CDK4/6 inhibitor for example treatment with palbociclib, abemaciclib, or ribociclib;
(f) a method for manufacturing a medicament intended for the therapeutic use of treating a disorder of abnormal cellular proliferation including a tumor or cancer, characterized in that a compound of the present invention as described herein is used in the manufacture;
(q) a pharmaceutical formulation comprising an effective host-treating amount of the compound of the present invention or a pharmaceutically acceptable salt or prodrug thereof together with a pharmaceutically acceptable carrier or diluent; (r) a compound of the present invention as described herein as a mixture of enantiomers or diastereomers (as relevant), including as a racemate;
(s) a compound of the present invention as described herein in enantiomerically or diastereomerically (as relevant) enriched form, including as an isolated enantiomer or disastereomer (i.e., greater than 85, 90, 95, 97 or 99% pure);
(t) a process for the preparation of therapeutic products that contain an effective amount of a compound of the present invention, as described herein.
(u) a solid dosage form of a compound of the present invention or its pharmaceutically acceptable salt in a pharmaceutically acceptable carrier for oral delivery;
(v); a parenteral dosage form of a compound of the present invention or its pharmaceutically acceptable salt in a pharmaceutically acceptable carrier for systemic delivery, including via intravenous delivery; and
(w) a method for manufacturing a medicament intended for anti-neoplastic therapy, characterized in that a compound of the present invention as described herein is used in the manufacture.
BRIEF DESCRIPTION OF THE FIGURES
FIG. 1 is a concentration curve to measure compound binding using the NanoBRET Target Engagement Intracellular Kinase Assay as described in Example 4. The binding of Compounds 1, 2, and 3 to CDK2/Cyclin E was measured and all three compounds exhibited potent binding. PF-0687300 was used as a reference compound. The x-axis is the concentration of the test compounds measured in molarity expressed as log[M] and the y-axis is the ratio (BRET ratio) of the acceptor emission value to the donor emission measured in milliBRET units (mBU).
FIG. 2A is a graphical depiction of all kinases inhibited greater than 95% by Compound
1 at a concentration of 100 nM (Compound 1 was assessed at 200x the biochemical IC50 of CDK2/CyclinE) as discussed in Example 5. CDK2 is represented by the labeled circle.
FIG. 2B is a graphical depiction of all kinases inhibited greater than 95% by Compound
2 at a concentration of 100 nM (Compound 2 was assessed at 1000x the biochemical IC50 of CDK2/CyclinE) as discussed in Example 5. CDK2 is represented by the labeled circle.
FIG. 2C is a graphical depiction of all kinases inhibited greater than 95% by Compound
3 at a concentration of 100 nM (Compound 3 was assessed at 250x the biochemical IC50 of CDK2/CyclinE) as discussed in Example 5. CDK2 is represented by the labeled circle. FIG. 3 A is an image of MCF7 parental cells (left) and MCF7 palbociclib-resistant cells (right) developed for four months as described in Example 6. The MCF7 palbociclib-resistant cells were maintained in complete media plus palbociclib for three months at ~IC90 (750nM) followed by one month at ImM.
FIG. 3B is a dose-response curve of palbociclib activity in the MCF7 cell line as described in Example 6. The IC50 of palbociclib in the MCF7 cell line was 33 nM. The x-axis is the concentration of palbociclib measured in molarity (M) expressed as the log[M] The y- axis is the luminescence measured in RLU.
FIG. 3C is a dose-response curve of palbociclib activity in the MCF7 Palbo-R cell line as described in Example 6. The IC50 of palbociclib in the MCF7 Palbo-R cell line was 2890 nM. The x-axis is the concentration of palbociclib measured in molarity (M) expressed as the log[M] The y-axis is the luminescence measured in RLU.
FIG. 4A is the pairwise comparison of transcript level in MCF7 palbociclib-resistant cells vs. MCF7 cells (control) as described in Example 6. Genes above the dashed line were differentially expressed (6,039 genes out of 17,383) with an adjusted p-value of 0.05. The x- axis is fold change expressed as log2. The y-axis is -logio(adjusted p value). The horizontal dashed line is -logio(0.05) and all points above the dashed line are statistically significant.
FIG. 4B is a graph of the fold change (expressed as log2) of specific genes in MCF7 palbociclib-resistant cells vs. MCF7 cells as described in Example 6. Three genes, CCNE1, CCNE2, and CDK6, were significantly upregulated in the palbociclib-resistant cells compared to the MCF7 cells. * denotes statistical significance (adjusted p value <0.05). The x-axis is fold change expressed as log 2 and the y-axis is labeled with the specific genes.
FIG. 4C are Western blots of MCF7 and MCF7 palbociclib-resistant cells that show the expression of Cyclin E and Rb (GAPDH is the control). The expression levels are also plotted on the graph below. As described in Example 6, there is an increase in the ratio of Cyclin E to Rb levels in the palbociclib-resistant cells compared to the MCF7 cells.
FIG. 5 A is a graph of the percentage of HCC1806 cells in the G0-G1 phase, the S phase, the G2 phase, and the M phase after increasing concentrations of Compound 1, Compound 2, Compound 3, and PF-06873600 (100 nM, 300 nM, and 1000 nM) as described in Example 6. The x-axis is the concentration of Compound 1, Compound 2, Compound 3, and PF-06873600 measured in nM and the y-axis is the percentage of the total cell population in each cell cycle phase.
FIG. 5B is a graph of the percentage of MCF7 cells in the G0-G1 phase, the S phase, the G2 phase, and the M phase after increasing concentrations of Compound 1, Compound 2, Compound 3, and PF-06873600 (100 nM, 300 nM, and 1000 nM) as described in Example 6. The x-axis is the concentration of Compound 1, Compound 2, Compound 3, and PF-06873600 measured in nM and the y-axis is the percentage of the total cell population in each cell cycle phase.
FIG. 5C is a graph of the percentage of MCF7 Palbo-R cells in the G0-G1 phase, the S phase, the G2 phase, and the M phase after increasing concentrations of Compound 1, Compound 2, Compound 3, and PF-06873600 (100 nM, 300 nM, and 1000 nM) as described in Example 6. The x-axis is the concentration of Compound 1, Compound 2, Compound 3, and PF-06873600 measured in nM and the y-axis is the percentage of the total cell population in each cell cycle phase.
FIG. 5D is a graph of the percentage of Hs68 cells in the G0-G1 phase, the S phase, the G2 phase, and the M phase after increasing concentrations of Compound 1, Compound 2, Compound 3, and PF-06873600 (100 nM, 300 nM, and 1000 nM) as described in Example 6. The x-axis is the concentration of Compound 1, Compound 2, Compound 3, and PF-06873600 measured in nM and the y-axis is the percentage of the total cell population in each cell cycle phase.
FIG. 5E is a representative flow gating schematic using the FxCycle DNA stain, Click- iTTM Edu assay to measure cellular proliferation of HCC1806 cells treated with vehicle as described in Example 6. The y-axis is the Alexa Fluor 488-A Edu fluorescence. The x-axis is the allophycocyanin (APC) fluorescence.
FIG. 5F is a representative flow gating schematic using the FxCycle DNA stain, Click- iTTM Edu assay to measure cellular proliferation of HCC1806 cells treated with 1000 nM of Compound 3 as described in Example 6. The y-axis is the Alexa Fluor 488-A Edu fluorescence. The x-axis is the allophycocyanin (APC) fluorescence.
FIG. 5G is a representative flow gating schematic using Phospho-Histone H3 conjugated antibody assay to measure cellular proliferation of HCC1806 cells treated with vehicle as described in Example 6. The y-axis is the Pacific Blue A Phospho-Histone H3 fluorescence. The x-axis is the allophycocyanin (APC) fluorescence.
FIG. 5H is a representative flow gating schematic using Phospho-Histone H3 conjugated antibody assay to measure cellular proliferation of HCC1806 cells treated with 1000 nM of Compound 3 as described in Example 6. The y-axis is the Pacific Blue A Phospho- Histone H3 fluorescence. The x-axis is the allophycocyanin (APC) fluorescence.
FIG. 6A are Western blots of HCC1806 cells treated with Compound 1, Compound 2, and Compound 3 that show the expression of p-Rb and p-21 (a-tubulin is the control) as described in Example 7. Compound 1, Compound 2, Compound 3, and PF-06873600 were administered at dosages of 100, 300, and 1000 nM. PF-06873600 was used as a reference compound. The expression levels are also plotted on the graph below.
FIG. 6B are Western blots of MCF7 Palbo-R cells treated with Compound 1, Compound 2, and Compound 3 that show the expression of p-Rb and p-21 (a-tubulin is the control) as described in Example 7. Compound 1, Compound 2, Compound 3, and PF- 06873600 were administered at dosages of 100, 300, and 1000 nM. PF-06873600 was used as a reference compound. The expression levels are also plotted on the graph below.
FIG. 6C are Western blots of Hs68 cells treated with Compound 1, Compound 2, and Compound 3 that show the expression of p-Rb and p-21 (a-tubulin is the control) as described in Example 7. Compound 1, Compound 2, Compound 3, and PF-06873600 were administered at dosages of 100, 300, and 1000 nM. PF-06873600 was used as a reference compound. The expression levels are also plotted on the graph below.
FIG. 7A is a dose-response curve of Compound 1, Compound 2, and Compound 3 activity in triple-negative breast cancer cell line HCC1806 as described in Example 8. PF- 06873600 was used as a reference compound. The IC50 value of Compound 1, Compound 2, Compound 3, and PF-06873600 against the HCC1806 cell line is given in Table 6. The x-axis is concentration of the compounds measured in molarity (M) expressed as the log[M] The y- axis is the luminescence measured in RLU.
FIG. 7B is a dose-response curve of Compound 1, Compound 2, and Compound 3 activity in triple-negative breast cancer cell line BT549 as described in Example 8. PF- 06873600 was used as a reference compound. The IC50 value of Compound 1, Compound 2, Compound 3, and PF-06873600 against the BT549 cell line is given in Table 6. The x-axis is concentration of the compounds measured in molarity (M) expressed as the log[M] The y- axis is the luminescence measured m RLU.
FIG. 7C is a dose-response curve of Compound 1, Compound 2, and Compound 3 activity in the MCF7 cell line as described in Example 8. PF-06873600 was used as a reference compound. The IC50 value of Compound 1, Compound 2, Compound 3, and PF-06873600 against the MCF7 cell line is given in Table 6. The x-axis is concentration of the compounds measured in molarity (M) expressed as the log[M] The y-axis is the luminescence measured in RLU.
FIG. 7D is a dose-response curve of Compound 1, Compound 2, and Compound 3 activity in the MCF7 Palbo-R cell line as described in Example 8. PF-06873600 was used as a reference compound. The IC50 value of Compound 1, Compound 2, Compound 3, and PF- 06873600 against the MCF7 Palbo-R cell line is given in Table 6. The x-axis is concentration of the compounds measured in molarity (M) expressed as the log[M] The y-axis is the luminescence measured in RLU.
FIG. 7E is a dose-response curve of Compound 1, Compound 2, and Compound 3 activity in the Hs68 cell line as described in Example 8. PF-06873600 was used as a reference compound. The IC50 value of Compound 1, Compound 2, Compound 3, and PF-06873600 against the Hs68 cell line is given in Table 6. The x-axis is concentration of the compounds measured in molarity (M) expressed as the log[M]. The y-axis is the luminescence measured in RLU.
FIG. 8A is a measure of the caspase 3/7 activity in HCC1806 cells after 24 hours of incubation with increasing concentrations of Compound 1, Compound 3, PF-06873600, and staurosporine as described in Example 9. The x-axis is the concentration of the compounds measured in molarity (M) expressed as the log[M]. The y-axis is the luminescence measured in RLU.
FIG. 8B is a measure of the caspase 3/7 activity in HS68 cells after 24 hours of incubation with increasing concentrations of Compound 1, Compound 3, PF-06873600, and staurosporine as described in Example 9. The x-axis is the concentration of the compounds measured in molarity (M) expressed as the log[M]. The y-axis is the luminescence measured in RLU.
FIG. 9 is a graph showing the in vivo evaluation of Compound 2 in a PDX model of human breast cancer in immunocompromised mice as described in Example 10. The mice were dosed for fourteen days with either a control vehicle, 100 mg/kg of Compound 2 once daily (QD), of 75 mg/kg twice daily (BID). The y-axis represents the change in tumor size at the end of the 14-day dosing period.
FIG. 10 is a graph of the percentage of Hs68 cells in the G0-G1 phase, the S phase, the G2 phase, and the M phase after increasing concentrations of Compound 74 (10 nM, 30 nM, 100 nM) as described in Example 11. The x-axis is the concentration of Compound measured in nM and the y-axis is the percentage of the total cell population in each cell cycle phase.
FIG. 11 is a graph of the percentage of Hs68 cells in the G0-G1 phase, the S phase, the G2 phase, and the M phase after increasing concentrations of Compound 75. The cell cycle arresting affect is compared to a vehicle control (VC), 300 nM dosing of Compound 2, the CDK4/6 inhibitor G1T38, as described in Example 11. The x-axis is the concentration of compound measured in nM and the y-axis is the percentage of the total cell population in each cell cycle phase. DETAILED DESCRIPTION
I. COMPOUNDS
In one embodiment, the compound of the present invention is of formula:
Figure imgf000032_0001
or a pharmaceutically acceptable salt, N-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In one embodiment, the compound of the present invention is of formula:
Figure imgf000032_0002
or a pharmaceutically acceptable salt, N-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In some embodiments, the compound of the present invention is of formula:
Figure imgf000033_0001
or a pharmaceutically acceptable salt. N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined herein.
In some embodiments, the compound of the present invention is of formula:
Figure imgf000033_0002
Figure imgf000034_0001
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In some embodiments, the compound of the present invention is of formula:
Figure imgf000034_0002
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables as defined above in the Summary or otherwise herein.
In one embodiment, the compound of the present invention is of formula:
Figure imgf000034_0003
or a pharmaceutically acceptable salt, N-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined herein.
In one embodiment, the compound of the present invention is of formula:
Figure imgf000035_0001
or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In one embodiment, the compound of the present invention is of formula:
Figure imgf000035_0002
Figure imgf000036_0001
or a pharmaceutically acceptable salt. N-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables as defined above in the Summary or otherwise herein.
In one embodiment, the compound of the present invention is of formula:
Figure imgf000036_0002
Figure imgf000037_0001
or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In one embodiment, the compound of the present invention is of formula:
Figure imgf000037_0002
Figure imgf000038_0001
or a pharmaceutically acceptable salt. N-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In one embodiment, the compound of the present invention is of formula:
Figure imgf000039_0001
or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In one embodiment, the compound of the present invention is of formula:
Figure imgf000040_0001
Figure imgf000041_0001
or a pharmaceutically acceptable salt N-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In one embodiment, the compound of the present invention is of formula:
Figure imgf000041_0002
Figure imgf000042_0001
or a pharmaceutically acceptable salt N-oxide isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In one embodiment, the compound of the present invention is of formula:
Figure imgf000042_0002
Figure imgf000043_0001
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In one embodiment, the compound of the present invention is of formula:
Figure imgf000043_0002
or a pharmaceutically acceptable salt, N-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In one embodiment, the compound of the present invention is of formula:
Figure imgf000044_0001
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In one embodiment, the compound of the present invention is of formula:
Figure imgf000045_0001
or a pharmaceutically acceptable salt, N-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In one embodiment, the compound of the present invention is selected from:
Figure imgf000045_0002
Figure imgf000046_0001
or a pharmaceutically acceptable salt N-oxide isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In one embodiment, the compound of the present invention is selected from:
Figure imgf000046_0002
or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In one embodiment, the compound of the present invention is selected from:
Figure imgf000047_0001
or a pharmaceutically acceptable salt, N-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In one embodiment, the compound of the present invention is selected from:
Figure imgf000047_0002
Figure imgf000048_0001
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In one embodiment, the compound of the present invention is selected from:
Figure imgf000048_0002
or a pharmaceutically acceptable salt, N-oxide , isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In one embodiment, the compound of the present invention is selected from:
Figure imgf000049_0001
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In one embodiment, the compound of the present invention is selected from:
Figure imgf000050_0001
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In one embodiment, the compound of the present invention is of formula:
Figure imgf000051_0001
Figure imgf000052_0001
or a pharmaceutically acceptable salt. /V-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In one embodiment, the compound of the present invention is of formula:
Figure imgf000052_0002
Figure imgf000053_0001
or a pharmaceutically acceptable salt. /V-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In certain embodiments, the compound of the present invention is selected from:
Figure imgf000053_0002
Figure imgf000054_0001
Figure imgf000055_0001
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In certain embodiments, the compound of the present invention is selected from:
Figure imgf000055_0002
Figure imgf000056_0001
Figure imgf000057_0002
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In certain embodiments, the compound of the present invention is selected from:
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In certain embodiments, the compound of the present invention is selected from:
Figure imgf000059_0002
Figure imgf000060_0001
or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In certain embodiments, the compound of the present invention is selected from:
Figure imgf000060_0002
Figure imgf000061_0001
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In certain embodiments, the compound of the present invention is selected from:
Figure imgf000062_0001
Figure imgf000063_0001
or a pharmaceutically acceptable salt N-oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In certain embodiments, the compound of the present invention is selected from:
Figure imgf000063_0002
Figure imgf000064_0001
or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In certain embodiments, the compound of the present invention is selected from:
Figure imgf000064_0002
Figure imgf000065_0001
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In certain embodiments, the compound of the present invention is selected from:
Figure imgf000066_0001
Figure imgf000067_0002
or a pharmaceutically acceptable salt. N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In one embodiment, the compound of the present invention is of formula:
Figure imgf000067_0001
or a pharmaceutically acceptable salt, N-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In one embodiment, the compound of the present invention is of formula:
Figure imgf000068_0001
or a pharmaceutically acceptable salt, N-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In one embodiment, the compound of the present invention is of formula:
Figure imgf000069_0001
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In one embodiment, the compound of the present invention is of formula:
Figure imgf000070_0001
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In one embodiment, the compound of the present invention is of formula:
Figure imgf000071_0001
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In certain embodiments, the compound of the present invention is selected from:
Figure imgf000072_0001
or a pharmaceutically acceptable salt, N-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In certain embodiments, the compound of the present invention is selected from:
Figure imgf000073_0001
or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In certain embodiments, the compound of the present invention is selected from:
Figure imgf000074_0001
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In certain embodiments, the compound of the present invention is selected from:
Figure imgf000075_0001
or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In certain embodiments, the compound of the present invention is selected from:
Figure imgf000076_0001
or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In certain embodiments the compound of the present invention is of formula:
Figure imgf000077_0001
Figure imgf000078_0002
or a pharmaceutically acceptable salt. N-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In certain embodiments the compound of the present invention is of formula:
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0002
or a pharmaceutically acceptable salt, N-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In certain embodiments the compound of the present invention is of formula:
Figure imgf000080_0001
Figure imgf000081_0001
or a pharmaceutically acceptable salt. V-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In certain embodiments the compound of the present invention is of formula:
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0002
or a pharmaceutically acceptable salt. N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In certain embodiments, the compound of the present invention is of formula:
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
or a pharmaceutically acceptable salt, N-oxide. isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In certain embodiments the compound of the present invention is selected from:
Figure imgf000087_0001
or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein. In certain embodiments the compound of the present invention is selected from:
Figure imgf000088_0001
or a pharmaceutically acceptable salt. N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In certain embodiments the compound of the present invention is selected from:
Figure imgf000088_0002
Figure imgf000089_0002
or a pharmaceutically acceptable salt. N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In certain embodiments the compound of the present invention is selected from:
Figure imgf000089_0001
Figure imgf000090_0001
or a pharmaceutically acceptable salt, N- oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof; wherein the variables are as defined above in the Summary or otherwise herein.
In another embodiment the compound is selected from:
Figure imgf000090_0002
Figure imgf000091_0001
Figure imgf000092_0001
or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof;
wherein:
R is hydrogen, C1-C6alkyl, -(C0-C2alkyl)(C3-C8carbocyclyl), -(C0-C2alkyl)(C3-C8heterocyclyl),-(C0-C2alkyl)(aryl), -(C0-C2alkyl)(heteroaryl), -COOalkyl, -COOarylalkyl, or -COOH.
In one aspect, a compound is provided selected from:
Figure imgf000092_0002
Figure imgf000093_0002
or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, prodrug, and/or a pharmaceutically acceptable composition thereof.
In another aspect, a compound is provided selected from:
Figure imgf000093_0001
Figure imgf000094_0002
or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, or prodrug, optionally in a pharmaceutically acceptable carrier to form a pharmaceutically acceptable composition.
In another aspect, a compound is provided selected from:
Figure imgf000094_0001
Figure imgf000095_0001
or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, or prodrug, optionally in a pharmaceutically acceptable carrier to form a pharmaceutically acceptable composition.
In one aspect, a compound is provided selected from:
Figure imgf000095_0002
Figure imgf000096_0001
or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, or prodrug, optionally in a pharmaceutically acceptable carrier to form a pharmaceutically acceptable composition.
In one aspect, a compound is provided selected from
Figure imgf000096_0002
Figure imgf000096_0003
or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, or prodrug, optionally in a pharmaceutically acceptable carrier to form a pharmaceutically acceptable composition.
Embodiments of“alkyl”
In one embodiment “alkyl” is a C1-C 10alkyl, C1-C9alkyl, C1-C8alkyl, C1-C7alkyl, C1-Cealkyl, C1-Csalkyl, C1-Cralkyl, C1-C3alkyl, or C1-C2alkyl.
In one embodiment“alkyl” has one carbon.
In one embodiment“alkyl” has two carbons.
In one embodiment“alkyl” has three carbons.
In one embodiment“alkyl” has four carbons.
In one embodiment“alkyl” has five carbons.
In one embodiment“alkyl” has six carbons.
Non-limiting examples of “alkyl” include: methyl, ethyl, propyl, butyl, pentyl, and hexyl.
Additional non-limiting examples of“alkyl” include: isopropyl, isobutyl, isopentyl, and isohexyl.
Additional non-limiting examples of “alkyl” include: vec-butyl, vec-pentyl. and sechexyl.
Additional non-limiting examples of “alkyl” include: tert- butyl, tert- pentyl, and tert-hexyl.
Additional non-limiting examples of“alkyl” include: neopentyl, 3-pentyl, and active pentyl.
In one embodiment“alkyl” is“substituted alkyl”
In one embodiment“alkenyl” is“substituted alkenyl”
In one embodiment“alkynyl” is“substituted alkynyl”
Embodiments of“haloalkyl”
In one embodiment“haloalkyl” is a C1-C 10haloalkyl, C1-C9haloalkyl, C1-C8haloalkyl, C1-C7haloalkyl, C1-C6haloalkyl, C1-C5haloalkyl, C1-C4haloalkyl, C1-C3haloalkyl, and C1- C2haloalkyl.
In one embodiment“haloalkyl” has one carbon.
In one embodiment“haloalkyl” has one carbon and one halogen.
In one embodiment“haloalkyl” has one carbon and two halogens.
In one embodiment“haloalkyl” has one carbon and three halogens. In one embodiment“haloalkyl” has two carbons.
In one embodiment“haloalkyl” has three carbons.
In one embodiment“haloalkyl” has four carbons.
In one embodiment“haloalkyl” has five carbons.
In one embodiment“haloalkyl” has six carbons.
Non-limiting examples of“haloalkyl” include:
Figure imgf000098_0002
Additional non-limiting examples of“haloalkyl” include:
Figure imgf000098_0003
Figure imgf000098_0001
Additional non-limiting examples of“haloalkyl” include:
Figure imgf000098_0004
Additional non-limiting examples of“haloalkyl” include:
Figure imgf000098_0005
Embodiments of“aryl”
In one embodiment“aryl” is a 6 carbon aromatic group (phenyl)
In one embodiment“aryl” is a 10 carbon aromatic group (napthyl)
In one embodiment“aryl” is a 6 carbon aromatic group fused to a heterocycle wherein the point of attachment is the aryl ring. Non-limiting examples of “aryl” include indoline, tetrahydroquinoline, tetrahydroisoquinoline. and dihydrobenzofuran wherein the point of attachment for each group is on the aromatic ring.
Figure imgf000098_0006
In one embodiment“aryl” is a 6 carbon aromatic group fused to a cycloalkyl wherein the point of attachment is the aryl ring. Non-limiting examples of“aryl” include dihydro-indene and tetrahydronaphthalene wherein the point of attachment for each group is on the aromatic ring. For example,
Figure imgf000099_0002
is an“aryl” group.
However, is a“cycloalkyl” group.
Figure imgf000099_0003
In one embodiment“aryl” is“substituted aryl”. Embodiments of“heteroaryl”
In one embodiment“heteroaryl” is a 5 membered aromatic group containing 1, 2, 3, or 4 nitrogen atoms.
Non-limiting examples of 5 membered“heteroaryl” groups include pyrrole, furan, thiophene, pyrazole, imidazole, triazole, tetrazole, isoxazole, oxazole, oxadiazole, oxatriazole, isothiazole, thiazole, thiadiazole, and thiatriazole.
Additional non-limiting examples of 5 membered“heteroaryl” groups include:
Figure imgf000099_0001
In one embodiment“heteroaryl” is a 6 membered aromatic group containing 1, 2, or 3 nitrogen atoms (i.e. pyridmyl, pyridazinyl, tnazinyl, pyrimidinyl, and pyrazinyl).
Non-limiting examples of 6 membered“heteroaryl” groups with 1 or 2 nitrogen atoms include:
Figure imgf000099_0004
In one embodiment“heteroaryl” is a 9 membered bicyclic aromatic group containing 1 or 2 atoms selected from nitrogen, oxygen, and sulfur. Non-limiting examples of heteroaryl groups that are bicyclic include indole, benzofuran, isoindole, indazole, benzimidazole, azaindole, azamdazole, purine, isobenzofuran, benzothiophene, benzoisoxazole, benzoisothiazole, benzooxazole, and benzothiazole.
Additional non-limiting examples of“heteroaryl” groups that are bicyclic include:
Additional non-limiting examples of“heteroaryl” groups that are bicyclic include:
Figure imgf000100_0001
Additional non-limiting examples of“heteroaryl” groups that are bicyclic include:
Figure imgf000100_0002
In one embodiment“heteroaryl” is a 10 membered bicyclic aromatic group containing 1 or 2 atoms selected from nitrogen, oxygen, and sulfur.
Non-limiting examples of “heteroaryl” groups that are bicyclic include quinoline, isoquinoline, quinoxaline, phthalazine, quinazoline, cinnoline, and naphthyridine.
Additional non-limiting examples of“heteroaryl” groups that are bicyclic include:
Figure imgf000100_0003
In one embodiment“heteroaryl” is“substituted heteroaryl”
Embodiments of“cycloalkyl”
In one embodiment “cycloalkyl” is a C3-C8cycloalkyl, C3-C7cycloalkyl, C3- C6cycloalkyl, C3-C5cycloalkyl, C3-C4cycloalkyl, C4-C8cycloalkyl, C5-C8cycloalkyl, or C6- C8cycloalkyl.
In one embodiment“cycloalkyl” has three carbons.
In one embodiment“cycloalkyl” has four carbons.
In one embodiment“cycloalkyl” has five carbons.
In one embodiment“cycloalkyl” has six carbons. In one embodiment“cycloalkyl” has seven carbons.
In one embodiment“cycloalkyl” has eight carbons.
In one embodiment“cycloalkyl” has nine carbons.
In one embodiment“cycloalkyl” has ten carbons.
Non-limiting examples of“cycloalkyl” include: cy clopropyl, cyclobutyl, cy clopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and cyclodecyl.
Additional non-limiting examples of “cycloalkyl” include dihydro-indene and tetrahydronaphthalene wherein the point of attachment for each group is on the cycloalkyl ring.
For example,
Figure imgf000101_0001
is an cycloalkyl group.
However,
Figure imgf000101_0002
In one embodiment“cycloalkyl” is a“substituted cycloalkyl”
Embodiments of“heterocycle”
In one embodiment“heterocycle” refers to a cyclic ring with one nitrogen and 3, 4, 5, 6, 7, or 8 carbon atoms.
In one embodiment“heterocycle” refers to a cyclic ring with one nitrogen and one oxygen and 3, 4, 5, 6, 7, or 8 carbon atoms.
In one embodiment“heterocycle” refers to a cyclic ring with two nitrogens and 3, 4, 5,
6, 7, or 8 carbon atoms.
In one embodiment“heterocycle” refers to a cyclic ring with one oxygen and 3, 4, 5, 6,
7, or 8 carbon atoms.
In one embodiment“heterocycle” refers to a cyclic ring with one sulfur and 3, 4, 5, 6, 7, or 8 carbon atoms.
Non-limiting examples of“heterocycle” include aziridine, oxirane, thiirane, azetidine, 1.3-diazetidine. oxetane. and thietane.
Additional non-limiting examples of“heterocycle” include pyrrolidine, 3-pyrroline, 2- pyrroline, pyrazolidine, and imidazolidine.
Additional non-limiting examples of “heterocycle” include tetrahydrofuran, 1,3- dioxolane, tetrahydrothiophene, 1,2-oxathiolane, and 1,3-oxathiolane. Additional non-limiting examples of “heterocycle” include piperidine, piperazine, tetrahydropyran, 1,4-dioxane, thiane, 1,3-dithiane, 1,4-dithiane, morpholine, and thiomorpholine.
Additional non-limiting examples of “heterocycle” include indoline, tetrahydroquinoline, tetrahydroisoquinoline, and dihydrobenzofuran wherein the point of attachment for each group is on the heterocyclic ring.
For example,
Figure imgf000102_0001
is a“heterocycle” group.
However, is an“aryl” group.
Figure imgf000102_0002
Non-limiting examples of“heterocycle” also include:
Figure imgf000102_0003
Additional non-limiting examples of“heterocycle” include:
Figure imgf000102_0004
Additional non-limiting examples of“heterocycle” include:
Figure imgf000102_0005
Non-limiting examples of“heterocycle” also include:
Figure imgf000102_0006
Non-limiting examples of“heterocycle” also include:
Figure imgf000102_0007
Additional non-limiting examples of“heterocycle” include:
Figure imgf000103_0001
Additional non-limiting examples of“heterocycle” include:
Figure imgf000103_0002
In one embodiment“heterocycle” is“substituted heterocycle”.
Embodiments of“-alkyl-aryl”
In one embodiment the“-alkyl-aryl” refers to a 1 carbon alkyl group substituted with an aryl group.
Non-limiting examples of“-alkyl-aryl” include:
Figure imgf000103_0003
In one embodiment“-alkyl-aryl” is
Figure imgf000103_0004
In one embodiment the“-alkyl-aryl” refers to a 2 carbon alkyl group substituted with an aryl group.
Non-limiting examples of“-alkyl-aryl” include:
Figure imgf000103_0005
In one embodiment the“alkyl-aryl” refers to a 3 carbon alkyl group substituted with an aryl group. Optional Substituents
In one embodiment a group described herein that can be substituted with 1 or 2 substituents is substituted with one substituent.
In one embodiment a group described herein that can be substituted with 1 or 2 substituents is substituted with two substituents. In one embodiment a group described herein that can be substituted with 1, 2, 3, or 4 substituents is substituted with one substituent.
In one embodiment a group described herein that can be substituted with 1. 2, 3, or 4 substituents is substituted with two substituents.
In one embodiment a group described herein that can be substituted with 1. 2. 3. or 4 substituents is substituted with three substituents.
In one embodiment a group described herein that can be substituted with 1, 2, 3, or 4 substituents is substituted with four substituents.
Embodiments of R1
In one embodiment y is 1 and R1 is aryl.
In one embodiment y is 1 and R1 is phenyl.
In one embodiment y is 1 and R1 is alkyl.
In one embodiment at least one R1 is hydroxyl.
In one embodiment at least one R1 is halogen.
In one embodiment at least one R1 is haloalkyl.
In one embodiment at least one R1 is flourine.
In one embodiment at least two R1s are flourine.
In one embodiment y is 3 and the two R1s are on the same ring atom and combine to form a 6-membered cycloalkyl.
In one embodiment y is 3 and the two R1s are on the same ring atom and combine to form a 6-membered cycloalkyl and the third R1 is hydroxyl.
In one embodiment y is 3 and the two R1s are on the same ring atom and combine to form a 6-membered cycloalkyl and the third R1 is haloalkyl.
In one embodiment y is 4 and the two R1s are on the same ring atom and combine to form a 6-membered cycloalkyl and the third and fourth R1s are halogen.
In one embodiment y is 4 and the two R1s are on the same ring atom and combine to form a 6-membered cycloalkyl and the third and fourth R1s are fluorine.
In one embodiment y is 2 and the two R1s are on the same ring atom and combine to form a 5 membered cycloalkyl. In one embodiment the cycloalkyl is substituted with one R50 substituent. In one embodiment the cycloalkyl is substituted with two R50 substituents. In one embodiment the cycloalkyl is substituted with NH2. In one embodiment the cycloalkyl is substituted with OR14. In one embodiment the cycloalkyl is substituted with OH. In one embodiment the cycloalkyl is substituted with alkyl. In one embodiment the cycloalkyl is substituted with CH3.
In one embodiment y is 2 and the two R1s are on the same ring atom and combine to form a 6 membered cycloalkyl. In one embodiment the cycloalkyl is substituted with one R50 substituent. In one embodiment the cycloalkyl is substituted with two R50 substituents. In one embodiment the cycloalkyl is substituted with NH2. In one embodiment the cycloalkyl is substituted with OR14. In one embodiment the cycloalkyl is substituted with OH. In one embodiment the cycloalkyl is substituted with alkyl. In one embodiment the cycloalkyl is substituted with CH3.
In one embodiment y is 2 and the two R1s are on the same ring atom and combine to form a 5 membered heterocycle. In one embodiment the heterocycle is substituted with one R50 substituent. In one embodiment the heterocycle is substituted with two R50 substituents. In one embodiment the heterocycle is substituted with NH2. In one embodiment the heterocycle is substituted with OR14. In one embodiment the heterocycle is substituted with OH. In one embodiment the heterocycle is substituted with alky l . In one embodiment the heterocycle is substituted with CH3.
In one embodiment y is 2 and the two R1s are on the same ring atom and combine to form a 6 membered heterocycle. In one embodiment the heterocycle is substituted with one R50 substituent. In one embodiment the heterocycle is substituted with two R50 substituents. In one embodiment the heterocycle is substituted with NH2. In one embodiment the heterocycle is substituted with OR14. In one embodiment the heterocycle is substituted with OH. In one embodiment the heterocycle is substituted with alkyl. In one embodiment the heterocycle is substituted with CH3.
In one embodiment y is 2 and the two R1s are on the same ring atom and combine to form a 5-membered spirocycle selected from:
Figure imgf000105_0001
Figure imgf000106_0001
In one embodiment y is 2 and the two R3s are on the same ring atom and combine to form a 6-membered spirocycle selected from:
Figure imgf000106_0002
Embodiments of R2:
In one embodiment R2 is -NR14C(O)R6, -NR14S(O)R6, -NR14S(O)2R6, -NR14C(S)R6, - OC(O)R6, -OS(O)R6, -OS(O)2R6, -OC(S)R6, -C(O)R6; -C(S)R6, -S(O)R6, or -S(O)2R6.
In one embodiment R2 is -C(O)R6; -C(S)R6, -S(O)R6, or -S(O)2R6.
In one embodiment R2 is -C(O)R6.
In one embodiment R2 is -C(O)NH2.
In one embodiment R2 is -C(O)CH3.
In one embodiment R2 is -S(O)2R6.
In one embodiment R2 is -S(O)2NH2.
Embodiments of R3:
(a) In one embodiment R3 is
Figure imgf000106_0003
(b) In one embodiment R3 is
Figure imgf000107_0001
(c) In one embodiment R3 is
Figure imgf000107_0002
(d) In one embodiment R3 is
Figure imgf000107_0003
(e) In one embodiment R3 is
Figure imgf000107_0004
(f) In one embodiment R3 is
Figure imgf000107_0005
,
(g) Any of embodiments (a)-(f), wherein R5 is hydrogen.
(h) Any of embodiments (a)-(I), wherein R5 is alkyl.
(i) Any of embodiments (a)-(f), wherein R5 is -C(O)R6.
(j) Any of embodiments (a)-(f), wherein R5 is -S(O)2R6.
(k) Any of embodiments (a)-(f), wherein R5 is aryl, cycloalkyl, haloalkyl, heteroaryl, or heterocycle,
(l) Any of embodiments (a)-(f), wherein R5 is -C(S)R6 or -S(O)R6.
(m) Any of embodiments (a)-(f), wherein R5 is -C(O)NR6R6, -C(S)NR6R6, -S(O)NR6R6, or -S(O)2NR6R6
(n) Any of embodiments (a)-(i), wherein R5 is -C(O)OR6, -C(S)OR6, -S(O)OR6, or -S(O)20R6.
(o) In one embodiment RJ is
Figure imgf000107_0006
(p) Embodiment (o), wherein R5 is -C(O)R6.
(q) Embodiment (o), wherein R5 is -S(O)2R6.
(r) Embodiment (o), wherein R5 is aryl, cycloalkyl, haloalkyl, heteroaryl, or heterocycle. (s) Embodiment (o), wherein R5 is -C(S)R6 or -S(O)R6.
(t) Embodiment (o), wherein R5 is -C(O)NR6R6, -C(S)NR6R6. -S(O)NR6R6, or - S(O)2NR6R6.
(u) Embodiment (o), wherein R5 is -C(O)OR6, -C(S)OR6, -S(O)OR6, or -S(O)20R6.
(v) Any of embodiments (a)-(u), wherein each R4 is independently hydrogen, alkyl, aryl, cycloalkyl haloalkyl, heteroaryl, heterocycle, -C(O)R6; -C(S)R6, -S(O)R6, or -S(O)2R6.
(w)Any of embodiments (a)-(u), wherein each R4 is independently alkyl, aryl, cycloalkyl, haloalkyl, heteroaryl, heterocycle, -C(O)R6; -C(S)R6, -S(O)R6, or -S(O)2R6.
(x) Any of embodiments (a)-(u), wherein each R4 is independently alkyl, aryl, cycloalkyl, haloalkyl, heteroaryl, or heterocycle.
(y) Any of embodiments (a)-(u), wherein each R4 is independently alkyl, aryl, cycloalkyl, haloalkyl, heteroaryl, or heterocycle.
(z) Any of embodiments (a)-(u), wherein each R4 is independently alkyl, aryl, or cycloalkyl.
(aa) Any of embodiments (a)-(u), wherein each R4 is independently alkyl.
(bb) Any of embodiments (a)-(aa), wherein x is 0.
(cc) Any of embodiments (a)-(aa), wherein x is 1.
(dd) Any of embodiments (a)-(aa), wherein x is 2.
(ee) Any of embodiments (a)-(aa), wherein x is 3.
(ff) Any of embodiments (a)-(aa), wherein x is 4.
(gg) Any of embodiments (dd)-(ff), wherein each R4 is selected from the same group. (hh) Any of embodiments (dd)-(ff), wherein each R4 is the same.
(ii) Any of embodiments (dd)-(ff), wherein each R4 is different.
Embodiments of R5
In one embodiment R5 is
Figure imgf000108_0001
In one embodiment R5 is
Figure imgf000108_0002
. In one embodiment R5 is
Figure imgf000109_0001
In one embodiment R5 is
In one embodiment R5 is
In one embodiment R5 is
Figure imgf000109_0002
In one embodiment R5 is
Figure imgf000109_0003
In one embodiment R5 is
Figure imgf000109_0004
In one embodiment R5 is
Figure imgf000109_0005
In one embodiment R5 is
Figure imgf000109_0006
In one embodiment R5 is
Figure imgf000109_0008
In one embodiment R5 is
Figure imgf000109_0007
In one embodiment R5 is
In one embodiment R5 is In one embodiment R" is
Figure imgf000110_0001
In one embodiment R5 is
Figure imgf000110_0002
In one embodiment R5 is
Figure imgf000110_0003
In one embodiment R5 is
Figure imgf000110_0004
In one embodiment R5 is
Figure imgf000110_0005
In one embodiment R5 is
Figure imgf000110_0010
In one embodiment R5 is
Figure imgf000110_0011
In one embodiment R5 is
Figure imgf000110_0006
In one embodiment R5 is
Figure imgf000110_0007
In one embodiment R5 is
Figure imgf000110_0008
In one embodiment R is
Figure imgf000110_0009
In one embodiment
Figure imgf000111_0001
In one embodiment
Figure imgf000111_0002
In one embodiment
Figure imgf000111_0003
In one embodiment
Figure imgf000111_0004
In one embodiment
Figure imgf000111_0005
In one embodiment
Figure imgf000111_0006
In one embodiment
Figure imgf000111_0007
In one embodiment
Figure imgf000111_0008
In one embodiment
Figure imgf000111_0009
4
In one embodiment R5 is
Figure imgf000112_0001
In one embodiment R5 is
Figure imgf000112_0002
In one embodiment R5 is
Figure imgf000112_0003
In one embodiment R5 is
Figure imgf000112_0004
In one embodiment R5 is
Figure imgf000112_0005
Embodiments of R11
In one embodiment R11 is hydrogen.
In one embodiment R11 is alkyl.
In one embodiment R11 is -NR12R13.
In one embodiment R11 is -S(O)alkyl.
In one embodiment R11 is -SO2alkyl.
In one embodiment R11 is cycloalkyl.
In one embodiment R11 is heterocycle. In one embodiment R11 is aryl.
In one embodiment R11 is heteroaryl.
In one embodiment R11 is alkyl-aryl.
In one embodiment R11 is alkyl-heteroaryl. Embodiments of R17
In one embodiment R17 is
Figure imgf000113_0001
In one embodiment R17 is
Figure imgf000113_0002
In one embodiment R17 is
Figure imgf000113_0003
In one embodiment R17 is
Figure imgf000113_0004
In one embodiment R17 is
Figure imgf000113_0005
.
In one embodiment R17 is
Figure imgf000113_0006
x
In one embodiment R17 is
Figure imgf000113_0007
In one embodiment R17 is
Figure imgf000113_0008
In one embodiment R17 is
Figure imgf000113_0009
In one embodiment R17 is
Figure imgf000113_0010
In one embodiment R17 is
Figure imgf000113_0011
In one embodiment R17 is
Figure imgf000114_0001
In one embodiment R17 is
Figure imgf000114_0002
In one embodiment R17 is
Figure imgf000114_0003
In one embodiment R17 is
Figure imgf000114_0004
In one embodiment R17 is
Figure imgf000114_0005
Embodiments of R18
In one embodiment R18 is cycloalkyl substituted with one R2 group.
In one embodiment R18 is heterocycle substituted with one R2 group.
In one embodiment R18 is aryl substituted with one R2 group.
In one embodiment R18 is substituted with 1 substituent selected from R4 and R5.
In one embodiment R18 is substituted with 2 substituents independently selected from R4 and R5.
In one embodiment R18 is substituted with 3 substituents independently selected from R4 and R5.
In one embodiment R18 is substituted with 4 substituents independently selected from R4 and R5.
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Additional Embodiments
5 1. In certain embodiments the compound is of Formula:
Figure imgf000120_0002
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0002
or a pharmaceutically acceptable salt thereof;
wherein
y is 0, 1, 2, 3, or 4;
x is 0, 1, 2, 3, or 4;
L is selected from -S(O)-, -S(O)2-, -C(O)-, and -C(S)-;
X1, X2, X3, X4, and X5 are independently selected from N, CH, CR2, and CR4; wherein at least one of X1, X2, X3, X4, and X5 is CR2; and wherein no more than 2 of X1, X2, X3, X4, and X5 are selected to be N;
X6, X7 X8, X9, and X10 are independently selected from N, CH, CR2, and CR4; and wherein no more than 2 of X6, X7, X8, X9, and X10 are selected to be N;
each R1 is independently halogen, -OR7, hydrogen, alky l, aryl, cycloalkyl, haloalkyl, heteroaryl, alkyl-hydroxyl, or heterocycle, wherein two R1s on adjacent ring atom(s) or on the same ring atom may come together with the ring atom(s) to which they are attached to optionally constitute a 3, 4, 5, 6, 7, or 8-membered cycloalkyl or 4, 5, 6, 7, or 8-membered heterocycle that has 1, 2, or 3 heteroatoms selected from N, O, and S; wherein the cycloalkyl or heterocycle formed by combining two R1s with the atom(s) to which they are attached can be optionally substituted with 1 or 2 substituents independently selected from R50;
each R2 is independently -NR14C(O)R6, -NR14S(O)R6, -NR14S(O)2R6, -NR14C(S)R6, - 0C(O)R6, -OS(O)R6, -OS(O)2R6, -OC(S)R6, -C(O)R6; -C(S)R6, -S(O)R6, or -S(O)2R6;
R3 is selected from:
Figure imgf000123_0001
Figure imgf000124_0001
each R4 is independently hydrogen, alky l, aryl, cycloalkyl, haloalkyl, heteroaryl, heterocycle, halogen, cyano, -OR14, -NR14R15, -NR14C(O)R6, -NR14S(O)R6, -NR14S(O)2R6, -NR14C(S)R6, -0C(O)R6, -OS(O)R6, -OS(O)2R6, -OC(S)R6, -C(O)R6; -C(S)R6, -S(O)R6, or -S(O)2R6;
R5 is hydrogen, alkyl, aryl, cycloalkyl, haloalkyl, heteroaryl, heterocycle, -C(O)R6, -C(S)R6, -S(O)R6, -S(O)2R6, -C(O)NR7R7, -C(S)NR7R7, -S(O)NR7R7, -S(O)2NR7R7, -C(O)0R7, -C(S)OR7, -S(O)0R7, or -S(O)20R7;
each R6 is selected independently from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycle, aryl, heteroaryl, NR7R7, and OR7 each of which R6 except hydrogen, NR7R7, and OR7 is optionally substituted with 1, 2, 3, or 4 R8 groups;
each R7 is independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycle, aryl, alkyl-aryl, alkyl-heteroaryl, and heteroaryl each of which R7 except hydrogen is optionally substituted with 1, 2, 3, or 4 R8 groups;
each R8 is independently selected from hydrogen, halogen, haloalkyl, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycle, aryl, heteroaryl, -S(O)2alkyl, NR12R13, alkyl-heteroaryl, alkyl-aryl, and OR12;
R11 is selected from halogen, haloalkyl, hydrogen, alkyl, -NR12R13, -S(O)alkyl, - SO2alkyl, cycloalkyl, heterocycle, aryl, heteroaryl, alkyl-aryl, and alkyl-heteroaryl:
each R12 and R13 are independently selected from hydrogen, alkyl, alkenyl, alkynyl, -C(O)alkyl, -C(S)alkyl, aryl, -SO2alkyl, -SOalkyl, heteroaryl, alkyl-aryl, cycloalkyl, heterocycle, and alkyl-heteroaryl;
each R14 and R15 are independently selected from hydrogen, alkyl, alkenyl, alkynyl, -C(O)R6, -C(O)alkyl, -C(S)alkyl, aryl, -SO2alkyl, heteroaryl, heterocycle, -alkyl-aryl, and -alkyl-heteroaryl;
R16 is selected from hydrogen, halogen, alkyl, hydroxyl, alkoxy, and haloalkyl; R17 is selected from:
Figure imgf000125_0001
R18 is selected from: cycloalkyl, heterocycle, and aryl, each of which R18 is substituted with at least one R2 group, and wherein each R18 is optionally substituted with 1, 2, 3, or 4 substituents independently selected from R4 and R5;
Figure imgf000125_0002
R20 is selected from alkyl, alkenyl, alkynyl, -C(O)R6, -C(O)alkyl, -C(S)alkyl, aryl, -SO2alkyl, heteroaryl, heterocycle, -alkyl-aryl, and -alkyl-heteroaryl;
R21 is selected from hydrogen, alkyl, -S(O)alkyl, -SO2alkyl, cycloalkyl, heterocycle, aryl, heteroaryl, alkyl-aryl, and alkyl-heteroaryl;
R50 is selected from hydrogen, amino, -NHR14, -NR14R15, hydroxyl, OR14, and R4; and y2 is 0, 1, 2, 3, or 4;
each m is independently 0 or 1;
each n is independently 0, 1, or 2;
each R31 is independently alkyl, aryl, cycloalkyl or haloalkyl, wherein each of said alkyl, cycloalkyl and haloalkyl groups optionally includes heteroatoms O, N, or S in place of a carbon in the chain and two R31s on adjacent ring atoms or on the same ring atom together with the ring atom(s) to which they are attached optionally form a 3-8-membered cycle or two R31s on adjacent ring atoms together with the ring atoms to which they are attached optionally form a 6-membered aryl ring; R32 is -(alkylene)m-heterocyclo, -(alkylene)m-heteroaryl, -(alkylene)m-NR33R34, -(alkylene)m-C(O)-NR33R34; -(alkylene)m-C(O)-O-alkyl; -(alkylene)m-O-R35, -(alkylene)m-S(O)n-R35, or -(alkylene)m-S(O)n-NR33R34 any of which may be optionally independently substituted with one or more Rx groups as allowed by valance, and wherein two Rx groups bound to the same or adjacent atom may optionally combine to form a ring;
R32* is -alkylene-heterocyclo, -(alkylene)m-heteroaryl, -alkylene-NR33R34, -(alkylene)m-C(O)-NR33R34; -(alkylene)m-C(O)-O-alkyl; -alkylene-S(O)n-R35, or -(alkylene)m-S(O)n-NR33R34 any of which may be optionally independently substituted with one or more Rx groups as allowed by valance, and wherein two Rx groups bound to the same or adjacent atom may optionally combine to form a ring;
R33 and R34 at each occurrence are independently:
(iii) hydrogen or
(iv) alkyl, cycloalkyl, heterocyclo, aryl, heteroaryl, cycloalkylalkyl, heterocycloalkyl, arylalkyl, or heteroarylalkyl; or R33 and R34 together with the nitrogen atom to which they are attached may combine to form a heterocyclo ring;
R35 is independently:
(i) hydrogen or
(ii) alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclo, aryl, heteroaryl, cycloalkylalkyl, heterocycloalkyl, arylalkyl, or heteroarylalkyl;
Rx at each occurrence is independently selected from halo, cyano, nitro, oxo, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclo, ary l heteroaryl, arylalky l, heteroarylalkyl, cycloalkylalkyl, heterocycloalkyl, -(alkylene)m-OR35, -(alkylene)m-O-alkylene-OR35, -(alkylene)m-S(O)n-R35, -(alkylene)m-NR33R34, -(alkylene)m-CN, -(alkylene)m-C(O)-R35,
-(alkylene)m-C(S)-R35, -(alky lene)m-C (O)-OR35, -(alkylene)m-O-C(O)-R35, -(alkylene)m-C(S)-OR35, -(alkylene)m-C(O)-(alkylene)m-NR33R34, -(alkylene)m-C(S)-NR33R34, -(alkylene)m-N(R33)-C(O)-NR33R34, -(alkylene)m-N(R33)-C(S)-NR33R34, -(alkylene)m-N(R33)-C(O)-R35, -(alkylene)m-N(R33)-C(S)-R35, -(alkylene)m-O-C(O)-NR33R34,
-(alkylene)m-O-C(S)-NR33R34, -(alkylene)m-S02-NR33R34, -(alkylene)m-N(R33)-SO2-R35,
-(alkylene)m-N(R33)-SO2-NR33R34, -(alkylene)m-N(R33)-C(O)-OR35,
-(alkylene)m-N(R33)-C(S)-OR35, or -(alkylene)m-N(R33)-SO2-R35. 2. The compound of embodiment 1 of formula:
Figure imgf000127_0001
or a pharmaceutically acceptable salt thereof.
3. The compound of embodiment 2, wherein R21 is hydrogen.
4. The compound of embodiment 2, wherein R21 is alkyl.
5. The compound of embodiment 2, wherein R21 is halogen.
6. The compound of embodiment 1 of formula:
Figure imgf000127_0002
or a pharmaceutically acceptable salt thereof.
7. The compound of embodiment 6, wherein R20 is alkyl.
8. The compound of embodiment 6, wherein R20 is methyl.
9. The compound of any one of embodiments 1-8, wherein R19 is
Figure imgf000127_0003
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
or a pharmaceutically acceptable salt thereof;
wherein
y is 0, 1, 2, 3, or 4;
x is 0, 1, 2, 3, or 4;
L is selected from -S(O)-, -S(O)2- -C(O)-, and -C(S)-;
X1, X2, X3, X4, and X5 are independently selected from N, CH, CR2, and CR4; wherein at least one of X1, X2, X3, X4, and X5 is CR2; and wherein no more than 2 of X1, X2, X3, X4, and X5 are selected to be N;
X6, X . X8, X9, and X10 are independently selected from N, CH, CR2, and CR4; and wherein no more than 2 of X6, X7, X8, X9, and X10 are selected to be N;
each R1 is independently halogen, -OR7, hydrogen, alkyl, aryl, cycloalkyl, haloalkyl, heteroaryl, alkyl-hydroxyl, or heterocycle, wherein two R4s on adjacent ring atom(s) or on the same ring atom may come together with the ring atom(s) to which they are attached to optionally constitute a 3, 4, 5, 6, 7, or 8-membered cycloalkyl or 4, 5, 6, 7, or 8-membered heterocycle that has 1, 2, or 3 heteroatoms selected from N, O, and S; wherein the cycloalkyl or heterocycle formed by combining two R 1 s with the atom(s) to which they are attached can be optionally substituted with 1 or 2 substituents independently selected from R50;
each R2 is independently -NR14C(O)R6, -NR14S(O)R6, -NR14S(O)2R6, -NR14C(S)R6, -
0C(O)R6, -OS(O)R6, -OS(O)2R6, -OC(S)R6, -C(O)R6; -C(S)R6, -S(O)R6, or -S(O)2R6; R3 is selected from:
Figure imgf000131_0001
each R4 is independently hydrogen, alkyl, aryl, cycloalkyl, haloalkyl, heteroaryl, heterocycle, halogen, cyano, -OR14, -NR14R15, -NR14C(O)R6, -NR14S(O)R6, -NR14S(O)2R6, -NR14C(S)R6, -0C(O)R6, -OS(O)R6, -OS(O)2R6, -OC(S)R6, -C(O)R6; -C(S)R6, -S(O)R6, or -S(O)2R6;
R5 is hydrogen, alkyl, aryl cycloalkyl, haloalkyl, heteroaryl, heterocycle, -C(O)R6, -C(S)R6, -S(O)R6, -S(O)2R6, -C(O)NR6R6, -C(S)NR6R6, -S(O)NR6R6, -S(O)2NR6R6, -C(O)0R6, -C(S)OR6, -S(O)0R6, or -S(O)20R6;
each R6 is selected independently from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocy cle, aryl, heteroaryl, NR7R7, and OR7 each of which R6 except hydrogen, NR7R7, and OR7 is optionally substituted with 1, 2, 3, or 4 R8 groups;
each R7 is independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycle, aryl, alkyl-aryl, alkyl-heteroaryl, and heteroaryl each of which R7 except hydrogen is optionally substituted with 1, 2, 3, or 4 R8 groups;
each R8 is independently selected from hydrogen, halogen, haloalkyl, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycle, aryl, heteroaryl, -S(O)2alkyl, NR12R13, alkyl-heteroaryl, alkyl-aryl, and OR12;
R11 is selected from halogen, haloalkyl, hydrogen, alkyl, -NR12R13, -S(O)alkyl, - SO2alkyl, cycloalkyl, heterocycle, aryl, heteroaryl, alkyl-aryl, and alkyl-heteroaryl;
each R12 and R13 are independently selected from hydrogen, alkyl, alkenyl, alkynyl, -C(O)alkyl, -C(S)alkyl, aryl, -SO2alkyl, -SOalkyl, heteroaryl, alkyl-aryl, cycloalkyl, heterocycle, and alkyl-heteroaryl;
each R14 and R15 are independently selected from hydrogen, alkyl, alkenyl, alkynyl, -C(O)R6, -C(O)alkyl, -C(S)alkyl, aryl, -SO2alkyl, heteroaryl, heterocycle, -alkyl-aryl, and -alkyl-heteroaryl;
R16 is selected from hydrogen, halogen, alkyl, hydroxyl, alkoxy, and haloalkyl; R17 is selected from:
Figure imgf000132_0001
R18 is selected from: cycloalkyl, heterocycle, and aryl, each of which R18 is substituted with at least one R2 group, and wherein each R18 is optionally substituted with 1, 2, 3, or 4 substituents independently selected from R4 and R5; and
R50 is selected from hydrogen, amino, -NHR14, -NR14R15, hydroxyl, OR14, and R4.
Figure imgf000132_0002
Figure imgf000133_0001
or a pharmaceutically acceptable salt thereof;
wherein:
each R1 is independently hydrogen, alkyl, aryl, cycloalkyl, haloalkyl, heteroaryl, alkyl-hydroxyl, or heterocycle, wherein two R1s on adjacent ring atom(s) or on the same ring atom may come together with the ring atom(s) to which they are attached to optionally constitute a 3, 4, 5, 6, 7, or 8-membered cycloalkyl or 4, 5, 6, 7, or 8-membered heterocycle that has 1 , 2, or 3 heteroatoms selected from N, 0, and S; wherein the cycloalkyl or heterocycle formed by combining two R1s with the atom(s) to which they are attached can be optionally substituted with 1 or 2 substituents independently selected from R50;
R3 is selected from:
Figure imgf000133_0002
R11 is selected from hydrogen, alkyl, -NR12R13, -S(O)alkyl, -SO2alkyl, cycloalkyl, heterocycle, aryl, heteroaryl, alkyl-aryl, and alkyl-heteroaryl.
14. The compound of any one of embodiments 1-13, wherein two R3s together with the carbon to which they are attached form a 3-8 membered cycle. 15. The compound of any one of embodiments 1-13, wherein two R3s together with the carbon to which they are attached form a 6-membered carbocycle.
16. The compound of any one of embodiments 1-15, wherein y is 3. 17. The compound of any one of embodiments 1-15, wherein y is 4.
18. The compound of any one of embodiments 1-17, wherein one R1 is halogen.
19. The compound of any one of embodiments 1-17, wherein one R1 is haloalkyl.
20. The compound of any one of embodiments 1-17, wherein one R1 is hydroxyl.
21. The compound of any one of embodiments 1-20, wherein R1 is selected from alkyl, aryl, cycloalkyl, and haloalkyl.
22. The compound of any one of embodiments 1-21, wherein R2 is -C(O)R6; -C(S)R6, - S(O)R6, or -S(O)2R6.
23. The compound of any one of embodiments 1-21, wherein R2 is -C(O)R6.
24. The compound of any one of embodiments 1-21, wherein R2 is -S(O)2R6.
25. The compound of any one of embodiments 1-24, wherein each R6 is selected independently from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycle, aryl, and heteroaryl.
26. The compound of any one of embodiments 1-24, wherein each R6 is selected independently from NR7R7 and OR7.
27. The compound of any one of embodiments 1-21, wherein R2 is -S(O)2NH2.
28. The compound of any one of embodiments 1-27, wherein R11 is hydrogen.
29. The compound of any one of embodiments 1-27, wherein R11 is halogen.
30. The compound of any one of embodiments 1-27, wherein R11 is haloalkyl. 31. The compound of any one of embodiments 1-30, wherein the compound is of Formula:
Figure imgf000135_0001
or a pharmaceutically acceptable salt thereof
32. The compound of any one of embodiments 1-30, wherein the compound is of
Formula:
Figure imgf000135_0003
or a pharmaceutically acceptable salt thereof
33. The compound of any one of embodiments 1-30, wherein the compound is of Formula:
Figure imgf000135_0002
Figure imgf000136_0001
or a pharmaceutically acceptable salt thereof.
34. The compound of any one of embodiments 1-30, wherein the compound is of
Formula:
Figure imgf000136_0002
35. The compound of any one of embodiments 1-30, wherein the compound is of
Formula:
Figure imgf000137_0001
or a pharmaceutically acceptable salt thereof.
36. The compound of any one of embodiments 1-30, wherein the compound is of Formula:
Figure imgf000137_0002
or a pharmaceutically acceptable salt thereof.
37. The compound of any one of embodiments 1-30, wherein the compound is of Formula:
Figure imgf000137_0003
Figure imgf000138_0001
or a pharmaceutically acceptable salt thereof.
38. The compound of any one of embodiments 34-37, wherein R16 is hydroxyl.
39. The compound of any one of embodiments 34-37, wherein R16 is fluorine.
40. The compound of any one of embodiments 1-39, wherein
Figure imgf000138_0002
41. The compound of any one of embodiments 1-39, wherein
Figure imgf000138_0003
42. The compound of any one of embodiments 1-39, wherein X1 is CH.
43. The compound of any one of embodiments 1-39, wherein X1 is N.
44. The compound of any one of embodiments 1-39, wherein X1 is CR2.
45. The compound of any one of embodiments 1-39, wherein X1 is CR4.
46. The compound of any one of embodiments 1-45, wherein X2 is CH. 47. The compound of any one of embodiments 1-45, wherein X2 is N.
48. The compound of any one of embodiments 1-45, wherein X2 is CR2.
49. The compound of any one of embodiments 1-45, wherein X2 is CR4.
50. The compound of any one of embodiments 1-49, wherein X3 is CH.
51. The compound of any one of embodiments 1-49, wherein X3 is N.
52. The compound of any one of embodiments 1-49, wherein X3 is CR2.
53. The compound of any one of embodiments 1-49, wherein X3 is CR4.
54. The compound of any one of embodiments 1-53, wherein X4 is CH.
55. The compound of any one of embodiments 1-53, wherein X4 is N.
56. The compound of any one of embodiments 1-53, wherein X4 is CR2.
57. The compound of any one of embodiments 1-53, wherein X4 is CR4.
58. The compound of any one of embodiments 1-57, wherein X1 is CH.
59. The compound of any one of embodiments 1-57, wherein X1 is N.
60. The compound of any one of embodiments 1-57, wherein X1 is CR2.
61. The compound of any one of embodiments 1-57, wherein X1 is CR4.
62. The compound of any one of embodiments 1-39, wherein R3 is
Figure imgf000139_0001
63. The compound of any one of embodiments 1-39, wherein R3 is
Figure imgf000140_0001
64. The compound of any one of embodiments 1-39, wherein R3 is
Figure imgf000140_0002
65. The compound of any one of embodiments 1-64, wherein R4 is OR14.
66. The compound of any one of embodiments 1-64, wherein R4 is halogen. 67. The compound of any one of embodiments 1-64, wherein R4 is alkyl.
68. The compound of any one of embodiments 1-39, wherein R3 is alkyl.
69. The compound of any one of embodiments 62-64, wherein x is 0.
70. The compound of any one of embodiments 62-67, wherein x is 1.
71. The compound of any one of embodiments 62-67, wherein x is 2. 72. The compound of any one of embodiments 62-67, wherein x is 3.
73. The compound of any one of embodiments 1-30, wherein the compound is of Formula:
Figure imgf000140_0003
Figure imgf000141_0001
or a pharmaceutically acceptable salt thereof.
74. The compound of any one of embodiment 73, wherein the compound is of Formula:
Figure imgf000141_0002
or a pharmaceutically acceptable salt thereof.
75. The compound of any one of embodiment 73, wherein the compound is of Formula:
Figure imgf000141_0003
or a pharmaceutically acceptable salt thereof.
76. The compound of any one of embodiments 73-75, wherein R17 is selected from:
Figure imgf000141_0004
77. The compound of any one of embodiments 73-75, wherein R17 is selected from:
Figure imgf000141_0005
78. The compound of any one of embodiments 73-75, wherein R is selected from:
Figure imgf000142_0001
79. The compound of any one of embodiments 73-75, wherein R17 is selected from:
Figure imgf000142_0002
80. The compound of any one of embodiment 73, wherein the compound is of Formula:
Figure imgf000142_0003
or a pharmaceutically acceptable salt thereof.
81. The compound of any one of embodiment 73, wherein the compound is of Formula:
Figure imgf000142_0004
or a pharmaceutically acceptable salt thereof.
82. The compound of embodiment 80 or 81, wherein R18 is cycloalkyl, each of which R18 is substituted with at least one R2 group, and wherein each R18 is optionally substituted with 1, 2, 3, or 4 substituents independently selected from R4 and R5. 83. The compound of embodiment 80 or 81, wherein R18 is heterocycle, each of which R18 is substituted with at least one R2 group, and wherein each R18 is optionally substituted with 1, 2, 3, or 4 substituents independently selected from R4 and R5. 84. The compound of embodiment 80 or 81, wherein R18 is aryl, each of which R18 is substituted with at least one R2 group, and wherein each R18 is optionally substituted with 1, 2, 3, or 4 substituents independently selected from R4 and R5.
85. In certain embodiments the compound is selected from:
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
5 or a pharmaceutically acceptable salt thereof. 86. In certain embodiments the compound is selected from:
Figure imgf000147_0001
Figure imgf000148_0001
or a pharmaceutically acceptable salt thereof.
87. In certain embodiments the compound is selected from:
Figure imgf000148_0002
or a pharmaceutically acceptable salt thereof.
88. The compound of embodiment 87 of structure:
Figure imgf000149_0002
or a pharmaceutically acceptable salt thereof.
89. The compound of embodiment 87 of structure:
Figure imgf000149_0003
or a pharmaceutically acceptable salt thereof 90. The compound of embodiment 87 of structure:
Figure imgf000149_0001
or a pharmaceutically acceptable salt thereof. 91. The compound of embodiment 87 of structure:
Figure imgf000150_0001
or a pharmaceutically acceptable salt thereof
92. The compound of embodiment 87 of structure:
Figure imgf000150_0002
or a pharmaceutically acceptable salt thereof.
93. The compound of embodiment 87 of structure:
Figure imgf000150_0003
or a pharmaceutically acceptable salt thereof. 94. The compound of embodiment 87 of structure:
Figure imgf000151_0001
or a pharmaceutically acceptable salt thereof
95. The compound of embodiment 87 of structure:
Figure imgf000151_0002
or a pharmaceutically acceptable salt thereof.
96. The compound of embodiment 87 of structure:
Figure imgf000151_0003
or a pharmaceutically acceptable salt thereof. 97. The compound of embodiment 87 of structure:
Figure imgf000152_0002
or a pharmaceutically acceptable salt thereof 98. The compound of embodiment 87 of structure:
Figure imgf000152_0001
or a pharmaceutically acceptable salt thereof.
99. A method for the treatment of a disorder associated with abnormal cellular proliferation comprising administering an effective amount to a host in need thereof of a compound of any one of embodiments 1-98, optionally in a pharmaceutically acceptable carrier is provided.
100. The method of embodiment 99, wherein the host is a human.
101. The method of embodiment 99 or 100, wherein the disorder is an inflammatory disorder.
102. The method of embodiment 99 or 100, wherein the disorder is a fibrotic disorder. 103. The method of embodiment 99 or 100, wherein the disorder is an autoimmune disorder.
104. The method of embodiment 99 or 100, wherein the disorder is a tumor.
105. The method of embodiment 99 or 100, wherein the disorder is a cancer.
106. The method of embodiment 99 or 100, wherein the disorder is rheumatoid arthritis.
107. In certain embodiments a method of reducing the effect of chemotherapy on healthy cells in a human being treated for cancer or abnormal cell proliferation, wherein said healthy cells are hematopoietic stem cells or hematopoietic progenitor cells, the method comprising administering to the human an effective amount of a compound of any one of embodiments 1-98, optionally in a pharmaceutically acceptable carrier is provided.
108. A pharmaceutical composition comprising a compound of any one of embodiments 1-98 or pharmaceutically acceptable salt thereof in a pharmaceutically acceptable carrier is provided.
109. The pharmaceutical composition of embodiment 108 for treating a disorder associated with abnormal cellular proliferation.
110. The pharmaceutical composition of embodiment 109, wherein the disorder is an inflammatory disorder.
111. The pharmaceutical composition of embodiment 109, wherein the disorder is a fibrotic disorder.
112. The pharmaceutical composition of embodiment 109, wherein the disorder is an autoimmune disorder.
113. The pharmaceutical composition of embodiment 109, wherein the disorder is a tumor. 114. The pharmaceutical composition of embodiment 109, wherein the disorder is a cancer.
115. The pharmaceutical composition of embodiment 109, wherein the disorder is rheumatoid arthritis.
116. The pharmaceutical composition of embodiment 108 for reducing the effect of chemotherapy on healthy cells in a human being treated for cancer or abnormal cell proliferation, wherein said healthy cells are hematopoietic stem cells or hematopoietic progenitor cells.
117. In certain embodiments a compound for use in the manufacture of a medicament to treat a disorder associated with abnormal cellular proliferation, wherein the compound is selected from any of embodiments 1-98 or a pharmaceutically acceptable salt thereof is provided.
118. The compound of embodiment 117, wherein the disorder is an inflammatory disorder.
119. The compound of embodiment 117, wherein the disorder is a fibrotic disorder.
120. The compound of embodiment 117, wherein the disorder is an autoimmune disorder.
121. The compound of embodiment 117, wherein the disorder is a tumor.
122. The compound of embodiment 117, wherein the disorder is a cancer.
123. The compound of embodiment 117, wherein the disorder is rheumatoid arthritis.
124. In certain embodiments a compound for use in the manufacture of a medicament to reduce the effect of chemotherapy on healthy cells in a human being treated for cancer or abnormal cell proliferation, wherein said healthy cells are hematopoietic stem cells or hematopoietic progenitor cells, wherein the compound is selected from any of embodiments 1-98 or a pharmaceutically acceptable salt thereof is provided. 125. In certain embodiments a use of a compound in the treatment of a disorder associated with abnormal cellular proliferation, wherein the compound is selected from any of embodiments 1-98 or a pharmaceutically acceptable salt thereof is provided.
126. The use of embodiment 125, wherein the disorder is an inflammatory disorder.
127. The use of embodiment 125, wherein the disorder is a fibrotic disorder.
128. The use of embodiment 125, wherein the disorder is an autoimmune disorder.
129. The use of embodiment 125, wherein the disorder is a tumor.
130. The use of embodiment 125, wherein the disorder is a cancer.
131. The use of embodiment 125, wherein the disorder is rheumatoid arthritis.
132. In certain embodiments a use of a compound in reducing the effect of chemotherapy on healthy cells in a human being treated for cancer or abnormal cell proliferation, wherein said healthy cells are hematopoietic stem cells or hematopoietic progenitor cells, wherein the compound is selected from any of embodiments 1-98 or a pharmaceutically acceptable salt thereof.
133. In the embodiments of 105, 114, 122 or 138, wherein the cancer is selected from breast cancer, prostate cancer (including androgen-resistant prostate cancer), another cancer of the reproductive system such as endometrial, ovarian or testicular cancer, small cell lung carcinoma, glioblastoma, colon, including metatatic colon, and head and/or neck cancer.
134. In the embodiments of 105, 114, 122 or 138, wherein the cancer is Rb-negative.
135. In the embodiments of 105, 114, 122 or 138, wherein the cancer is Rb-positive.
136. The pharmaceutical composition of embodiment 108 in a solid dosage form.
137. The embodiment of 136, wherein the solid dosage form is for oral delivery. 138. The pharmaceutical composition of embodiment 108 in a dosage form for parenteral delivery.
II. TERMINOLOGY
Compounds are described using standard nomenclature. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this invention belongs.
The compounds in any of the Formulas described herein include racemates, enantiomers, mixtures of enantiomers, diastereomers, mixtures of diastereomers, tautomers, N- oxides, isomers; such as rotamers, as if each is specifically described.
The terms“a” and“an” do not denote a limitation of quantity, but rather denote the presence of at least one of the referenced item. The term“or” means“and/or”. Recitation of ranges of values are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. The endpoints of all ranges are included within the range and independently combinable. All methods described herein can be performed in a suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of examples, or exemplary language (e.g.,“such as”), is intended merely to better illustrate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. Unless defined otherwise, technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this invention belongs.
The present invention includes compounds of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, with at least one desired isotopic substitution of an atom, at an amount above the natural abundance of the isotope, i.e., enriched. Isotopes are atoms having the same atomic number but different mass numbers, i.e., the same number of protons but a different number of neutrons.
Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine and iodine such as 2H, 3H, 11C, 13C, 14C, 15N, 17O, 18O,18F 31P, 32P, 35S, 36CI, and 125I respectively. In one non-limiting embodiment, isotopically labelled compounds can be used in metabolic studies (with 14C), reaction kinetic studies (with, for example 2H or 3H), detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients. In particular, an 18F labeled compound may be particularly desirable for PET or SPECT studies. Isotopically labeled compounds of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
By way of general example and without limitation, isotopes of hydrogen, for example, deuterium (2H) and tritium (¾) may be used anywhere in described structures that achieves the desired result. Alternatively or in addition, isotopes of carbon, e.g., 13C and 14C, may be used.
Isotopic substitutions, for example deuterium substitutions, can be partial or complete. Partial deuterium substitution means that at least one hydrogen is substituted with deuterium. In certain embodiments, the isotope is 90, 95 or 99% or more enriched in an isotope at any location of interest. In one non-limiting embodiment, deuterium is 90, 95 or 99% enriched at a desired location.
In one non-limiting embodiment, the substitution of one or more hydrogen atoms for a deuterium atoms can be provided in any Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV. In one non-limiting embodiment, the substitution of a hydrogen atom for a deuterium atom occurs within a group selected from any of R1, R2, R3, R4, R5, R6, R7, R8, R11, R12, R13, R14, R15, R16, R17, R18, R19, R20, R21, R31, R32, R32*, R33, R34, and R35. For example, when any of the groups are, or contain for example through substitution, methyl, ethyl, or methoxy, the alkyl residue may be deuterated (in non-limiting embodiments, CDH2, CD2H, CD3, CD2 CD3, CD2CD3, CHDCFED, CH2CD3, CHDCHD2, OCDH2, OCD2H, or OCD3 etc.). In certain other embodiments, when two substituents are combined to form a cycle the unsubstituted carbons may be deuterated.
The compound of the present invention may form a solvate with solvents (including water). Therefore, in one non-limiting embodiment, the invention includes a solvated form of the compound. The term "solvate" refers to a molecular complex of a compound of the present invention (including a salt thereof) with one or more solvent molecules. Non-limiting examples of solvents are water, ethanol, dimethyl sulfoxide, acetone and other common organic solvents. The term "hydrate" refers to a molecular complex comprising a compound of the invention and water. Pharmaceutically acceptable solvates in accordance with the invention include those wherein the solvent may be isotopically substituted, e.g. D2O, d6-acetone, d6- DMSO. A solvate can be in a liquid or solid form. A dash that is not between two letters or symbols is used to indicate a point of attachment for a substituent. For example, -(C=O)NH2 is attached through carbon of the keto (C=O) group.
‘“Alkyl” is a branched or straight chain saturated aliphatic hydrocarbon group. In one non-limiting embodiment, the alkyl group contains from 1 to about 12 carbon atoms, more generally from 1 to about 6 carbon atoms or from 1 to about 4 carbon atoms. In one non- limiting embodiment, the alkyl contains from 1 to about 8 carbon atoms. In certain embodiments, the alkyl is C1-C2, C1-C3, C1-C4, C1-C5, or C1-C6. The specified ranges as used herein indicate an alkyl group having each member of the range described as an independent species. For example, the term C1-C6 alkyl as used herein indicates a straight or branched alkyl group having from 1, 2, 3, 4, 5, or 6 carbon atoms and is intended to mean that each of these is described as an independent species. For example, the term C1-C4 alkyl as used herein indicates a straight or branched alkyl group having from 1, 2, 3, or 4 carbon atoms and is intended to mean that each of these is described as an independent species. Examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, /-butyl, n- pentyl, isopentyl, tert- pentyl, neopentyl, n-hexyl, 2-methylpentane, 3-methylpentane, 2,2- dimethylbutane, and 2,3-dimethylbutane. In an alternative embodiment, the alkyl group is optionally substituted. The term“Alkyl” also encompasses cycloalkyl or carbocyclic groups. For example, when a term is used that includes“alk” then“cycloalkyl” or“carbocyclic” can be considered part of the definition, unless unambiguously excluded by the context. For example, and without limitation, the terms alkyl, -O-alkyl, haloalkyl, etc. can all be considered to include the cyclic forms of alkyl, unless unambiguously excluded by context.
As used herein“substituted alkyl” refers to an alkyl group that is substituted with the described substituents. If no substituents are explicitly described“substituted alkyl” refers to an alkyl group that is substituted with 1, 2, 3, or 4 substituents independently selected from F, Cl, Br, I, cyano, hydroxy, -O-alkyl, -SH, -Salkyl, -COOH, -COOalkyl, -COalkyl, -COH, -CONH2, -CONHalkyl, -CON(alkyl)2, -OC(O)alkyl, -NHC(O)alkyl, -NalkylC(O)alkyl, nitro, amino, -NHalkyl, N(alkyl)2, cyano, haloalkyl, aryl, heteroaryl, alkenyl, alkynyl, haloalkyl, cycloalkyl, alkyl-aryl, alkyl-heteroaryl, alkyl-cycloalkyl, alkyl-heterocycle, heterocycle, -COOaryl, -COaryl, -CONHaiyl, -CON(alkyl)(aiyl), -OC(O)aryl, -NHC(O)aiyl, -NalkylC(O)aryl, -COOheteroaryl, -COheteroaryl, -CONHheteroaiyl,
-CON(alkyl)(heteroaryl), -OC(O)heteroaryl, -NHC(O)heteroaryl, -NalkylC(O)heteroaryl, -COOheterocycle, -COheterocycle, -CONHheterocycle, -CON(alkyl)(heterocycle), -OC(O)heterocycle, -NHC(O)heterocycle, and -NalkylC(O)heterocycle. “Alkenyl” is a linear or branched aliphatic hydrocarbon groups having one or more carbon-carbon double bonds that may occur at a stable point along the chain. The specified ranges as used herein indicate an alkenyl group having each member of the range described as an independent species, as described above for the alky l moiety. Examples of alkenyl radicals include, but are not limited to ethenyl, propenyl, allyl, propenyl, butenyl and 4-methylbutenyl. The term“alkenyl” also embodies“cis” and“trans” alkenyl geometry or alternatively,“E” and “Z” alkenyl geometry. In an alternative embodiment, the alkenyl group is optionally substituted. The term“Alkenyl” also encompasses cycloalkyl or carbocyclic groups possessing at least one point of unsaturation. As used herein“substituted alkenyl” can be substituted with the groups described above for alkyl.
“Alkynyl” is a branched or straight chain aliphatic hydrocarbon group having one or more carbon-carbon triple bonds that may occur at any stable point along the chain. The specified ranges as used herein indicate an alkynyl group having each member of the range described as an independent species, as described above for the alkyl moiety. Examples of alkynyl include, but are not limited to, ethynyl, propynyl, 1-butynyl, 2-butynyl, 3-butynyl, 1- pentynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1-hexynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl and 5-hexynyl. In an alternative embodiment, the alkynyl group is optionally substituted. The term “Alkynyl” also encompasses cycloalkyl or carbocyclic groups possessing at least one point of unsaturation. As used herein“substituted alkynyl” can be substituted with the groups described above for alkyl.
“Halo” and“Halogen” is fluorine, chlorine, bromine or iodine.
“Haloalkyl” is a branched or straight-chain alkyl groups substituted with 1 or more halo atoms described above, up to the maximum allowable number of halogen atoms. Examples of haloalky l groups include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, pentafluoroethyl, heptafluoropropyl, difluorochloromethyl, dichlorofluoromethyl, difluoroethyl, difluoropropyl, dichloroethyl and dichloropropyl.“Perhaloalkyl” means an alkyl group having all hydrogen atoms replaced with halogen atoms. Examples include but are not limited to, trifluoromethyl and pentafluoroethyl.
As used herein,“aryl” refers to a radical of a monocyclic or polycyclic ( e.g ., bicyclic or tricyclic) 4n+2 aromatic ring system (e.g., having 6, 10, or 14 p electrons shared in a cyclic array) having 6-14 ring carbon atoms and zero heteroatoms provided in the aromatic ring system (“C6- 14 aryl”). In some embodiments, an aryl group has 6 ring carbon atoms (“Ce aryl”; e.g., phenyl). In some embodiments, an aryl group has 10 ring carbon atoms (“C 10 aryl”; e.g., naphthyl such as 1-naphthyl and 2-naphthyl). In some embodiments, an aryl group has 14 ring carbon atoms (“C14 aryl”; e.g., anthracyl). “Aryl” also includes ring systems wherein the aryl ring, as defined above, is fused with one or more cycloalkyl or heterocycle groups wherein the radical or point of attachment is on the aryl ring, and in such instances, the number of carbon atoms continue to designate the number of carbon atoms in the aryl ring system. The one or more fused cycloalkyl or heterocycle groups can be 4 to 7-membered saturated or partially unsaturated cycloalkyl or heterocycle groups. As used herein“substituted aryl” refers to an aiyl group that is substituted with the described substituents. If no substituents are explicitly described“substituted aryl” refers to an aryl group that is substituted with 1, 2, 3, or 4 substituents independently selected from F, Cl, Br, I, cyano, hydroxy, -O-alkyl, -SH, -Salkyl, -COOH, -COOalkyl, -COalkyl, -COH, -CONH2, -CONHalkyl, -CON(alkyl)2, -OC(O)alkyl, -NHC(O)alkyl, -NalkylC(O)alkyl, nitro, amino, -NHalkyl, Ntalkylf. cyano, haloalkyl, aryl, heteroaryl, alkyl, alkenyl, alkynyl, haloalkyl, cycloalkyl, alkyl-aryl, alkyl-heteroaryl, alkyl- cycloalkyl, alkyl-heterocycle, heterocycle, -COOaryl, -COaryl, -CONHaryl, -CON(alkyl)(aryl), -OC(O)aryl, -NHC(O)aryl, -NalkylC(O)aryl, -COOheteroaryl, -COheteroaryl, -CONHheteroaryl, -CON(alkyl)(heteroaryl), -OC(O)heteroaryl, -NHC(O)heteroaryl, -NalkylC(O)heteroaryl, -COOheterocycle, -COheterocycle, -CONHheterocycle, -CON(alkyl)(heterocycle), -OC(O)heterocycle, -NHC(O)heterocycle, and -NalkylC(O)heterocycle.
The terms“heterocyclyl” and“heterocycle” include saturated, and partially saturated heteroatom-containing ring radicals, where the heteroatoms may be selected from nitrogen, sulfur, boron, silicone, and oxygen. Heterocyclic rings comprise monocyclic 3-10 membered rings, as well as 5-16 membered bicyclic ring systems (which can include bridged fused and spiro-fused bicyclic ring systems). It does not include rings containing -O-O-.-O-S- or -S-S- portions. Examples of saturated heterocycle groups include saturated 3- to 6-membered heteromonocyclic groups containing 1 to 4 nitrogen atoms [e g. pyrrolidinyl, imidazolidinyl, piperidinyl, pyrrolinyl, piperazinyl]; saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms [e.g. morpholinyl]; saturated 3 to 6- membered heteromonocyclic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms [e.g., thiazolidinyl]. Examples of partially saturated heterocycle radicals include but are not limited to, dihydrothienyl, dihydropyranyl, dihydrofuryl, and dihydrothiazolyl. Examples of partially saturated and saturated heterocycle groups include but are not limited to, pyrrolidinyl, imidazolidinyl, piperidinyl, pyrrolinyl, pyrazolidinyl, piperazinyl, morpholinyl, tetrahydropyranyl, thiazolidinyl, dihydrothienyl, 2,3-dihydro-benzo[l,4]dioxanyl, indolinyl, isoindolinyl, dihydrobenzothienyl, dihydrobenzofuryl, isochromanyl, chromanyl, 1,2- dihydroquinolyl, 1,2, 3, 4- tetrahydro-isoquinolyl, 1 ,2,3,4-tetrahydro-quinolyl, 2, 3, 4, 4a, 9,9a- hexahydro-lH-3-aza-fluorenyl, 5,6,7- trihydro-1, 2, 4-triazolo[3,4-a]isoquinolyl, 3,4-dihydro- 2H-benzo[l,4]oxazinyl, benzo[l,4]dioxanyl, 2,3- dihydro-1 H-ll -benzo[d]sothiazol-6-yl, dihydropyranyl, dihydrofuryl and dihydrothiazolyl. As used herein“substituted heterocycle” refers to a heterocycle group that is substituted with the described substituents. If no substituents are explicitly described“substituted heterocycle” refers to a heterocycle group that is substituted with 1, 2, 3, or 4 substituents independently selected from oxo, F, Cl, Br, I, cyano, hydroxy, -O-alkyl, -SH, -Salkyl, -COOH, -COOalkyl, -COalkyl, -COH, -CONH2, -CONHalkyl, -CON(alkyl)2, -OC(O)alkyl, -NHC(O)alkyl, -NalkylC(O)alkyl, nitro, amino, -NHalkyl, N(alkyl)2, cyano, haloalkyl, aryl, heteroaryl, alkyl, alkenyl, alkynyl, haloalkyl, cycloalkyl, alkyl-aryl, alkyl-heteroaryl, alkyl-cycloalkyl, alkyl-heterocycle, heterocycle, -COOaryl, -COaryl, -CONHaryl, -CON(alkyl)(aryl), -OC(O)aryl, -NHC(O)aryl, -NalkylC(O)aryl, -COOheteroaryl, -COheteroaryl, -CONHheteroaryl,
-CON(alkyl)(heteroaryl), -OC(O)heteroaryl,-NHC(O)heteroaryl, -NalkylC(O)heteroaryl, -COOheterocycle, -COheterocycle, -CONHheterocycle, -CON(alkyl)(heterocycle), -OC(O)heterocycle, -NHC(O)heterocycle, and -NalkylC(O)heterocycle.
“Heterocycle” also includes groups wherein the heterocyclic radical is fused/condensed with an aryl or carbocycle radical, wherein the point of attachment is the heterocycle ring. For example partially unsaturated condensed heterocyclic group containing 1 to 5 nitrogen atoms, for example, indoline, isoindoline, partially unsaturated condensed heterocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, partially unsaturated condensed heterocyclic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms, and saturated condensed heterocyclic group containing 1 to 2 oxygen or sulfur atoms.
The term“heteroaryl” denotes stable aromatic ring systems that contain one or more heteroatoms selected from O, N, and S, wherein the ring nitrogen and sulfur atom(s) are optionally oxidized, and nitrogen atom(s) are optionally quartemized. Examples include but are not limited to, unsaturated 5 to 6 membered heteromonocyclyl groups containing 1 to 4 nitrogen atoms, such as pyrrolyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazolyl [e g., 4H-l,2,4-triazolyl, IH-1 ,2,3-triazolyl, 2H- 1,2,3-triazolyl]; unsaturated 5- to 6-membered heteromonocyclic groups containing an oxygen atom, for example, pyranyl, 2-furyl, 3-furyl, etc.; unsaturated 5 to 6-membered heteromonocyclic groups containing a sulfur atom, for example, 2-thienyl, 3-thienyl, etc.; unsaturated 5- to 6-membered heteromonocyclic groups containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, for example, oxazolyl, isoxazolyl, oxadiazolyl [e.g., 1,2,4-oxadiazolyl, 1.3.4-oxadiazolyl. 1,2,5- oxadiazolyl]; unsaturated 5 to 6-membered heteromonocyclic groups containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms, for example, thiazolyl, thiadiazolyl [e.g., 1,2,4-thiadiazolyl, 1,3,4-thiadiazolyl, 1,2, 5 -thiadiazolyl]. In one embodiment the “heteroaryl” group is a 8, 9, or 10 membered bi cyclic ring system. Examples of 8, 9, or 10 membered bicyclic heteroaryl groups include benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, quinolinyl, isoquinolinyl, benzofuranyl, indolyl, indazolyl, and benzotriazolyl. As used herein“substituted heteroaryl” refers to a heteroaryl group that is substituted with the described substituents. If no substituents are explicitly described“substituted heteroaryl” refers to a heteroaryl group that is substituted with 1, 2, 3, or 4 substituents independently selected from F, Cl, Br, I, cyano, hydroxy, -O-alkyl, -SH, -Salkyl, -COOH, -COOalkyl, -COalkyl, -COH, -CONH2, -CONHalkyl, -CON(alkyl)2, -OC(O)alkyl, -NHC(O)alkyl, -NalkylC(O)alkyl, nitro, amino, -NHalkyl, N(alkyl)2, cyano, haloalkyl, aryl, heteroaryl, alkyl, alkenyl, alkynyl, haloalkyl, cycloalkyl, alkyl-aryl, alkyl-heteroaryl, alkyl-cycloalkyl, alkyl-heterocycle, heterocycle, -COOaryl, -COaryl, -CONHaryl, -CON(alkyl)(aryl), -OC(O)aryl, -NHC(O)aryl, -NalkylC(O)aryl, -COOheteroaryl, -COheteroaryl, -CONHheteroaryl,
-CON(alkyl)(heteroaryl), -OC(O)heteroaryl, -NHC(O)heteroaryl, -NalkylC(O)heteroaryl, -COOheterocycle, -COheterocycle, -CONHheterocycle, -CON(alkyl)(heterocycle), -OC(O)heterocycle, -NHC(O)heterocycle, and -NalkylC(O)heterocycle.
The term“sulfonyl”, whether used alone or linked to other terms such as alkylsulfonyl, denotes respectively divalent radicals -SO2-.
“Alkyl-heterocycle” is an alkyl group as defined herein with a heterocycle substituent. Examples include but are not limited to, piperidylmethyl and morpholinylethyl.
“Alkyl-aryl” is an alkyl group as defined herein with an aryl substituent. Non-limiting
Figure imgf000162_0001
“Alkyl-heteroaryl” is an alkyl group as defined herein with a heteroaryl substituent.
Non-limiting examples of alkyl-heteroaryl groups include:
Figure imgf000162_0002
Figure imgf000163_0001
As used herein, “carbocyclyl”, “carbocyclic”, “carbocycle” or “cycloalkyl” is a saturated or partially unsaturated (i.e., not aromatic) group containing all carbon ring atoms and from 3 to 14 ring carbon atoms (“C3-14 cycloalkyl”) and zero heteroatoms in the non aromatic ring system. In some embodiments, a cycloalkyl group has 3 to 10 ring carbon atoms (“C3-10 cycloalkyl”). In some embodiments, a cycloalkyl group has 3 to 9 ring carbon atoms (“C3-9 cycloalkyl”). In some embodiments, a cycloalkyl group has 3 to 8 ring carbon atoms (“C3-8 cycloalkyl”). In some embodiments, a cycloalkyl group has 3 to 7 ring carbon atoms (“C3-7 cycloalkyl”). In some embodiments, a cycloalkyl group has 3 to 6 ring carbon atoms (“C3-6 cycloalkyl”). In some embodiments, a cycloalkyl group has 4 to 6 ring carbon atoms (“C4-6 cycloalkyl”). In some embodiments, a cycloalkyl group has 5 to 6 ring carbon atoms (“C5-6 cycloalkyl”). In some embodiments, a cycloalkyl group has 5 to 10 ring carbon atoms (“C5-10 cycloalkyl”). Exemplary C3-6 cycloalkyl groups include, without limitation, cyclopropyl (C3), cyclopropenyl (C3), cyclobutyl (C4), cyclobutenyl (C4), cyclopentyl (C5), cyclopentenyl (C5), cyclohexyl (C6) cyclohexenyl (C6) cyclohexadienyl (C6), and the like. Exemplary C3-8 cycloalkyl groups include, without limitation, the aforementioned C3-6 cycloalkyl groups as well as cycloheptyl (C7), cycloheptenyl (C7), cycloheptadienyl (C7), cycloheptatrienyl (C7), cyclooctyl (C8), cyclooctenyl (C8), and the like. Exemplary C3-10 cycloalkyl groups include, without limitation, the aforementioned C3-8 cycloalkyl groups as well as cyclononyl (C9), cyclononenyl (C9), cyclodecyl (C10), cyclodecenyl (C10), and the like. As the foregoing examples illustrate, in certain embodiments, the cycloalkyl group can be saturated or can contain one or more carbon-carbon double or triple bonds. In an alternative embodiment,“cycloalkyl” also includes ring systems wherein the cycloalkyl ring, as defined above, is fused with one heterocycle, aryl or heteroaryl ring wherein the point of attachment is on the cycloalkyl ring, and in such instances, the number of carbons continue to designate the number of carbons in the carbocyclic ring system. In an alternative embodiment, each instance of cycloalky l is optionally substituted with one or more substituents. In certain embodiments, the cycloalkyl group is an unsubstituted C3-14 cycloalkyl.
“Alkyl-cycloalkyl” is an alkyl group as defined herein with a cycloalkyl substituent. Non-limiting examples of alkyl-cycloalkyl groups include:
Figure imgf000164_0001
Figure imgf000164_0002
The term“oxo” as used herein contemplates an oxygen atom attached with a double bond.
“Intrinsic resistance,” also known as primary resistance, as used herein, refers to a condition wherein a cancer is not responsive to the inhibitory effects of initial CDK4/6 inhibitor treatment. Mutations and conditions associated with CDK4/6 inhibitor intrinsic resistance include, but are not limited to: increased activity of cyclin-dependent kinase 1 (CDK1); increased activity of cyclin-dependent kinase 2 (CDK2); loss, deficiency, or absence of retinoblastoma tumor suppressor protein (Rb)(Rb-null); high levels of pl6Ink4a expression; high levels of MYC expression; increased expression of cyclin El, cyclin E2, and cyclin A; and combinations thereof. The cancer may be characterized by reduced expression of the retinoblastoma tumor suppressor protein or a retinoblastoma family member protein or proteins (such as, but not limited to pl07 and pl30). In certain embodiments, a tumor or cancer that is intrinsically resistant to selective CDK4/6 inhibitor inhibition is a tumor or cancer whose cell population, as a whole, does not experience substantial G1 cell-cycle arrest when exposed to a selective CDK4/6 inhibitor. In certain embodiments, a tumor or cancer that is intrinsically resistant to CDK4/6 inhibitor inhibition is a tumor or cancer who has a cell population wherein less than 25%, 20%, 15%, 10%, or 5% of its cells experience G1 cell-cycle arrest when exposed to a selective CDK4/6 inhibitor.
“Acquired resistance,” as used herein, refers to a condition wherein a cancer that was or is initially sensitive to the inhibitory effects of at least one selective CDK4/6 inhibitor becomes non-responsive or less-responsive over time to the effects of that selective CDK4/6 inhibitor. Without wishing to be bound by any one theory, it is believed that acquired resistance to CDK4/6 inhibitors occurs due to one or more additional mutations or genetic alterations in bypass signaling that develops after the onset of CDK4/6 inhibitor treatment regimen. For example, non-limiting exemplary causes of acquired resistance to CDK4/6 inhibitors may be a result of: the development of one or more genetic aberrations associated with“intrinsic resistance.” In addition, other non-limiting exemplary causes of acquired resistance to CDK4/6 inhibitors may include an increase in cyclin E expression; CCNE1/2 amplification; E2F amplification; CDK2 amplification; amplification of CDK6; amplification of CDK4; pl6 amplification; WEE1 overexpression; MDM2 overexpression; CDK7 overexpression; loss of FZR1 ; E1DAC activation; activation of the FGFR pathway; activation of the PI3K/AKT/mTOR pathway; loss of ER or PR expression; higher transcriptional activity of AP-1; epithelial- mesenchymal transition; Smad 3 suppression; autophagy activation; Rbl-loss or inactivating RBI mutations; or a combination thereof A general review of CDK4/6 resistant mechanisms can be found, for example, in Pandey et al., Molecular mechanisms of resistance to CDK4/6 inhibitors in breast cancer: A review. Int. J. Cancer: 00, 1-10 (2019), incorporated herein by reference. In certain embodiments, a tumor or cancer that has acquired resistance to selective CDK4/6 inhibitor inhibition is a tumor or cancer whose cell population, as a whole, no longer experiences substantial G1 cell-cycle arrest when exposed to a selective CDK4/6 inhibitor, resulting in disease progression. In certain embodiments, a tumor or cancer that has acquired resistance to CDK4/6 inhibitor inhibition is a tumor or cancer who has a cell population wherein less than 50%, 40%, 30% 20%, 15%, 10%, or 5% of its cells experience G1 cell-cycle arrest when exposed to a selective CDK4/6 inhibitor, leading to disease progression.
Determining intrinsic resistance to selective CDK4/6 inhibitors, for example by determining the loss or absence of retinoblastoma (Rb) tumor suppressor protein (Rb-null), can be determined through any of the standard assays known to one of ordinary skill in the art. For example, Rb-status in a cancer can be determined by, for example but not limited to, Western Blot, ELISA (enzyme linked immunoadsorbent assay), IHC (immunohistochemistry), and FACS (fluorescent activated cell sorting). The selection of the assay will depend upon the tissue, cell line or surrogate tissue sample that is utilized e.g., for example Western Blot and ELISA may be used with any or all types of tissues, cell lines or surrogate tissues, whereas the IHC method would be more appropriate wherein the tissue utilized in the methods of described herein was a tumor biopsy. FACs analysis would be most applicable to samples that were single cell suspensions such as cell lines and isolated peripheral blood mononuclear cells. See for example, US 20070212736 “Functional Immunohistochemical Cell Cycle Analysis as a Prognostic Indicator for Cancer”.
Alternatively, molecular genetic testing may be used for determination of retinoblastoma gene status. Molecular genetic testing for retinoblastoma includes the following as described in Lohmann and Gallie“Retinoblastoma. Gene Reviews” (2010) or Parsam et al. “A comprehensive, sensitive and economical approach for the detection of mutations in the RBI gene in retinoblastoma” Journal of Genetics, 88(4), 517-527 (2009). Increased activity of CDK1 or CDK2. high levels of MYC expression, increased cyclin E and increased cyclin A can be determined through any of the standard assays known to one of ordinary skill in the art, including but not limited to Western Blot, ELISA (enzyme linked immunoadsorbent assay), IHC (immunohistochemistry), and FACS (fluorescent activated cell sorting). The selection of the assay will depend upon the tissue, cell line, or surrogate tissue sample that is utilized e.g., for example Western Blot and ELISA may be used with any or all types of tissues, cell lines, or surrogate tissues, whereas the IHC method would be more appropriate wherein the tissue utilized in the methods was a tumor biopsy. FACs analysis would be most applicable to samples that were single cell suspensions such as cell lines and isolated peripheral blood mononuclear cells.
Numerous methods can be utilized to measure markers believed to contribute to CDK4/6 inhibitor acquired resistance. Current methods include immunohistochemistry (IHC), immunocytochemistry, mass spectrometry. An alternative method includes the use of immunofluorescence (IF) and image analysis to determine the relative abundance of a protein of interest in formalin-fixed, paraffin-embedded (FFPE) tissue samples. The most frequently used methods for determining gene expression levels is immunohistochemistry (IHC), although western blot allows for assessment of total as well as isoform-specific expression. mRNA from the gene of interest can also be measured by reverse transcription polymerase chain reaction (RT-PCR).
Immunohistochemistry (IHC) and immunocytochemistry (ICC) are techniques employed to localize expression and are dependent on specific epitope-antibody interactions. IHC refers to the use of tissue sections, whereas ICC describes the use of cultured cells or cell suspensions. In both methods, positive staining is visualized using a molecular label, which can be fluorescent or chromogenic. Briefly, samples are fixed to preserve cellular integrity and then subjected to incubation with blocking reagents to prevent non-specific binding of the antibodies. Samples are subsequently incubated with primary and secondary antibodies, and the signal is visualized for microscopic analysis.
The western blot technique uses three elements to identify specific proteins from a complex mixture of proteins extracted from cells: separation by size, transfer to a solid support, and marking target protein using a proper primary and secondary' antibody to visualize. The most common version of this method is immunoblotting. This technique is used to detect specific proteins in a given sample of tissue homogenate or extract. The sample of proteins is first electrophoresed by SDS-PAGE to separate the proteins based on molecular weight. The proteins are then transferred to a membrane where they are probed using antibodies specific to the target protein.
Genomic alterations and mRNA expression can be determined through fluorescence in situ hybridization (FISH), targeted sequencing, and microarray analysis. Commonly mutated genes, as well as differentially expressed and co-expressed genes can be identified.
Fluorescence in situ hybridization (FISH) is a cytogenic technique used for the detection and localization of RNA sequences within tissues or cells. It is particularly important for defining the spatial-temporal patterns of gene expression. FISH relies on fluorescent probes that bind to complementary sequences of the RNA of interest. A series of hybridization steps are performed to achieve signal amplification of the target of interest. This amplification is then viewed using a fluorescent microscope. This technique can be used on formalin-fixed paraffin embedded (FFPE) tissue, frozen tissues, fresh tissues, cells and circulating tumor cells.
Targeted RNA-sequencing (RNA-Seq) is a highly accurate method for selecting and sequencing specific transcripts of interest. It offers both quantitative and qualitative information. Targeted RNA-Seq can be achieved via either enrichment or amplicon-based approaches, both of which enable gene expression analysis in a focused set of genes of interest. Enrichment assays also provide the ability to detect both known and novel gene fusion partners in many sample types including formalin-fixed paraffin-embedded (FFPE) tissue. RNA enrichment provides quantitative expression information as well as the detection of small variants and gene fusions.
In a microarray analysis, mRNA molecules are typically collected from both an experimental sample and a reference sample. For example, the reference sample could be collected from a healthy individual, and the experimental sample could be collected from an individual with a disease such as cancer. The two mRNA samples are then converted into complementary DNA (cDNA), and each sample is labeled with a fluorescent probe of a different color. The experimental cDNA sample may be labeled with a red fluorescent dye, whereas the reference cDNA may be labeled with a green fluorescent dye. The two samples are then mixed together and allowed to hybridize to the microarray slide. Following hybridization, the microarray is scanned to measure the expression of each gene printed on the slide. If the expression of a particular gene is higher in the experimental sample than in the reference sample, then the corresponding spot on the microarray appears red. In contrast, if the expression in the experimental sample is lower than in the reference sample, then the spot appears green. Finally, if there is equal expression in the two samples, then the spot appears yellow. The data gathered through microarrays can be used to create gene expression profiles, which show simultaneous changes in the expression of many genes in response to a particular condition or treatment.
The term“selective CDK4/6 inhibitor” used in the context of the compounds described herein includes compounds that inhibit CDK4 activity, CDK6 activity, or both CDK4 and CDK6 activity at an IC50 molar concentration at least about 300, or 400, or 500, or 1000, or 1500, or 1800, or 2000, or 5000, or 10,000 times less than the IC50 molar concentration necessary to inhibit to the same degree of CDK2 activity in a standard phosphorylation assay.
III. METHODS OF TREATMENT
In one aspect, a method of treating a proliferative disorder in a host, including a human, is provided comprising administering an effective amount of a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, or its pharmaceutically acceptable salt, N-oxide, deuterated derivative, prodrug, and/or a pharmaceutically acceptable composition thereof as described herein optionally in a pharmaceutically acceptable carrier. Non-limiting examples of disorders include tumors, cancers, disorders related to abnormal cellular proliferation, inflammatory disorders, immune disorders, and autoimmune disorders. In certain embodiments the disorder is mediated by CDK2, CDK4, CDK6, or CDK9. In certain embodiments the disorder is mediated by CDK2. In certain embodiments the disorder is mediated by CDK4. In certain embodiments the disorder is mediated by CDK6. In certain embodiments the disorder is mediated by CDK9.
A compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, is useful as therapeutic agents when administered in an effective amount to a host, including a human, to treat a tumor, cancer (solid, non-solid, diffuse, hematological, etc.), abnormal cellular proliferation, immune disorder, inflammatory disorder, blood disorder, a myelo- or lymphoproliferative disorder such as B- or T-cell lymphomas, multiple myeloma, breast cancer, prostate cancer, AML, ALL, CLL, myelodysplastic syndrome (MDS), mesothelioma, renal cell carcinoma (RCC), cholangiocarcinoma, lung cancer, pancreatic cancer, colon cancer, skin cancer, melanoma, Waldenstrom’s macroglobulinemia, Wiskott-Aldrich syndrome, or a post-transplant lymphoproliferative disorder; an autoimmune disorder, for example, Lupus, Crohn’s Disease, Addison disease, Celiac disease, dermatomyositis, Graves disease, thyroiditis, multiple sclerosis, pernicious anemia, reactive arthritis, or type I diabetes; a disease of cardiologic malfunction, including hypercholesterolemia; an infectious disease, including a viral and/or bacterial infection; an inflammatory condition, including asthma, chronic peptic ulcers, tuberculosis, rheumatoid arthritis, periodontitis, ulcerative colitis, or hepatitis.
In one embodiment the compound of the present invention is used to treat breast cancer. In one embodiment the breast cancer is HR+ and HER2-. In one embodiment the breast cancer is HR- and HER2+.
In one embodiment the compound of the present invention is used to treat non-small cell lung cancer (NSCLC). In one embodiment the NSCLC has an EGFR mutation. In one embodiment the NSCLC has an EGFR mutation and an EGFR inhibitor failed (e.g. 2nd line therapy). In one embodiment an ALK inhibitor failed (e.g. 2nd line therapy). In one embodiment the NSCLC has an KRAS mutation.
In one embodiment the compound of the present invention is used to treat prostate cancer. In one embodiment the prostate cancer is castration resistant. In one embodiment a prior chemotherapeutic agent already failed (e.g. 2nd line therapy).
In one embodiment the compound of the present invention is used to treat lymphoma. In one embodiment the lymphoma is mantel cell lymphoma (MCL), marginal zone lymphoma (MZL), chronic lymphocytic leukemia (CLL), follicular lymphoma (FL), or diffuse large B- cell lymphoma (DLBCL). In one embodiment a prior chemotherapeutic agent already failed (e.g. 2nd line therapy).
In one embodiment the compound of the present invention is used to treat melanoma. In one embodiment the melanoma is has a BRAF mutation.
In one embodiment the compound of the present invention is used to treat RAS mutated cancer. In one embodiment the RAS mutated cancer is colon cancer (CLC). In one embodiment the RAS mutated cancer is pancreatic cancer. In one embodiment the RAS mutated cancer is cholangiocarcinoma.
In one embodiment the compound of the present invention is used to treat a gastrointestinal stromal tumor (GIST). In one embodiment the treatment with imatinib or sunitinib already failed (e.g. 2nd line therapy).
Exemplary proliferative disorders include, but are not limited to, benign growths, neoplasms, tumors, cancer (Rb positive or Rb negative), autoimmune disorders, inflammatory disorders graft-versus-host rejection, and fibrotic disorders.
Non-limiting examples of cancers that can be treated according to the present invention include, but are not limited to, acoustic neuroma, adenocarcinoma, adrenal gland cancer, anal cancer, angiosarcoma (e.g., lymphangiosarcoma, lymphangioendotheliosarcoma, hemangiosarcoma), appendix cancer, benign monoclonal gammopathy, biliary cancer (e.g., cholangiocarcinoma), bladder cancer, breast cancer (e.g., adenocarcinoma of the breast, papillary carcinoma of the breast, mammary cancer, medullary carcinoma of the breast), brain cancer (e.g., meningioma; glioma, e.g., astrocytoma, oligodendroglioma; medulloblastoma), bronchus cancer, carcinoid tumor, cervical cancer (e.g., cervical adenocarcinoma), choriocarcinoma, chordoma, craniopharyngioma, colorectal cancer (e.g., colon cancer, rectal cancer, colorectal adenocarcinoma), epithelial carcinoma, ependymoma, endotheliosarcoma (e.g., Kaposi’s sarcoma, multiple idiopathic hemorrhagic sarcoma), endometrial cancer (e.g., uterine cancer, uterine sarcoma), esophageal cancer (e.g., adenocarcinoma of the esophagus, Barrett’s adenocarcinoma), Ewing’s sarcoma, eye cancer (e.g., intraocular melanoma, retinoblastoma), familiar hypereosinophilia, gall bladder cancer, gastric cancer (e.g., stomach adenocarcinoma), gastrointestinal stromal tumor (GIST), head and neck cancer (e.g., head and neck squamous cell carcinoma, oral cancer (e.g., oral squamous cell carcinoma (OSCC), throat cancer (e.g., laryngeal cancer, pharyngeal cancer, nasopharyngeal cancer, oropharyngeal cancer)), hematopoietic cancers (e.g., leukemia such as acute lymphocytic leukemia (ALL) - also known as acute lymphoblastic leukemia or acute lymphoid leukemia (e.g., B-cell ALL, T-cell ALL), acute myelocytic leukemia (AML) (e.g., B-cell AML, T-cell AML), chronic myelocytic leukemia (CML) (e.g., B-cell CML, T-cell CML), and chronic lymphocytic leukemia (CLL) (e.g., B-cell CLL, T-cell CLL); lymphoma such as Hodgkin lymphoma (HL) (e.g., B-cell HL, T-cell HL) and non-Hodgkin lymphoma (NHL) (e.g., B-cell NHL such as diffuse large cell lymphoma (DLCL) (e.g., diffuse large B-cell lymphoma (DLBCL)), follicular lymphoma, chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL), mantle cell lymphoma (MCL), marginal zone B-cell lymphomas (e.g., mucosa- associated lymphoid tissue (MALT) lymphomas, nodal marginal zone B-cell lymphoma, splenic marginal zone B-cell lymphoma), primary mediastinal B-cell lymphoma, Burkitt lymphoma, lymphoplasmacytic lymphoma (i.e.,“Waldenstrom's macroglobulinemia”), hairy cell leukemia (HCL), immunoblastic large cell lymphoma, precursor B-lymphoblastic lymphoma and primary central nervous system (CNS) lymphoma; and T-cell NHL such as precursor T-lymphoblastic lymphoma/leukemia, peripheral T-cell lymphoma (PTCL) (e.g., cutaneous T-cell lymphoma (CTCL) (e.g., mycosis fungiodes, Sezary syndrome), angioimmunoblastic T-cell lymphoma, extranodal natural killer T-cell lymphoma, enteropathy type T-cell lymphoma, subcutaneous panniculitis-like T-cell lymphoma, anaplastic large cell lymphoma); a mixture of one or more leukemia/lymphoma as described above; and multiple myeloma (MM)), heavy chain disease (e.g., alpha chain disease, gamma chain disease, mu chain disease), hemangioblastoma, inflammatory myofibroblastic tumors, immunocytic amyloidosis, kidney cancer (e.g., nephroblastoma a.k.a. Wilms’ tumor, renal cell carcinoma), liver cancer (e.g., hepatocellular cancer (HCC), malignant hepatoma), lung cancer (e.g., bronchogenic carcinoma, small cell lung cancer (SCLC), non-small cell lung cancer (NSCLC), adenocarcinoma of the lung), leiomyosarcoma (LMS), mastocytosis (e.g., systemic mastocytosis), myelodysplastic syndrome (MDS), mesothelioma, myeloproliferative disorder (MPD) (e.g., polycythemia Vera (PV), essential thrombocytosis (ET), agnogenic myeloid metaplasia (AMM) a.k.a. myelofibrosis (MF), chronic idiopathic myelofibrosis, chronic myelocytic leukemia (CML), chronic neutrophilic leukemia (CNL), hypereosinophilic syndrome (HES)), neuroblastoma, neurofibroma (e.g., neurofibromatosis (NF) type 1 or type 2, schwannomatosis), neuroendocrine cancer (e.g., gastroenteropancreatic neuroendoctrine tumor (GEP-NET), carcinoid tumor), osteosarcoma, ovarian cancer (e.g., cystadenocarcinoma, ovarian embryonal carcinoma, ovarian adenocarcinoma), papillary adenocarcinoma, pancreatic cancer (e.g., pancreatic andenocarcinoma, intraductal papillary mucinous neoplasm (IPMN), Islet cell tumors), penile cancer (e.g., Paget’s disease of the penis and scrotum), pineal oma, primitive neuroectodermal tumor (PNT), prostate cancer (e.g., prostate adenocarcinoma), rectal cancer, rhabdomyosarcoma, salivary gland cancer, skin cancer (e.g., squamous cell carcinoma (SCC), keratoacanthoma (KA), melanoma, basal cell carcinoma (BCC)), small bowel cancer (e.g., appendix cancer), soft tissue sarcoma (e.g., malignant fibrous histiocytoma (MFH), liposarcoma, malignant peripheral nerve sheath tumor (MPNST), chondrosarcoma, fibrosarcoma, myxosarcoma), sebaceous gland carcinoma, sweat gland carcinoma, synovioma, testicular cancer (e.g., seminoma, testicular embryonal carcinoma), thyroid cancer (e.g., papillary carcinoma of the thyroid, papillary thyroid carcinoma (PTC), medullary thyroid cancer), urethral cancer, vaginal cancer and vulvar cancer (e.g., Paget’s disease of the vulva).
In another embodiment, the disorder is myelodysplastic syndrome (MDS).
In certain embodiments, the cancer is a hematopoietic cancer. In certain embodiments, the hematopoietic cancer is a lymphoma. In certain embodiments, the hematopoietic cancer is a leukemia. In certain embodiments, the leukemia is acute myelocytic leukemia (AML).
In certain embodiments, the proliferative disorder is a myeloproliferative neoplasm. In certain embodiments, the myeloproliferative neoplasm (MPN) is primary myelofibrosis (PMF).
In certain embodiments, the cancer is a solid tumor. A solid tumor, as used herein, refers to an abnormal mass of tissue that usually does not contain cysts or liquid areas. Different types of solid tumors are named for the type of cells that form them. Examples of classes of solid tumors include, but are not limited to, sarcomas, carcinomas, and lymphomas, as described above herein. Additional examples of solid tumors include, but are not limited to, squamous cell carcinoma, colon cancer, breast cancer, prostate cancer, lung cancer, liver cancer, pancreatic cancer, and melanoma
In certain embodiments, the condition treated with a Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, is a disorder related to abnormal cellular proliferation.
Abnormal cellular proliferation, notably hyperproliferation, can occur as a result of a wide variety of factors, including genetic mutation, infection, exposure to toxins, autoimmune disorders, and benign or malignant tumor induction.
There are a number of skin disorders associated with cellular hyperprobferation. Psoriasis, for example, is a benign disease of human skin generally characterized by plaques covered by thickened scales. The disease is caused by increased proliferation of epidermal cells of unknown cause. Chronic eczema is also associated with significant hyperprobferation of the epidermis. Other diseases caused by hyperprobferation of skin cells include atopic dermatitis, lichen planus, warts, pemphigus vulgaris, actinic keratosis, basal cell carcinoma and squamous cell carcinoma.
Other hyperproliferative cell disorders include blood vessel proliferation disorders, fibrotic disorders, autoimmune disorders, graft-versus-host rejection, tumors and cancers.
Blood vessel proliferative disorders include angiogenic and vasculogemc disorders. Proliferation of smooth muscle cells in the course of development of plaques in vascular tissue cause, for example, restenosis, retinopathies and atherosclerosis. Both cell migration and cell proliferation play a role in the formation of atherosclerotic lesions.
Fibrotic disorders are often due to the abnormal formation of an extracellular matrix. Examples of fibrotic disorders include hepatic cirrhosis and mesangial proliferative cell disorders. Hepatic cirrhosis is characterized by the increase in extracellular matrix constituents resulting in the formation of a hepatic scar. Hepatic cirrhosis can cause diseases such as cirrhosis of the liver. An increased extracellular matrix resulting in a hepatic scar can also be caused by viral infection such as hepatitis. Lipocytes appear to play a major role in hepatic cirrhosis.
Mesangial disorders are brought about by abnormal proliferation of mesangial cells. Mesangial hyperproliferative cell disorders include various human renal diseases, such as glomerulonephritis, diabetic nephropathy, malignant nephrosclerosis, thrombotic micro- angiopathy syndromes, transplant rejection, and glomerulopathies.
Another disease with a proliferative component is rheumatoid arthritis. Rheumatoid arthritis is generally considered an autoimmune disease that is thought to be associated with activity of autoreactive T cells, and to be caused by autoantibodies produced against collagen and IgE.
Other disorders that can include an abnormal cellular proliferative component include Bechet’s syndrome, acute respiratory distress syndrome (ARDS), ischemic heart disease, post- dialysis syndrome, leukemia, acquired immune deficiency syndrome, vasculitis, lipid histiocytosis, septic shock and inflammation in general.
In certain embodiments, a compound of the present invention and its pharmaceutically acceptable derivatives or pharmaceutically acceptable formulations containing these compounds are also useful in the prevention and treatment of HBV infections and other related conditions such as anti-HBV antibody positive and HBV-positive conditions, chronic liver inflammation caused by HBV, cirrhosis, acute hepatitis, fulminant hepatitis, chronic persistent hepatitis, and fatigue. These compounds or formulations can also be used prophylactically to prevent or retard the progression of clinical illness in individuals who are anti-HBV antibody or HBV-antigen positive or who have been exposed to HBV.
In certain embodiments, the condition is associated with an immune response.
Cutaneous contact hypersensitivity and asthma are just two examples of immune responses that can be associated with significant morbidity. Others include atopic dermatitis, eczema, Sjogren's Syndrome, including keratoconjunctivitis sicca secondary to Sjogren's Syndrome, alopecia areata, allergic responses due to arthropod bite reactions, Crohn's disease, aphthous ulcer, iritis, conjunctivitis, keratoconjunctivitis, ulcerative colitis, cutaneous lupus erythematosus, scleroderma, vaginitis, proctitis, and drug eruptions. These conditions may result in any one or more of the following symptoms or signs: itching, swelling, redness, blisters, crusting, ulceration, pain, scaling, cracking, hair loss, scarring, or oozing of fluid involving the skin, eye, or mucosal membranes.
In atopic dermatitis, and eczema in general, immunologically mediated leukocyte infiltration (particularly infiltration of mononuclear cells, lymphocytes, neutrophils, and eosinophils) into the skin importantly contributes to the pathogenesis of these diseases. Chronic eczema also is associated with significant hyperproliferation of the epidermis. Immunologically mediated leukocyte infiltration also occurs at sites other than the skin, such as in the airways in asthma and in the tear producing gland of the eye in keratoconjunctivitis sicca. In one non-limiting embodiment compounds of the present invention are used as topical agents in treating contact dermatitis, atopic dermatitis, eczematous dermatitis, psoriasis, Sjogren's Syndrome, including keratoconjunctivitis sicca secondary to Sjogren's Syndrome, alopecia areata, allergic responses due to arthropod bite reactions, Crohn's disease, aphthous ulcer, iritis, conjunctivitis, keratoconjunctivitis, ulcerative colitis, asthma, allergic asthma, cutaneous lupus erythematosus, scleroderma, vaginitis, proctitis, and drug eruptions. The novel method may also be useful in reducing the infiltration of skin by malignant leukocytes in diseases such as mycosis fungoides. These compounds can also be used to treat an aqueous- deficient dry eye state (such as immune mediated keratoconjunctivitis) in a patient suffering therefrom, by administering the compound topically to the eye.
The term“neoplasia” or“cancer” is used throughout the specification to refer to the pathological process that results in the formation and growth of a cancerous or malignant neoplasm, i.e., abnormal tissue (solid) or cells (non-solid) that grow by cellular proliferation, often more rapidly than normal and continues to grow after the stimuli that initiated the new growth cease. Malignant neoplasms show partial or complete lack of structural organization and functional coordination with the normal tissue and most invade surrounding tissues, can metastasize to several sites, are likely to recur after attempted removal and may cause the death of the patient unless adequately treated. As used herein, the term neoplasia is used to describe all cancerous disease states and embraces or encompasses the pathological process associated with malignant hematogenous, ascitic and solid tumors. Exemplary cancers which may be treated by the present disclosed compounds either alone or in combination with at least one additional anti-cancer agent include squamous-cell carcinoma, basal cell carcinoma, adenocarcinoma, hepatocellular carcinomas, and renal cell carcinomas, cancer of the bladder, bowel, breast, cervix, colon, esophagus, head, kidney, liver, lung, neck, ovary, pancreas, prostate, and stomach; leukemias; benign and malignant lymphomas, particularly Burkitt's lymphoma and Non-Hodgkin's lymphoma; benign and malignant melanomas; myeloproliferative diseases; sarcomas, including Ewing's sarcoma, hemangiosarcoma, Kaposi's sarcoma, liposarcoma, myosarcomas, peripheral neuroepithelioma, synovial sarcoma, gliomas, astrocytomas, oligodendrogliomas, ependymomas, gliobastomas, neuroblastomas, ganglioneuromas, gangliogliomas, medulloblastomas, pineal cell tumors, meningiomas, meningeal sarcomas, neurofibromas, and Schwannomas; bowel cancer, breast cancer, prostate cancer, cervical cancer, uterine cancer, lung cancer, ovarian cancer, testicular cancer, thyroid cancer, astrocytoma, esophageal cancer, pancreatic cancer, stomach cancer, liver cancer, colon cancer, melanoma; carcinosarcoma, Hodgkin's disease, Wilms' tumor and teratocarcinomas. Additional cancers which may be treated using the disclosed compounds according to the present invention include, for example, acute granulocytic leukemia, acute lymphocytic leukemia (ALL), acute myelogenous leukemia (AML), adenocarcinoma, adenosarcoma, adrenal cancer, adrenocortical carcinoma, anal cancer, anaplastic astrocytoma, angiosarcoma, appendix cancer, astrocytoma, Basal cell carcinoma, B-Cell lymphoma, bile duct cancer, bladder cancer, bone cancer, bone marrow cancer, bowel cancer, brain cancer, brain stem glioma, breast cancer, triple (estrogen, progesterone and HER-2) negative breast cancer, double negative breast cancer (two of estrogen, progesterone and HER-2 are negative), single negative (one of estrogen, progesterone and HER-2 is negative), estrogen-receptor positive, HER2- negative breast cancer, estrogen receptor-negative breast cancer, estrogen receptor positive breast cancer, metastatic breast cancer, luminal A breast cancer, luminal B breast cancer, Her2- negative breast cancer, HER2-positive or negative breast cancer, progesterone receptor- negative breast cancer, progesterone receptor-positive breast cancer, recurrent breast cancer, carcinoid tumors, cervical cancer, cholangiocarcinoma, chondrosarcoma, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), colon cancer, colorectal cancer, craniopharyngioma, cutaneous lymphoma, cutaneous melanoma, diffuse astrocytoma, ductal carcinoma in situ (DCIS), endometrial cancer, ependymoma, epithelioid sarcoma, esophageal cancer, ewing sarcoma, extrahepatic bile duct cancer, eye cancer, fallopian tube cancer, fibrosarcoma, gallbladder cancer, gastric cancer, gastrointestinal cancer, gastrointestinal carcinoid cancer, gastrointestinal stromal tumors (GIST), germ cell tumor glioblastoma multiforme (GBM), glioma, hairy cell leukemia, head and neck cancer, hemangioendothelioma, Hodgkin lymphoma, hypopharyngeal cancer, infiltrating ductal carcinoma (IDC), infiltrating lobular carcinoma (ILC), inflammatory breast cancer (IBC), intestinal Cancer, intrahepatic bile duct cancer, invasive/infiltrating breast cancer, Islet cell cancer, jaw cancer, Kaposi sarcoma, kidney cancer, laryngeal cancer, leiomyosarcoma, leptomeningeal metastases, leukemia, lip cancer, liposarcoma, liver cancer, lobular carcinoma in situ, low-grade astrocytoma, lung cancer, lymph node cancer, lymphoma, male breast cancer, medullary carcinoma, medulloblastoma, melanoma, meningioma, Merkel cell carcinoma, mesenchymal chondrosarcoma, mesenchymous, mesothelioma metastatic breast cancer, metastatic melanoma metastatic squamous neck cancer, mixed gliomas, monodermal teratoma, mouth cancer mucinous carcinoma, mucosal melanoma, multiple myeloma, Mycosis Fungoides, myelodysplastic syndrome, nasal cavity cancer, nasopharyngeal cancer, neck cancer, neuroblastoma, neuroendocrine tumors (NETs), non-Hodgkin's lymphoma, non-small cell lung cancer (NSCLC), oat cell cancer, ocular cancer, ocular melanoma, oligodendroglioma, oral cancer, oral cavity cancer, oropharyngeal cancer, osteogenic sarcoma, osteosarcoma, ovarian cancer, ovarian epithelial cancer ovarian germ cell tumor, ovarian primary peritoneal carcinoma, ovarian sex cord stromal tumor, Paget's disease, pancreatic cancer, papillary carcinoma, paranasal sinus cancer, parathyroid cancer, pelvic cancer, penile cancer, peripheral nerve cancer, peritoneal cancer, pharyngeal cancer, pheochromocytoma, pilocytic astrocytoma, pineal region tumor, pineoblastoma, pituitary gland cancer, primary central nervous system (CNS) lymphoma, prostate cancer, rectal cancer, renal cell carcinoma, renal pelvis cancer, rhabdomyosarcoma, salivary gland cancer, soft tissue sarcoma, bone sarcoma, sarcoma, sinus cancer, skin cancer, small cell lung cancer (SCLC), small intestine cancer, spinal cancer, spinal column cancer, spinal cord cancer, squamous cell carcinoma, stomach cancer, synovial sarcoma, T-cell lymphoma, testicular cancer, throat cancer, thymoma/thymic carcinoma, thyroid cancer, tongue cancer, tonsil cancer, transitional cell cancer, tubal cancer, tubular carcinoma, undiagnosed cancer, ureteral cancer, urethral cancer, uterine adenocarcinoma, uterine cancer, uterine sarcoma, vaginal cancer, vulvar cancer, T-cell lineage acute lymphoblastic leukemia (T-ALL), T-cell lineage lymphoblastic lymphoma (T- LL), peripheral T-cell lymphoma, Adult T-cell leukemia, Pre-B ALL, Pre-B lymphomas, large B-cell lymphoma, Burkitts lymphoma, B-cell ALL, Philadelphia chromosome positive ALL, Philadelphia chromosome positive CML, juvenile myelomonocytic leukemia (JMML), acute promyelocytic leukemia (a subtype of AML), large granular lymphocytic leukemia, Adult T- cell chronic leukemia, diffuse large B cell lymphoma, follicular lymphoma; Mucosa- Associated Lymphatic Tissue lymphoma (MALT), small cell lymphocytic lymphoma, mediastinal large B cell lymphoma, nodal marginal zone B cell lymphoma (NMZL); splenic marginal zone lymphoma (SMZL); intravascular large B-cell lymphoma; primary effusion lymphoma; or lymphomatoid granulomatosis;; B-cell prolymphocytic leukemia; splenic lymphoma/leukemia, unclassifiable, splenic diffuse red pulp small B-cell lymphoma; lymphoplasmacytic lymphoma; heavy chain diseases, for example, Alpha heavy chain disease, Gamma heavy chain disease, Mu heavy chain disease, plasma cell myeloma, solitary plasmacytoma of bone; extraosseous plasmacytoma; primary cutaneous follicle center lymphoma, T cell/histocyte rich large B-cell lymphoma, DLBCL associated with chronic inflammation; Epstein-Barr virus (EBV)+ DLBCL of the elderly; primary mediastinal (thymic) large B-cell lymphoma, primary cutaneous DLBCL, leg type, ALK+ large B-cell lymphoma, plasmablastic lymphoma; large B-cell lymphoma arising in HHV8-associated multicentric, Castleman disease; B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma, or B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma and classical Hodgkin lymphoma.
In another aspect, a method of increasing BIM expression (e.g., BCLC2L11 expression) is provided to induce apoptosis in a cell comprising contacting a compound of the present invention or a pharmaceutically acceptable composition, salt, isotopic analog, or prodrug thereof with the cell. In certain embodiments, the method is an in vitro method. In certain embodiments, the method is an in vivo method. BCL2L11 expression is tightly regulated in a cell. BCL2L11 encodes for BIM, a proapoptotic protein. BCL2L11 is downregulated in many cancers and BIM is inhibited in many cancers, including chronic myelocytic leukemia (CML) and non-small cell lung cancer (NSCLC) and that suppression of BCL2L11 expression can confer resistance to tyrosine kinase inhibitors. See, e.g., Ng et al, Nat. Med. (2012) 18:521— 528.
In yet another aspect, a method of treating a condition associated with angiogenesis is provided, such as, for example, a diabetic condition (e.g., diabetic retinopathy), an inflammatory condition (e.g., rheumatoid arthritis), macular degeneration, obesity, atherosclerosis, or a proliferative disorder, comprising administering to a subject in need thereof a compound of the present invention or a pharmaceutically acceptable composition, salt, isotopic analog, or prodrug thereof.
In certain embodiments, the condition associated with angiogenesis is macular degeneration. In certain embodiments, provided is a method of treating macular degeneration comprising administering to a subject in need thereof a compound of the present invention or a pharmaceutically acceptable composition, salt, isotopic analog, or prodrug thereof.
In certain embodiments, the condition associated with angiogenesis is obesity. As used herein,“obesity” and“obese” as used herein, refers to class I obesity, class II obesity, class III obesity and pre-obesity (e.g., being “over-weight”) as defined by the World Health Organization. In certain embodiments, a method of treating obesity is provided comprising administering to a subject in need thereof a compound of the present invention or a pharmaceutically acceptable composition, salt, isotopic analog, or prodrug thereof.
In certain embodiments, the condition associated with angiogenesis is atherosclerosis. In certain embodiments, provided is a method of treating atherosclerosis comprising administering to a subject in need thereof a compound of the present invention or a pharmaceutically acceptable composition, salt, isotopic analog, or prodrug thereof.
In certain embodiments, the condition associated with angiogenesis is a proliferative disorder. In certain embodiments, provided is a method of treating a proliferative disorder comprising administering to a subject in need thereof a compound of the present invention or a pharmaceutically acceptable composition, salt, isotopic analog, or prodrug thereof.
IV. METHODS TO REDUCE THE SIDE EFFECTS RELATED TO CHEMOTHERAPY
In certain embodiments, compounds of the present invention decrease the effect of chemotherapeutic agent toxicity on CDK4/6 replication dependent healthy cells, such as hematopoietic stem cells and hematopoietic progenitor cells (together referred to as HSPCs), and/or renal epithelial cells, in subjects, typically humans, that will be, are being, or have been exposed to the chemotherapeutic agent (typically a DNA-damaging agent).
In one embodiment, the subject has been exposed to a chemotherapeutic agent, and, using a compound described herein, the subject’s CDK4/6-replication dependent healthy cells are placed in G1 arrest following exposure in order to mitigate, for example, DNA damage. In one embodiment, the compound is administered at least ½ hour, at least 1 hour, at least 2 hours, at least 3 hours, at least 4 hours, at least 5 hours, at least 6 hours, at least 7 hours, at least 8 hours, at least 10 hours, at least 12 hours, at least 14 hours, at least 16 hours, at least 18 hours, at least 20 hours or more post chemotherapeutic agent exposure.
In one embodiment, the compound can allow for dose intensification (e.g., more therapy can be given in a fixed period of time) in medically related chemotherapies, which will translate to better efficacy. Therefore, the presently disclosed methods can result in chemotherapy regimens that are less toxic and more effective.
In some embodiments, the use of a compound described herein may result in reduced or be substantially free of off-target effects, for example, related to inhibition of kinases other than CDK4 and/or CDK6 and/or CDK2. Furthermore, in certain embodiments, the use of the compounds described herein should not induce cell cycle arrest in CDK4/6 replication independent cells.
In some embodiments, the use of a compound descnbed herein reduces the risk of undesirable off-target effects including, but not limited to, long term toxicity anti-oxidant effects, and estrogenic effects. Anti-oxidant effects can be determined by standard assays known in the art. For example, a compound with no significant anti-oxidant effects is a compound that does not significantly scavenge free-radicals, such as oxygen radicals. The anti-oxidant effects of a compound can be compared to a compound with known anti-oxidant activity, such as genistein. Thus, a compound with no significant anti-oxidant activity can be one that has less than about 2, 3, 5, 10, 30, or 100 fold anti-oxidant activity relative to genistein. Estrogenic activities can also be determined via known assays. For instance, a non-estrogenic compound is one that does not significantly bind and activate the estrogen receptor. A compound that is substantially free of estrogenic effects can be one that has less than about 2, 3, 5, 10, 20, or 100 fold estrogenic activity relative to a compound with estrogenic activity, e.g., genistein.
V. METHODS TO TREAT ABNORMAL PROLIFERATION OF T-CELLS, B- CELLS AND/OR NK-CELLS
In certain aspects, the invention includes the use of an effective amount of a compound described herein, or its pharmaceutically acceptable salt, prodrug or isotopic variant optionally in a pharmaceutical composition, to treat a host, typically a human, with a selected cancer, tumor, hyperproliferative condition or an inflammatory or immune disorder. Some of the disclosed compounds are highly active against T-cell proliferation. Given the paucity of drugs for T-cell cancers and abnormal proliferation, the identification of such uses represents a substantial improvement in the medical therapy for these diseases.
Abnormal proliferation of T-cells, B-cells, and/or NK-cells can result in a wide range of diseases such as cancer, proliferative disorders and inflammatory/immune diseases. A host, for example a human, afflicted with any of these disorders can be treated with an effective amount of a compound as described herein to achieve a decrease in symptoms (a palliative agent) or a decrease in the underlying disease (a disease modifying agent).
Examples include T-cell or NK-cell lymphoma, for example, but not limited to: peripheral T-cell lymphoma; anaplastic large cell lymphoma, for example anaplastic lymphoma kinase (ALK) positive, ALK negative anaplastic large cell lymphoma, or primary cutaneous anaplastic large cell lymphoma; angioimmunoblastic lymphoma; cutaneous T-cell lymphoma, for example mycosis fungoides, Sezary syndrome, primary cutaneous anaplastic large cell lymphoma, primary cutaneous CD30+ T-cell lymphoproliferative disorder; primary cutaneous aggressive epidermotropic CD8+ cytotoxic T-cell lymphoma; primary cutaneous gamma-delta T-cell lymphoma; primary cutaneous small/medium CD4+ T-cell lymphoma, and lymphomatoid papulosis; Adult T-cell Leukemia/Lymphoma (ATLL); Blastic NK-cell Lymphoma; Enteropathy-type T-cell lymphoma; Hematosplenic gamma-delta T-cell Lymphoma; Lymphoblastic Lymphoma; Nasal NK/T-cell Lymphomas; Treatment-related T- cell lymphomas; for example lymphomas that appear after solid organ or bone marrow transplantation; T-cell prolymphocytic leukemia; T-cell large granular lymphocytic leukemia; Chronic lymphoproliferative disorder of NK-cells; Aggressive NK cell leukemia; Systemic EBV+ T-cell lymphoproliferative disease of childhood (associated with chronic active EBV infection); Hydroa vacciniforme-like lymphoma; Adult T-cell leukemia/ lymphoma; Enteropathy-associated T-cell lymphoma; Hepatosplenic T-cell lymphoma; or Subcutaneous panniculitis-like T-cell lymphoma.
In one embodiment, a compound disclosed herein, or its salt, prodrug, or isotopic variant can be used in an effective amount to treat a host, for example a human, with a lymphoma or lymphocytic or myelocytic proliferation disorder or abnormality. For example, the compounds as described herein can be administered to a host suffering from a Hodgkin Lymphoma or a Non-Hodgkin Lymphoma. For example, the host can be suffering from a Non- Hodgkin Lymphoma such as, but not limited to: an AIDS-Related Lymphoma; Anaplastic Large-Cell Lymphoma; Angioimmunoblastic Lymphoma; Blastic NK-Cell Lymphoma; Burkitt’s Lymphoma; Burkitt-like Lymphoma (Small Non-Cleaved Cell Lymphoma); Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma; Cutaneous T-Cell Lymphoma; Diffuse Large B-Cell Lymphoma; Enteropathy-Type T-Cell Lymphoma; Follicular Lymphoma; Hepatosplenic Gamma-Delta T-Cell Lymphoma; Lymphoblastic Lymphoma; Mantle Cell Lymphoma; Marginal Zone Lymphoma; Nasal T-Cell Lymphoma; Pediatric Lymphoma; Peripheral T-Cell Lymphomas; Primary Central Nervous System Lymphoma; T- Cell Leukemias; Transformed Lymphomas; Treatment-Related T-Cell Lymphomas; or Waldenstrom's Macroglobulinemia.
Alternatively, a compound disclosed herein, or its salt, prodrug, or isotopic variant can be used in an effective amount to treat a host, for example a human, with a Hodgkin Lymphoma, such as, but not limited to: Nodular Sclerosis Classical Hodgkin’s Lymphoma (CHL); Mixed Cellularity CHL; Lymphocyte-depletion CHL; Lymphocyte-rich CHL; Lymphocyte Predominant Hodgkin Lymphoma; or Nodular Lymphocyte Predominant HL.
Alternatively, a compound disclosed herein, or its salt, prodrug, or isotopic variant can be used in an effective amount to treat a host, for example a human with a specific B-cell lymphoma or proliferative disorder such as, but not limited to: multiple myeloma; Diffuse large B cell lymphoma; Follicular lymphoma; Mucosa-Associated Lymphatic Tissue lymphoma (MALT); Small cell lymphocytic lymphoma; Mediastinal large B cell lymphoma; Nodal marginal zone B cell lymphoma (NMZL); Splenic marginal zone lymphoma (SMZL); Intravascular large B-cell lymphoma; Primary effusion lymphoma; or Lymphomatoid granulomatosis;; B-cell prolymphocytic leukemia; Hairy cell leukemia; Splenic lymphoma/leukemia, unclassifiable; Splenic diffuse red pulp small B-cell lymphoma; Hairy cell leukemia-variant; Lymphoplasmacytic lymphoma; Heavy chain diseases, for example, Alpha heavy chain disease, Gamma heavy chain disease, Mu heavy chain disease; Plasma cell myeloma; Solitary plasmacytoma of bone; Extraosseous plasmacytoma; Primary cutaneous follicle center lymphoma; T cell/histiocyte rich large B-cell lymphoma; DLBCL associated with chronic inflammation; Epstein-Barr virus (EBV)+ DLBCL of the elderly; Primary mediastinal (thymic) large B-cell lymphoma; Primary cutaneous DLBCL, leg type; ALK+ large B-cell lymphoma; Plasmablastic lymphoma; Large B-cell lymphoma arising in HHV8- associated multicentric; Castleman disease; B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma; or B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma and classical Hodgkin lymphoma.
In one embodiment, a compound disclosed herein, or its salt, prodrug, or isotopic variant can be used in an effective amount to treat a host, for example a human with leukemia. For example, the host may be suffering from an acute or chronic leukemia of a lymphocytic or myelogenous origin, such as, but not limited to: Acute lymphoblastic leukemia (ALL); Acute myelogenous leukemia (AML); Chronic lymphocytic leukemia (CLL); Chronic myelogenous leukemia (CML); juvenile myelomonocytic leukemia (JMML); hairy cell leukemia (HCL); acute promyelocytic leukemia (a subtype of AML); large granular lymphocytic leukemia; or Adult T-cell chronic leukemia. In one embodiment, the patient suffers from an acute myelogenous leukemia, for example an undifferentiated AML (M0); myeloblastic leukemia (Ml; with/without minimal cell maturation); myeloblastic leukemia (M2; with cell maturation); promyelocytic leukemia (M3 or M3 variant [M3V]); myelomonocytic leukemia (M4 or M4 variant with eosinophilia [M4E]); monocytic leukemia (M5); erythroleukemia (M6); or megakaryoblastic leukemia (M7).
VI. PHARMACEUTICAL COMPOSITIONS AND DOSAGE FORMS
An active compound described herein, or its salt, isotopic analog, or prodrug can be administered in an effective amount to a host to treat any of the disorders described herein using any suitable approach which achieves the desired therapeutic result. The amount and timing of active compound administered will, of course, be dependent on the host being treated, the instructions of the supervising medical specialist, on the time course of the exposure, on the manner of administration, on the pharmacokinetic properties of the particular active compound, and on the judgment of the prescribing physician. Thus, because of host to host variability, the dosages given below are a guideline and the physician can titrate doses of the compound to achieve the treatment that the physician considers appropriate for the host. In considering the degree of treatment desired, the physician can balance a variety of factors such as age and weight of the host, presence of preexisting disease, as well as presence of other diseases.
The pharmaceutical composition may be formulated as any pharmaceutically useful form, e.g., as an aerosol, a cream, a gel, a pill, an injection or infusion solution, a capsule, a tablet, a syrup, a transdermal patch, a subcutaneous patch, a dry powder, an inhalation formulation, in a medical device, suppository, buccal, or sublingual formulation, parenteral formulation, or an ophthalmic solution. Some dosage forms, such as tablets and capsules, are subdivided into suitably sized unit doses containing appropriate quantities of the active components, e.g., an effective amount to achieve the desired purpose.
The therapeutically effective dosage of any active compound described herein will be determined by the health care practitioner depending on the condition, size and age of the patient as well as the route of delivery. In one non-limited embodiment, a dosage from about 0.1 to about 200 mg/kg has therapeutic efficacy, with all weights being calculated based upon the weight of the active compound, including the cases where a salt is employed. In one embodiment, the dosage is at about or greater than 0.1, 0.5, 1, 5, 10, 25, 50, 75, 100, 125, 150, 175, or 200 mg/kg. In some embodiments, the dosage may be the amount of compound needed to provide a serum concentration of the active compound of up to about 10 nM, 50 nM, 100 nM, 200 nM, 300 nM, 400 nM, 500 nM, 600 nM, 700 nM, 800 nM, 900 nM, 1 mM, 5 mM, 10 mM, 20 mM, 30 mM, or 40 mM.
In certain embodiments, the pharmaceutical composition is in a dosage form that contains from about 0.1 mg to about 2000 mg, from about 10 mg to about 1000 mg, from about 100 mg to about 800 mg, or from about 200 mg to about 600 mg of the active compound and optionally from about 0.1 mg to about 2000 mg, from about 10 mg to about 1000 mg, from about 100 mg to about 800 mg, or from about 200 mg to about 600 mg of an additional active agent in a unit dosage form. Examples of dosage forms with at least 5, 10, 15, 20, 25, 50, 100, 200, 250, 300, 400, 500, 600, 700, or 750 mg of active compound, or its salt. The pharmaceutical composition may also include a molar ratio of the active compound and an additional active agent, in a ratio that achieves the desired results.
In some embodiments, compounds disclosed herein or used as described are administered once a day (QD), twice a day (BID), or three times a day (TID). In some embodiments, compounds disclosed herein or used as described are administered at least once a day for at least 21 days, at least 24 days, at least 28 days, at least 35 days, at least 45 days, at least 60 days, at least 75 days, at least 90 days, at least 120 days, at least 180 days, or longer. Compounds disclosed herein or used as described herein may be administered orally, topically, parenterally, by inhalation or spray, sublingually, via implant, including ocular implant, transdermally, via buccal administration, rectally, as an ophthalmic solution, injection, including ocular injection, intravenous, intramuscular, inhalation, intra-aortal, intracranial, subdermal, intraperitoneal, subcutaneous, transnasal, sublingual, or rectal or by other means, in dosage unit formulations containing conventional pharmaceutically acceptable carriers. For ocular delivery, the compound can be administered, as desired, for example, via intravitreal, intrastromal, intracameral, sub-tenon, sub-retinal, retro-bulbar, peribulbar, suprachorodial, conjunctival, subconjunctival, episcleral, periocular, transscleral, retrobulbar, posterior juxtascleral, circumcomeal, or tear duct injections, or through a mucus, mucin, or a mucosal barrier, in an immediate or controlled release fashion or via an ocular device.
In accordance with the presently disclosed methods, an oral administration can be in any desired form such as a solid, gel or liquid, including a solution, suspension, or emulsion. In some embodiments, the compounds or salts are administered by inhalation, intravenously, or intramuscularly as a liposomal suspension. When administered through inhalation the active compound or salt may be in the form of a plurality of solid particles or droplets having any desired particle size, and for example, from about 0.01, 0.1 or 0.5 to about 5, 10, 20 or more microns, and optionally from about 1 to about 2 microns. Compounds as disclosed in the present invention have demonstrated good pharmacokinetic and pharmacodynamics properties, for instance when administered by the oral or intravenous routes.
The pharmaceutical formulations can comprise an active compound described herein or a pharmaceutically acceptable salt thereof, in any pharmaceutically acceptable carrier. If a solution is desired, water may sometimes be the carrier of choice for water-soluble compounds or salts. With respect to the water-soluble compounds or salts, an organic vehicle, such as glycerol, propylene glycol, polyethylene glycol, or mixtures thereof, can be suitable. In the latter instance, the organic vehicle can contain a substantial amount of water. The solution in either instance can then be sterilized in a suitable manner known to those in the art, and for illustration by filtration through a 0.22-micron filter. Subsequent to sterilization, the solution can be dispensed into appropriate receptacles, such as depyrogenated glass vials. The dispensing is optionally done by an aseptic method. Sterilized closures can then be placed on the vials and, if desired, the vial contents can be lyophilized.
Carriers include excipients and diluents and must be of sufficiently high purity and sufficiently low toxicity to render them suitable for administration to the patient being treated. The carrier can be inert or it can possess pharmaceutical benefits of its own. 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.
Classes of carriers include, but are not limited to binders, buffering agents, coloring agents, diluents, disintegrants, emulsifiers, flavorants, glidents, lubricants, preservatives, stabilizers, surfactants, tableting agents, and weting agents. Some carriers may be listed in more than one class, for example vegetable oil may be used as a lubricant in some formulations and a diluent in others. Exemplary pharmaceutically acceptable carriers include sugars, starches, celluloses, powdered tragacanth, malt, gelatin; talc, and vegetable oils. Optional active agents may be included in a pharmaceutical composition, which do not substantially interfere with the activity of the compound of the present invention.
Additionally, auxiliary substances, such as weting or emulsifying agents, biological buffering substances, surfactants, and the like, can be present in such vehicles. A biological buffer can be any solution which is pharmacologically acceptable and which provides the formulation with the desired pH, i.e., a pH in the physiologically acceptable range. Examples of buffer solutions include saline, phosphate buffered saline, Tris buffered saline, Hank’s buffered saline, and the like.
Depending on the intended mode of administration, the pharmaceutical compositions can be in the form of solid, semi-solid or liquid dosage forms, such as, for example, tablets, suppositories, pills, capsules, powders, liquids, suspensions, creams, ointments, lotions or the like, preferably in unit dosage form suitable for single administration of a precise dosage. The compositions will include an effective amount of the selected drug in combination with a pharmaceutically acceptable carrier and, in addition, can include other pharmaceutical agents, adjuvants, diluents, buffers, and the like.
Thus, the compositions of the disclosure can be administered as pharmaceutical formulations including those suitable for oral (including buccal and sub-lingual), rectal, nasal, topical, pulmonary, vaginal or parenteral (including intramuscular, intra-arterial, intrathecal, subcutaneous and intravenous) administration or in a form suitable for administration by inhalation or insufflation. The preferred manner of administration is intravenous or oral using a convenient daily dosage regimen which can be adjusted according to the degree of affliction.
For solid compositions, conventional nontoxic solid carriers include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talc, cellulose, glucose, sucrose, magnesium carbonate, and the like. Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, and the like, an active compound as described herein and optional pharmaceutical adjuvants in an excipient, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to thereby form a solution or suspension. If desired, the pharmaceutical composition to be administered can also contain minor amounts of nontoxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents and the like, for example, sodium acetate, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and the like. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington’s Pharmaceutical Sciences, referenced above.
In yet another embodiment is the use of permeation enhancer excipients including polymers such as: polycations (chitosan and its quaternary ammonium derivatives, poly-L- arginine, animated gelatin); polyanions N-carboxymethyl chitosan, poly-acrylic acid); and, thiolated polymers (carboxymethyl cellulose-cysteine, polycarbophil-cysteine, chitosan- thiobutylamidine, chitosan-thiogly colic acid, chitosan-glutathione conjugates).
For oral administration, the composition will generally take the form of a tablet, capsule, a softgel capsule or can be an aqueous or nonaqueous solution, suspension or syrup. Tablets and capsules are preferred oral administration forms. Tablets and capsules for oral use can include one or more commonly used carriers such as lactose and com starch. Lubricating agents, such as magnesium stearate, are also typically added. Typically, the compositions of the disclosure can be combined with an oral, non-toxic, pharmaceutically acceptable, inert carrier such as lactose, starch, sucrose, glucose, methyl cellulose, magnesium stearate, dicalcium phosphate, calcium sulfate, mannitol, sorbitol and the like. Moreover, when desired or necessary , suitable binders, lubricants, disintegrating agents, and coloring agents can also be incorporated into the mixture. Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, com sweeteners, natural and synthetic gums such as acacia, tragacanth, or sodium alginate, carboxymethylcellulose, polyethylene gly col, waxes, and the like. Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum, and the like.
When liquid suspensions are used, the active agent can be combined with any oral, non- toxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, and the like and with emulsifying and suspending agents. If desired, flavoring, coloring and/or sweetening agents can be added as well. Other optional components for incorporation into an oral formulation herein include, but are not limited to, preservatives, suspending agents, thickening agents, and the like. Parenteral formulations can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solubilization or suspension in liquid prior to injection, or as emulsions. Preferably, sterile injectable suspensions are formulated according to techniques known in the art using suitable carriers, dispersing or wetting agents and suspending agents. The sterile injectable formulation can also be a sterile injectable solution or a suspension in a nontoxic parenterally acceptable diluent or solvent. Among the acceptable vehicles and solvents that can be employed are water, Ringer’s solution and isotonic sodium chloride solution. In addition, sterile, fixed oils, fatty esters or polyols are conventionally employed as solvents or suspending media. In addition, parenteral administration can involve the use of a slow release or sustained release system such that a constant level of dosage is maintained.
Parenteral administration includes intraarticular, intravenous, intramuscular, intradermal, intraperitoneal, and subcutaneous routes, and include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. Administration via certain parenteral routes can involve introducing the formulations of the disclosure into the body of a patient through a needle or a catheter, propelled by a sterile syringe or some other mechanical device such as an continuous infusion system. A formulation provided by the disclosure can be administered using a syringe, injector, pump, or any other device recognized in the art for parenteral administration.
In addition to the active compounds or their salts, the pharmaceutical formulations can contain other additives, such as pH-adjusting additives. In particular, useful pH-adjusting agents include acids, such as hydrochloric acid, bases or buffers, such as sodium lactate, sodium acetate, sodium phosphate, sodium citrate, sodium borate, or sodium gluconate. Further, the formulations can contain antimicrobial preservatives. Useful antimicrobial preservatives include methylparaben, propylparaben, and benzyl alcohol. An antimicrobial preservative is typically employed when the formulations is placed in a vial designed for multi- dose use. The pharmaceutical formulations described herein can be lyophilized using techniques well known in the art.
For oral administration, a pharmaceutical composition can take the form of a solution suspension, tablet, pill, capsule, powder, and the like. Tablets containing various excipients such as sodium citrate, calcium carbonate and calcium phosphate may be employed along with various disintegrants such as starch (e g., potato or tapioca starch) and certain complex silicates, together with binding agents such as polyvinylpyrrolidone, sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, sodium lauryl sulfate, and talc are often very useful for tableting purposes. Solid compositions of a similar type may be employed as fillers in soft and hard-filled gelatin capsules. Materials in this connection also include lactose or milk sugar as well as high molecular weight polyethylene glycols. When aqueous suspensions and/or elixirs are desired for oral administration, the compounds of the presently disclosed host matter can be combined with various sweetening agents, flavoring agents, coloring agents, emulsifying agents and/or suspending agents, as well as such diluents as water, ethanol, propylene glycol, glycerin and various like combinations thereof.
In yet another embodiment of the host matter described herein, there are provided injectable, stable, sterile formulations comprising an active compound as described herein, or a salt thereof, in a unit dosage form in a sealed container. The compound or salt is provided in the form of a lyophilizate, which is capable of being reconstituted with a suitable pharmaceutically acceptable carrier to form liquid formulation suitable for injection thereof into a host. When the compound or salt is substantially water-insoluble, a sufficient amount of emulsifying agent, which is physiologically acceptable, can be employed in sufficient quantity to emulsify the compound or salt in an aqueous carrier. Particularly useful emulsifying agents include phosphatidyl cholines and lecithin.
Additional embodiments provided herein include liposomal formulations of the active compounds disclosed herein. The technology for forming liposomal suspensions is well known in the art. When the compound is an aqueous-soluble salt, using conventional liposome technology, the same can be incorporated into lipid vesicles. In such an instance, due to the water solubility of the active compound, the active compound can be substantially entrained within the hydrophilic center or core of the liposomes. The lipid layer employed can be of any conventional composition and can either contain cholesterol or can be cholesterol-free. When the active compound of interest is water-insoluble, again employing conventional liposome formation technology, the salt can be substantially entrained within the hydrophobic lipid bilayer that forms the structure of the liposome. In either instance, the liposomes that are produced can be reduced in size, as through the use of standard soni cation and homogenization techniques. The liposomal formulations comprising the active compounds disclosed herein can be lyophilized to produce a lyophilizate, which can be reconstituted with a pharmaceutically acceptable carrier, such as water, to regenerate a liposomal suspension. Pharmaceutical formulations also are provided which are suitable for administration as an aerosol by inhalation. These formulations comprise a solution or suspension of a desired compound described herein or a salt thereof, or a plurality of solid particles of the compound or salt. The desired formulations can be placed in a small chamber and nebulized. Nebulization can be accomplished by compressed air or by ultrasonic energy to form a plurality of liquid droplets or solid particles comprising the compounds or salts. The liquid droplets or solid particles may for example have a particle size in the range of about 0.5 to about 10 microns, and optionally from about 0.5 to about 5 microns. In one embodiment, the solid particles provide for controlled release through the use of a degradable polymer. The solid particles can be obtained by processing the solid compound or a salt thereof, in any appropriate manner known in the art, such as by micronization. Optionally, the size of the solid particles or droplets can be from about 1 to about 2 microns. In this respect, commercial nebulizers are available to achieve this purpose. The compounds can be administered via an aerosol suspension of respirable particles in a manner set forth in U.S. Pat. No. 5,628,984, the disclosure of which is incorporated herein by reference in its entirety.
Pharmaceutical formulations also are provided which provide a controlled release of a compound described herein, including through the use of a degradable polymer, as known in the art.
When the pharmaceutical formulations suitable for administration as an aerosol is in the form of a liquid, the formulations can comprise a water-soluble active compound in a carrier that comprises water. A surfactant can be present, which lowers the surface tension of the formulations sufficiently to result in the formation of droplets within the desired size range when hosted to nebulization.
The term "pharmaceutically acceptable salts" as used herein refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with hosts (e.g., human hosts) without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the presently disclosed host matter.
Thus, the term "salts" refers to the relatively non-toxic, inorganic and organic acid addition salts of the presently disclosed compounds. These salts can be prepared during the final isolation and purification of the compounds or by separately reacting the purified compound in its free base form with a suitable organic or inorganic acid and isolating the salt thus formed. Basic compounds are capable of forming a wide variety of different salts with various inorganic and organic acids. Acid addition salts of the basic compounds are prepared by contacting the free base form with a sufficient amount of the desired acid to produce the salt in the conventional manner. The free base form can be regenerated by contacting the salt form with a base and isolating the free base in the conventional manner. The free base forms may differ from their respective salt forms in certain physical properties such as solubility in polar solvents. Pharmaceutically acceptable base addition salts may be formed with metals or amines, such as alkali and alkaline earth metal hydroxides, or of organic amines. Examples of metals used as cations, include, but are not limited to, sodium, potassium, magnesium, calcium, and the like. Examples of suitable amines include, but are not limited to, N,N'- dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N- methylglucamine, and procaine. The base addition salts of acidic compounds are prepared by contacting the free acid form with a sufficient amount of the desired base to produce the salt in the conventional manner. The free acid form can be regenerated by contacting the salt form with an acid and isolating the free acid in a conventional manner. The free acid forms may differ from their respective salt forms somewhat in certain physical properties such as solubility in polar solvents.
Salts can be prepared from inorganic acids sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, nitrate, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydriodic, phosphorus, and the like. Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, nitrate, acetate, oxalate, valerate, oleate, palmitate, stearate, laurate, borate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate mesylate, glucoheptonate, lactobionate, laurylsulphonate and isethionate salts, and the like. Salts can also be prepared from organic acids, such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, etc. and the like. Representative salts include acetate, propionate, caprylate, isobutyrate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, mandelate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, phthalate, benzenesulfonate, toluenesulfonate, phenylacetate, citrate, lactate, maleate, tartrate, methanesulfonate, and the like. Pharmaceutically acceptable salts can include cations based on the alkali and alkaline earth metals, such as sodium, lithium, potassium, calcium, magnesium and the like, as well as non-toxic ammonium, quaternary ammonium, and amine cations including, but not limited to, ammonium, tetramethylammonium, tetraethylammomum, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. Also contemplated are the salts of amino acids such as arginate. gluconate, galacturonate, and the like. See, for example, Berge et al., J. Pharm. Sci., 1977, 66, 1-19, which is incorporated herein by reference.
Preferably, sterile injectable suspensions are formulated according to techniques known in the art using suitable carriers, dispersing or wetting agents and suspending agents. The sterile injectable formulation can also be a sterile injectable solution or a suspension in a nontoxic parenterally acceptable diluent or solvent. Among the acceptable vehicles and solvents that can be employed are water, Ringer’s solution and isotonic sodium chloride solution. In addition, sterile, fixed oils, fatty esters or polyols are conventionally employed as solvents or suspending media. In addition, parenteral administration can involve the use of a slow release or sustained release system such that a constant level of dosage is maintained.
Preparations according to the disclosure for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, or emulsions. Examples of non-aqueous solvents or vehicles are propylene glycol, polyethylene glycol, vegetable oils, such as olive oil and com oil, gelatin, and injectable organic esters such as ethyl oleate. Such dosage forms can also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents. They can be sterilized by, for example, filtration through a bacteria retaining filter, by incorporating sterilizing agents into the compositions, by irradiating the compositions, or by heating the compositions. They can also be manufactured using sterile water, or some other sterile injectable medium, immediately before use.
Sterile injectable solutions are prepared by incorporating one or more of the compounds of the disclosure in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. Thus, for example, a parenteral composition suitable for administration by injection is prepared by stirring 1.5% by weight of active ingredient in 10% by volume propylene glycol and water. The solution is made isotonic with sodium chloride and sterilized.
Formulations suitable for rectal administration are typically presented as unit dose suppositories. These may be prepared by admixing the active disclosed compound with one or more conventional solid carriers, for example, cocoa butter, and then shaping the resulting mixture.
Formulations suitable for topical application to the skin preferably take the form of an ointment, cream, lotion, paste, gel, spray, aerosol, or oil. Carriers which may be used include petroleum jelly, lanoline, polyethylene glycols, alcohols, transdermal enhancers, and combinations of two or more thereof.
Formulations suitable for transdermal administration may be presented as discrete patches adapted to remain in intimate contact with the epidermis of the recipient for a prolonged period of time. Formulations suitable for transdermal administration may also be delivered by iontophoresis (see, for example, Pharmaceutical Research 3 (6):318 (1986)) and typically take the form of an optionally buffered aqueous solution of the active compound. In one embodiment, microneedle patches or devices are provided for delivery of drugs across or into biological tissue, particularly the skin. The microneedle patches or devices permit drug delivery at clinically relevant rates across or into skin or other tissue barriers, with minimal or no damage, pain, or irritation to the tissue.
Formulations suitable for administration to the lungs can be delivered by a wide range of passive breath driven and active power driven single/-multiple dose dry powder inhalers (DPI). The devices most commonly used for respiratory delivery include nebulizers, metered- dose inhalers, and dry powder inhalers. Several types of nebulizers are available, including jet nebulizers, ultrasonic nebulizers, and vibrating mesh nebulizers. Selection of a suitable lung delivery device depends on parameters, such as nature of the drug and its formulation, the site of action, and pathophysiology of the lung.
Additional non-limiting examples of drug delivery devices and methods include, for example, US20090203709 titled“Pharmaceutical Dosage Form For Oral Administration Of Tyrosine Kinase Inhibitor” (Abbott Laboratories); US20050009910 titled“Delivery of an active drug to the posterior part of the eye via subconjunctival or periocular delivery of a prodrug”, US 20130071349 titled“Biodegradable polymers for lowering intraocular pressure”, US 8,481,069 titled“Tyrosine kinase microspheres”, US 8,465,778 titled“Method of making tyrosine kinase microspheres”, US 8,409,607 titled“Sustained release intraocular implants containing tyrosine kinase inhibitors and related methods”, US 8,512,738 and US 2014/0031408 titled“Biodegradable intravitreal tyrosine kinase implants”, US 2014/0294986 titled“Microsphere Drug Delivery System for Sustained Intraocular Release”, US 8,911,768 titled“Methods For Treating Retinopathy With Extended Therapeutic Effect” (Allergan, Inc.); US 6,495,164 titled“Preparation of injectable suspensions having improved injectability” (Alkermes Controlled Therapeutics, Inc ); WO 2014/047439 titled “Biodegradable Microcapsules Containing Filling Material” (Akina, Inc.); WO 2010/132664 titled “Compositions And Methods For Drug Delivery” (Baxter International Inc. Baxter Healthcare SA); US20120052041 titled “Polymeric nanoparticles with enhanced drug loading and methods of use thereof’ (The Brigham and Women’s Hospital, Inc.); US20140178475, US20140248358, and US20140249158 titled “Therapeutic Nanoparticles Comprising a Therapeutic Agent and Methods of Making and Using Same” (BIND Therapeutics, Inc.); US 5,869,103 titled“Polymer microparticles for drug delivery (Danbiosyst UK Ltd.); US 8628801 titled“Pegylated Nanoparticles” (Universidad de Navarra); US2014/0107025 titled “Ocular drug delivery system” (Jade Therapeutics, LLC); US 6,287,588 titled “Agent delivering system comprised of microparticle and biodegradable gel with an improved releasing profde and methods of use thereof’, US 6,589,549 titled“Bioactive agent delivering system comprised of microparticles within a biodegradable to improve release profiles” (Macromed, Inc.); US 6,007,845 and US 5,578,325 titled“Nanoparticles and microparticles of non-linear hydrophilic hydrophobic multiblock copolymers” (Massachusetts Institute of Technology); US20040234611, US20080305172, US20120269894, and US20130122064 titled “Ophthalmic depot formulations for periocular or subconjunctival administration (Novartis Ag); US 6,413,539 titled“Block polymer” (Poly-Med, Inc.); US 20070071756 titled “Delivery of an agent to ameliorate inflammation” (Peyman); US 20080166411 titled “Injectable Depot Formulations And Methods For Providing Sustained Release Of Poorly Soluble Drugs Comprising Nanoparticles” (Pfizer, Inc.); US 6,706,289 titled“Methods and compositions for enhanced delivery of bioactive molecules” (PR Pharmaceuticals, Inc.); and US 8,663,674 titled“Microparticle containing matrices for drug delivery” (Surmodics).
VII. COMBINATION THERAPY
The disclosed compounds of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, can be used in an effective amount alone or in combination with another compound of the present invention or another bioactive agent (therapeutic agent) to treat a host such as a human with a disorder as described herein.
The disclosed compounds described herein can be used in an effective amount alone or in combination with another compound of the present invention or another bioactive agent to treat a host such as a human with a disorder as described herein. The term“bioactive agent” or“therapeutic agent” is used to describe an agent, other than the selected compound according to the present invention, which can be used in combination or alternation with a compound of the present invention to achieve a desired result of therapy. In one embodiment, the compound of the present invention and the bioactive agent are administered in a manner that they are active in vivo during overlapping time periods, for example, have time-period overlapping Cmax, Tmax, AUC or other pharmacokinetic parameter. In another embodiment, the compound of the present invention and the bioactive agent are administered to a host in need thereof that do not have overlapping pharmacokinetic parameter, however, one has a therapeutic impact on the therapeutic efficacy of the other.
In one aspect of this embodiment, the bioactive agent is a chemotherapeutic.
In another aspect of this embodiment, the bioactive agent is a growth factor.
In one aspect of this embodiment, the bioactive agent is an immune modulator, including but not limited to a checkpoint inhibitor, including as non-limiting examples, a PD- 1 inhibitor, PD-L1 inhibitor, PD-L2 inhibitor, CTLA-4 inhibitor, LAG-3 inhibitor, TIM-3 inhibitor, V-domain Ig suppressor of T-cell activation (VISTA) inhibitors, small molecule, peptide, nucleotide, or other inhibitor. In certain aspects, the immune modulator is an antibody, such as a monoclonal antibody.
Immune Checkpoint Inhibitors
Immune checkpoint inhibitors for use in the methods described herein include, but are not limited to PD-1 inhibitors, PD-L1 inhibitors, PD-L2 inhibitors, CTLA-4 inhibitors, LAG- 3 inhibitors, TIM-3 inhibitors, and V-domain Ig suppressor of T-cell activation (VISTA) inhibitors, or combinations thereof. In some embodiments, an immune checkpoint inhibitor is administered in an effective amount in combination with a compound described herein to treat a cancer, including but not limited to, Hodgkin lymphoma, melanoma, non-small cell lung cancer, including NSCLC with EGFR or ALK genomic tumor aberrations, squamous cell carcinoma of the head and neck, small cell lung cancer, hepatocellular carcinoma, renal cell carcinoma, urothelial carcinoma, colorectal cancer, colorectal cancer, hepatocellular carcinoma, renal cell carcinoma, small-cell lung carcinoma, bladder carcinoma, B-cell lymphoma, gastric cancer, cervical cancer, liver cancer, advanced Merkel cell carcinoma, esophageal squamous cell carcinoma, or ovarian cancer.
In one embodiment, the immune checkpoint inhibitor is a PD-1 inhibitor that blocks the interaction of PD-1 and PD-L1 by binding to the PD-1 receptor, and in turn inhibits immune suppression. In one embodiment, the immune checkpoint inhibitor is a PD-1 immune checkpoint inhibitor selected from nivolumab (Opdivo®), pembrolizumab (Keytruda®), pidilizumab, (AMP-224 (AstraZeneca and Medlmmune), PF-06801591 (Pfizer), MEDI0680 (AstraZeneca), PDR001 (Novartis), cemiplimad/REGN2810 (Libtayo® Regeneron), MGA012 (MacroGenics), BGB-A317 (BeiGene) SHR-12-1 (Jiangsu Hengrui Medicine Company and Incyte Corporation), TSR-042 (Tesaro), and the PD-L1/VISTA inhibitor CA-170 (Curis Inc.).
In one embodiment, the immune checkpoint inhibitor is the PD-1 immune checkpoint inhibitor nivolumab (Opdivo®) administered in an effective amount with a compound described herein for the treatment of Hodgkin lymphoma, melanoma, non-small cell lung cancer, including NSCLC with EGFR or ALK genomic tumor aberrations, squamous cell carcinoma of the head and neck, small cell lung cancer, hepatocellular carcinoma, renal cell carcinoma, squamous cell carcinoma, urothelial carcinoma, colorectal cancer, colorectal cancer, hepatocellular carcinoma, or ovarian cancer. Nivolumab has been FDA approved for the use of Hodgkin lymphoma, melanoma, non-small cell lung cancer, including NSCLC with EGFR or ALK genomic tumor aberrations, squamous cell carcinoma of the head and neck, small cell lung cancer, hepatocellular carcinoma, renal cell carcinoma, squamous cell carcinoma, urothelial carcinoma, colorectal cancer, progressive classical Hodgkin lymphoma (cHL), colorectal cancer, urothelial cancer, squamous cell carcinoma of the head and neck, or ovarian cancer. In another aspect of this embodiment, the immune checkpoint inhibitor is the PD-1 immune checkpoint inhibitor pembrolizumab (Keytruda®) administered in an effective amount for the treatment of melanoma, non-small cell lung cancer, small cell lung cancer, head and neck cancer, bladder cancer, urothelial carcinoma, renal cell carcinoma, classical Hodgkin lymphoma, gastric cancer, cervical cancer, liver cancer, primary mediastinal B-cell lymphoma, advanced Merkel cell carcinoma, esophageal squamous cell carcinoma, or urothelial cancer. In an additional aspect of this embodiment, the immune checkpoint inhibitor is the PD-1 immune checkpoint inhibitor pidilizumab (Medivation) administered in an effective amount for refractory diffuse large B-cell lymphoma (DLBCL) or metastatic melanoma. In an additional aspect of this embodiment, the immune checkpoint inhibitor is the PD-1 immune checkpoint inhibitor cemiplimab (Libtayo/Regeneron) administered in an effective amount for cutaneous squamous cell carcinoma.
In one embodiment, the immune checkpoint inhibitor is a PD-L1 inhibitor that blocks the interaction of PD-1 and PD-L1 by binding to the PD-L1 receptor, and in turn inhibits immune suppression. PD-L1 inhibitors include, but are not limited to, atezolizumab, durvalumab, KN035CA-170 (Curis Inc.), and LY3300054 (Eli Lilly). In one embodiment, the PD-L1 inhibitor is atezolizumab. In one embodiment, the PD-L1 inhibitor blocks the interaction between PD-L1 and CD80 to inhibit immune suppression.
In one embodiment, the immune checkpoint inhibitor is the PD-L1 immune checkpoint inhibitor atezolizumab (Tecentriq®) administered in an effective amount for the treatment of metastatic bladder cancer, small cell lung cancer, metastatic melanoma, metastatic non-small cell lung cancer, or metastatic renal cell carcinoma. In another aspect of this embodiment, the immune checkpoint inhibitor is durvalumab (Imfinzi®; AstraZeneca and Medlmmune) administered in an effective amount for the treatment of small cell lung cancer, non-small cell lung cancer, or bladder cancer. In one embodiment, the immune checkpoint inhibitor is the PD- L1 immune checkpoint inhibitor avelumab (Bavencio®; EMD Serono/Pfizer) administered in an effective amount for the treatment of Merkel cell carcinoma or urothelial carcinoma. In yet another aspect of the embodiment, the immune checkpoint inhibitor is KN035 (Alphamab) administered in an effective amount for the treatment of PD-L1 positive solid tumors.
In one aspect of this embodiment, the immune checkpoint inhibitor is a CTLA-4 immune checkpoint inhibitor that binds to CTLA-4 and inhibits immune suppression. CTLA- 4 inhibitors include, but are not limited to, ipilimumab, tremelimumab (AstraZeneca and Medlmmune), AGEN1884 and AGEN2041 (Agenus).
In one embodiment, the CTLA-4 immune checkpoint inhibitor is ipilimumab (Yervoy®) administered in an effective amount for the treatment of metastatic melanoma, adjuvant melanoma, or non-small cell lung cancer.
In another embodiment, the immune checkpoint inhibitor is a LAG-3 immune checkpoint inhibitor. Examples of LAG-3 immune checkpoint inhibitors include, but are not limited to, BMS-986016 (Bristol-Myers Squibb), GSK2831781 (GlaxoSmithKline), IMP321 (Prima BioMed), LAG525 (Novartis), and the dual PD-1 and LAG-3 inhibitor MGD013 (MacroGenics). In yet another aspect of this embodiment, the immune checkpoint inhibitor is a TIM-3 immune checkpoint inhibitor. A specific TIM-3 inhibitor includes, but is not limited to, TSR-022 (Tesaro).
Other immune checkpoint inhibitors for use in the invention described herein include, but are not limited to, B7-H3/CD276 immune checkpoint inhibitors such as MGA217, indoleamine 2,3-dioxygenase (IDO) immune checkpoint inhibitors such as Indoximod and INCB024360, killer immunoglobulin-like receptors (KIRs) immune checkpoint inhibitors such as Lirilumab (BMS-986015), carcinoembryomc antigen cell adhesion molecule (CEACAM) inhibitors (e.g., CEACAM-1, -3 and/or -5). Exemplary anti-CEACAM-1 antibodies are described in WO 2010/125571, WO 2013/082366 and WO 2014/022332, e.g., a monoclonal antibody 34B1, 26H7, and 5F4; or a recombinant form thereof, as described in, e.g., US 2004/0047858, U.S. Pat. No. 7,132,255 and WO 99/052552. In other embodiments, the anti- CEACAM antibody binds to CEACAM-5 as described in, e.g., Zheng et al. PLoS One. 2010 September 2; 5(9). pii: el2529 (D01: 10: 1371/joumal.pone.0021146), or cross-reacts with CEACAM-1 and CEACAM-5 as described in, e.g., WO 2013/054331 and US 2014/0271618. Still other checkpoint inhibitors can be molecules directed to B and T lymphocyte attenuator molecule (BTLA), for example as described in Zhang et al., Monoclonal antibodies to B and T lymphocyte attenuator (BTLA) have no effect on in vitro B cell proliferation and act to inhibit in vitro T cell proliferation when presented in a cis, but not trans, format relative to the activating stimulus, Clin Exp Immunol. 2011 Jan; 163(1): 77-87.
Chemotherapeutic Agents
As contemplated herein, a CDK inhibitor described herein can be in combination with any standard chemotherapeutic agent treatment modality. In one embodiment, a CDK inhibitor described herein can be in combination with any standard chemotherapeutic agent treatment modality and in further combination with an immune checkpoint inhibitor.
In one embodiment, the chemotherapeutic agent is toxic to immune effector cells. In one embodiment the chemotherapeutic agent inhibits cell growth. In one embodiment, the cytotoxic chemotherapeutic agent administered is a DNA damaging chemotherapeutic agent. In one embodiment, the chemotherapeutic agent is a protein synthesis inhibitor, a DNA- damaging chemotherapeutic, an alkylating agent, a topoisomerase inhibitor, an RNA synthesis inhibitor, a DNA complex binder, a thiolate alkylating agent, a guanine alkylating agent, a tubulin binder, DNA polymerase inhibitor, an anticancer enzyme, RAC1 inhibitor, thymidylate synthase inhibitor, oxazophosphorine compound, integrin inhibitor such as cilengitide, camptothecin or homocamptothecin, antifolate or a folate antimetabohte.
In some embodiments, the additional therapeutic agent is selected from elotuzumab, rituximab, lenalidomide, cytarabine, daratumumab, adalimumab, idealisib, gilteritinib, glasdegib, valaciclovir, acalabrutinib, ibrutinib, midostaurin, ruxolitinib, bortezomib, lapatinib, bendamstine, enzalutamide, azacitadine, obinutuzumab, decitabine, erdafitinib, or venetoclax.
In one embodiment the additional therapeutic agent is trastuzumab. In one embodiment the additional therapeutic agent is lapatinib. In one embodiment the compound of the present invention is dosed with 2, 3, or 4 additional therapeutic agents. In one embodiment there are 2 additional therapeutic agents. In one embodiment the two additional therapeutic agents are lapatinib and trastuzumab. In one embodiment the additional therapeutic agent is osimertinib mesylate (Tagrisso®).
In one embodiment the additional therapeutic agent is alectinib (Alecensa®).
In one embodiment the additional therapeutic agent is a MEK inhibitor.
In one embodiment the additional therapeutic agent is an Androgen Receptor ligand.
In one embodiment the additional therapeutic agent is a BTK inhibitor, for example but not limited to ibrutinib (Imbruvica®) or acalabrutinib (Calquence®).
In one embodiment the additional therapeutic agents are a MEK inhibitor and a RAF inhibitor
In one embodiment the additional therapeutic agent is a RAF inhibitor.
In one embodiment the additional therapeutic agent is regorafenib.
Cytotoxic Chemotherapeutic Agents
Cytotoxic, DNA-damaging chemotherapeutic agents tend to be non-specific and, particularly at high doses, toxic to normal, rapidly dividing cells such as HSPC and immune effector cells. As used herein the term“DNA-damaging” chemotherapy or chemotherapeutic agent refers to treatment with a cytostatic or cytotoxic agent (i.e., a compound) to reduce or eliminate the growth or proliferation of undesirable cells, for example cancer cells, wherein the cytotoxic effect of the agent can be the result of one or more of nucleic acid intercalation or binding, DNA or RNA alkylation, inhibition of RNA or DNA synthesis, the inhibition of another nucleic acid-related activity (e.g., protein synthesis), or any other cytotoxic effect. Such compounds include, but are not limited to, DNA damaging compounds that can kill cells. “DNA damaging” chemotherapeutic agents include, but are not limited to, alkylating agents, DNA intercalators, protein synthesis inhibitors, inhibitors of DNA or RNA synthesis, DNA base analogs, topoisomerase inhibitors, telomerase inhibitors, and telomeric DNA binding compounds. For example, alkylating agents include alkyl sulfonates, such as busulfan, improsulfan, and piposulfan; aziridines, such as a benzodizepa, carboquone, meturedepa, and uredepa; ethylenimines and methylmelamines, such as altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide, and trimethylol melamine; nitrogen mustards such as chlorambucil, chlomaphazine, cyclophosphamide, estramustine, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichine, phenesterine, prednimustine, trofosfamide, and uracil mustard; and nitroso ureas, such as carmustme, chlorozotocin, fotemustine, lomustine, nimustine, and ranimustine. Other DNA- damaging chemotherapeutic agents include daunorubicin, doxorubicin, idarubicin, epirubicin, mitomycin, and streptozocin. Chemotherapeutic antimetabolites include gemcitabine, mercaptopurine, thioguanine, cladribine, fludarabine phosphate, fluorouracil (5-FU), lloxuridine, cytarabine, pentostatin, methotrexate, azathioprine, acyclovir, adenine β-1-D- arabinoside, amethopterin, aminopterin, 2-aminopurme, aphidicolin, 8-azaguanine, azaserine, 6-azauracil, 2'-azido-2'-deoxynucleosides, 5-bromodeoxycytidine, cytosine β-1 -D- arabinoside, diazooxynorleucine, dideoxynucleosides, 5-fluorodeoxycytidine, 5- fluorodeoxyuridine, and hydroxyurea.
Chemotherapeutic protein synthesis inhibitors include abrin, aurintricarboxylic acid, chloramphenicol, colicin E3, cycloheximide, diphtheria toxin, edeine A, emetine, erythromycin, ethionine, fluoride, 5-fluorotryptophan, fusidic acid, guanylyl methylene diphosphonate and guanylyl imidodiphosphate, kanamycin, kasugamycin, kirromycin, and O- methyl threonine. Additional protein synthesis inhibitors include modeccin, neomycin, norvaline, pactamycin, paromomycine, puromycin, ricin, shiga toxin, showdomycin, sparsomycin, spectinomycin, streptomycin, tetracycline, thiostrepton, and trimethoprim.
Inhibitors of DNA synthesis, include alkylating agents such as dimethyl sulfate, nitrogen and sulfur mustards; intercalating agents, such as acridine dyes, actinomycins, anthracenes, benzopyrene, ethidium bromide, propidium diiodide-intertwining; and other agents, such as distamycin and netropsin. Topoisomerase inhibitors, such as irinotecan, teniposide, coumermycin, nalidixic acid, novobiocin, and oxolinic acid; inhibitors of cell division, including colcemide, mitoxantrone, colchicine, vinblastine, and vincristine; and RNA synthesis inhibitors including actinomycin D, a-amanitine and other fungal amatoxins, cordycepin (3 '-deoxy adenosine), dichlororibofuranosyl benzimidazole, rifampicine, streptovaricm, and streptolydigm also can be used as the DNA damaging compound.
In one embodiment the chemotherapeutic agent is a DNA complex binder such as camptothecin, or etoposide; a thiolate alkylating agent such as nitrosourea, BCNU, CCNU, ACNU, or fotesmustine; a guanine alkylating agent such as temozolomide, a tubulin binder such as vinblastine, vincristine, vinorelbine, vinflunine, cryptophycin 52, halichondrins, such as halichondrin B, dolastatins, such as dolastatin 10 and dolastatin 15, hemiasterlins, such as hemiasterlin A and hemiasterlin B, colchicine, combrestatins, 2-methoxy estradiol, E7010, paclitaxel, docetaxel, epothilone, discodermolide; a DNA polymerase inhibitor such as cytarabine; an anti cancer enzyme such as asparaginase; a Racl inhibitor such as 6-thioguanine; a thymidylate synthase inhibitor such as capecitabine or 5-FU; a oxazophosphorine compound such as Cytoxan; a integrin inhibitor such as cilengitide; an antifolate such as pralatrexate; a folate antimetabolite such as pemetrexed; or a camptothecin or homocamptothecin such as diflomotecan.
In one embodiment the topoisomerase inhibitor is a type I inhibitor. In another embodiment the topoisomerase inhibitor is a type II inhibitor.
Other DNA-damaging chemotherapeutic agents whose toxic effects can be mitigated by the presently disclosed selective CDK4/6 inhibitors include but are not limited to, cisplatin, hydrogen peroxide, carboplatin, procarbazine, ifosfamide, bleomycin, plicamycin, taxol, transplatinum, thiotepa, oxaliplatin, and the like, and similar acting-type agents. In one embodiment, the DNA damaging chemotherapeutic agent is selected from the group consisting of cisplatin, carboplatin, camptothecin, and etoposide.
Other suitable chemotherapeutic agents include, but are not limited to, radioactive molecules, toxins, also referred to as cytotoxins or cytotoxic agents, which includes any agent that is detrimental to the viability of cells, agents, and liposomes or other vesicles containing chemotherapeutic compounds. General anticancer pharmaceutical agents include: Vincristine (Oncovin®), liposomal vincristine (Marqibo®), Cytarabine (cytosine arabinoside, ara-C, or Cytosar®), L-asparaginase (Elspar®) or PEG-L-asparaginase (pegaspargase or Oncaspar®), Etoposide (VP-16), Teniposide (Vumon®), 6-mercaptopurine (6-MP or Purinethol®), Prednisone, and Dexamethasone (Decadron). Examples of additional suitable chemotherapeutic agents include but are not limited to 5-fluorouracil, dacarbazine, alkylating agents, anthramycin (AMC)), anti-mitotic agents, cis-dichlorodiamine platinum (II) (DDP) cisplatin), diamino dichloro platinum, anthracyclines, antibiotics, antimetabolites, asparaginase, BCG live (intravesical), bleomycin sulfate, calicheamicin, cytochalasin B, dactinomycin (formerly actinomycin), daunorubicin HC1, daunorubicin citrate, denileukin diftitox, dihydroxy anthracin dione, Docetaxel, doxorubicin HC1, E. coli L-asparaginase, Erwinia L-asparaginase, etoposide citrovomm factor, etoposide phosphate, gemcitabine HC1, idarubicin HC1, interferon a-2b, irinotecan HC1, maytansinoid, mechlorethamine HC1, melphalan HC1, mithramycin, mitomycin C, mitotane, polifeprosan 20 with carmustine implant, procarbazine HC1, streptozotocin, teniposide, thiotepa, topotecan HC1, valrubicin, vinblastine sulfate, vincristine sulfate, and vinorelbine tartrate.
Additional cytotoxic chemotherapeutic agents for use with the present invention include: epirubicm, abraxane, taxotere, epothilone, tafluposide, vismodegib, azacytidine, doxifluridine, vindesine, and vinorelbine. In one embodiment the chemotherapeutic agent is not an aromatase inhibitor. In one embodiment the chemotherapeutic agent is not a steroid. In one embodiment the chemotherapeutic agent is not a BCR-ABL inhibitor.
In one embodiment the chemotherapeutic agent is a DNA complex binder. In one embodiment the chemotherapeutic agent is a tubulin binder. In one embodiment the chemotherapeutic agent is an alkylating agent. In one embodiment the chemotherapeutic agent is a thiolate alkylating agent.
Additional Chemotherapeutic Agents
Additional chemotherapeutic agents that may be used as descnbed herein may include 2-methoxyestradiol or 2ME2, fmasunate, etaracizumab (MEDI-522), HLL1, huN901-DMl, atiprimod, saquinavir mesylate, ritonavir, nelfmavir mesylate, indinavir sulfate, plitidepsin, P276-00, tipifamib, lenalidomide, thalidomide, pomalidomide, simvastatin, and celecoxib. Chemotherapeutic agents useful in the present invention include, but are not limited to, Trastuzumab (Herceptin®), Pertuzumab (Perjeta™), Lapatinib (Tykerb®), Gefitinib (Iressa®), Erlotinib (Tarceva®), Cetuximab (Erbitux®), Panitumumab (Vectibix®), Vandetanib (Caprelsa®), Vemurafenib (Zelboraf®), Vorinostat (Zolinza®), Romidepsin (Istodax®), Bexarotene (Targretin®), Alitretinoin (Panretin®), Tretinoin (Vesanoid®), Carfilzomib (Kyprolis™), Pralatrexate (Folotyn®), Bevacizumab (Avastin®), Ziv-aflibercept (Zaltrap®), Sorafenib (Nexavar®), Sunitinib (Sutent®), Pazopanib (Votrient®), Regorafenib (Stivarga®), and Cabozantinib (Cometriq™).
Additional chemotherapeutic agents contemplated include, but are not limited to, a calcineurin inhibitor, e.g. a cyclosporin or an ascomycin, e.g. Cyclosporin A (Neoral®), FK506 (tacrolimus), pimecrolimus, a mTOR inhibitor, e.g. rapamycin or a derivative thereof, e.g. Sirolimus (Rapamune®), Everolimus (Certican®), temsirohmus, zotarolimus, biolimus-7, biolimus-9, a rapalog, e.g. ridaforolimus, campath 1H, a SIP receptor modulator, a dual mTORCl and mTORC2 inhibitor, eg. Vistusertib (AZD2014), e.g. fmgolimod or an analogue thereof, an anti IL-8 antibody, mycophenolic acid or a salt thereof, e.g. sodium salt, or a prodrug thereof, e.g. Mycophenolate Mofetil (CellCept®), OKT3 (Orthoclone OKT3®), Prednisone, ATGAM®, Thymoglobulin®, Brequinar Sodium, OKT4, T10B9.A-3A, 33B3.1, 15- deoxyspergualin, tresperimus, Leflunomide Arava®, anti-CD25, anti-IL2R, Basiliximab (Simulect®), Daclizumab (Zenapax®), mizoribine, dexamethasone, ISAtx-247, SDZ ASM 981 (pimecrolimus, Elidel®), Abatacept, belatacept, LFA31g, etanercept (sold as Enbrel® by ImmuneXcite), adalimumab (Humira®), infliximab (Remicade®), an anti-LFA-1 antibody, natalizumab (Antegren®), Enlimomab, gavilimomab, Golimumab, antithymocyte immunoglobulin, siplizumab, Alefacept, efalizumab, Pentasa, mesalazine, asacol, codeine phosphate, benorylate, fenbufen, naprosyn, diclofenac, etodolac, indomethacin, dasatinib (Sprycel®) nilotinib (Tasigna®), bosutinib (Bosulif®), Imatinib mesylate (Gleevec®) and ponatinib (Iclusig™) amifostine, dolasetron mesylate, dronabinol, epoetin-a, etidronate, filgrastim, fluconazole, goserelin acetate, gramicidin D, granisetron, leucovorin calcium, lidocaine, Mesna, ondansetron HC1, pilocarpine HC1, porfimer sodium, vatalanib, 1- dehydrotestosterone, allopurinol sodium, Betamethasone, sodium phosphate and betamethasone acetate, calcium leucovorin, conjugated estrogens, Dexrazoxane, Dibromomannitol, esterified estrogens, estradiol, estramustine phosphate sodium, ethinyl estradiol, flutamide, folinic acid, glucocorticoids, leuprolide acetate, levamisole HC1, medroxyprogesterone acetate, megestrol acetate, methyltestosterone, nilutamide, octreotide acetate, pamidronate disodium, procaine, propranolol, testolactone, tetracaine, toremifene citrate, and sargramostim.
In one embodiment the chemotherapeutic agent is an estrogen receptor ligands such as tamoxifen, raloxifene, fulvestrant, anordrin, bazedoxifene, broparestriol, chlorotrianisene, clomiphene citrate, cyclofenil, lasofoxifene, ormeloxifene, or toremifene; an androgen receptor ligand such as bicalutamide, enzalutamide, apalutamide, cyproterone acetate, chlormadinone acetate, spironolactone, canrenone, drospirenone, ketoconazole, topilutamide, abiraterone acetate, or cimetidine; an aromatase inhibitor such as letrozole, anastrozole, or exemestane; an anti-inflammatory such as prednisone; an oxidase inhibitor such as allopurinol; an anticancer antibody; an anticancer monoclonal antibody; an antibody against CD40 such as lucatumumab or dacetuzumab; an antibody against CD20 such as rituximab; an antibody that binds CD52 such as alemtuzumab; an antibody that binds integrin such as volociximab or natalizumab; an antibody against interleukin-6 receptor such as tocilizumab; an interleukin-2 memetic such as aldesleukin; an antibody that targets IGF1 like figitumumab; an antibody that targets DR4 such as mapatumumab; an antibody that targets TRAIL-R2 such as lexatumumab or dulanermin; a fusion protein such as atacicept; a B cell inhibitor such as atacicept; a proteasome inhibitor such as carfilzomib, bortezomib, or marizomib; a HSP90 inhibitor such as tanespimycin; a HDAC inhibitor such as vorinostat, belinostat or panobinostat; a MAPK ligand such as talmapimod; a PKC inhibitor such as enzastaurin; a HER2 receptor ligand such as trastuzumab, lapatinib, or pertuzumab; an EGFR inhibitor such as gefitimb, erlotinib, cetuximab, panitumumab, or vandetanib; a natural product such as romidepsin; a retinoid such as bexarotene, tretinoin, or alitretinoin; a receptor tyrosine kinase (RTK) inhibitor such as sunitinib, regorafenib, or pazopanib; or a VEGF inhibitor such as ziv-aflibercept, bevacizumab or dovitinib.
In one embodiment, the combinations of a CDK4/6 inhibitor, chemotherapeutic agent, and immune checkpoint inhibitor is further combined with the use of hematopoietic growth factors including, but not limited to, granulocyte colony stimulating factor (G-CSF, for example, sold as Neupogen® (filgrastim), Neulasta® (peg-filgrastim), or lenograstim), granulocyte-macrophage colony stimulating factor (GM-CSF, for example sold as molgramostim and sargramostim (Leukine®)), M-CSF (macrophage colony stimulating factor), Thrombopoietin (megakaryocyte growth development factor (MGDF), for example sold as Romiplostim® and Eltrombopag®) interleukin (IL)-12, interleukin-3, interleukin- 11 (adipogenesis inhibiting factor or oprelvekin), SCF (stem cell factor, steel factor, kit-ligand, or KL) and erythropoietin (EPO), and their derivatives (sold as for example epoetin-a as Darbepoetin, Epocept, Nanokine, Epofit, Epogen, Eprex, and Procrit; epoetin-b sold as for example NeoRecormon, Recormon and Micera), epoetin-delta (sold as for example Dynepo), epoetin- omega (sold as for example Epomax), epoetin zeta (sold as for example Silapo and Retacrit) as well as for example Epocept, Epotrust, Erypro Safe, Repoitin, Vintor, Epofit, Erykine, Wepox, Espogen, Relipoietin, Shanpoietin, Zyrop and EPIAO).
Additional active compounds contemplated herein, particularly in the treatment of abnormal tissue of the female reproductive system such as breast, ovarian, endometrial, or uterine cancer include a CDK9 inhibitor described herein in combination with an estrogen inhibitor including but not limited to a SERM (selective estrogen receptor modulator), a SERD (selective estrogen receptor degrader), a complete estrogen receptor degrader, or another form of partial or complete estrogen antagonist. Partial anti-estrogens include raloxifene and tamoxifen retain some estrogen-like effects. Complete anti-estrogens include fulvestrant. Non-limiting examples of anti-estrogen compounds are provided in WO 2014/19176 assigned to Astra Zeneca, WO2013/090921, WO 2014/203129, WO 2014/203132, and US2013/0178445 assigned to Olema Pharmaceuticals, W02017/100712, W02017/100715, WO2018/081168, and WO2018/148576 assigned to G1 Therapeutics, and U.S. Patent Nos. 9,078,871, 8,853,423, and 8,703,810, as well as US 2015/0005286, WO 2014/205136, and WO 2014/205138. Additional non-limiting examples of anti-estrogen compounds include: SERMS such as anordrin, arzoxifene, bazedoxifene, broparestriol, clomiphene citrate, cyclofenil, droloxifene, endoxifen, idoxifene, lasofoxifene, ormeloxifene, pipendoxifene, raloxifene, tamoxifen, toremifene, and fulvestrant; aromatase inhibitors such as aminoglutethimide, testolactone, anastrozole, exemestane, fadrozole, formestane, and letrozole; and antigonadotropins such as leuprorelin, cetrorelix, allylestrenol, chloromadinone acetate, delmadinone acetate, dydrogesterone, medroxyprogesterone acetate, megestrol acetate, nomegestrol acetate, norethisterone acetate, progesterone, and spironolactone. Additional non-limiting examples of anti-estrogen compounds include: SERDS such as fulvestrant, rintodestrant (G1T48), brilanestrant (GDC0810), elacestrant (RAD1901), etacstil (GW5638), GW7604, AZD9496, GDC-0927, GDC 9545 (RG6171), LSZ102, and SAR439859.
In certain embodiments the SERD compound of the Formula described in WO 2017/100712.
Figure imgf000203_0001
or a pharmaceutically acceptable salt or prodrug thereof
wherein:
m4 is 0, 1, 2, 3, or 4;
n4 is 0, 1, 2, 3, or 4;
XA is selected from -O-, -CH2-, -S-, -NH-, -NMe-, -CF2-, and C3cycloalkyl;
Ring B is phenyl, naphthyl, quinolinyl, 5- or 6- membered monocyclic heteroaryl or 7-, 8-, 9- or 10 membered bicyclic heterocycle;
Ring C is phenyl, thienyl, 5- or 6- membered monocyclic heteroaryl or 7-, 8-, 9- or 10- membered bicyclic heterocycle;
R41 is selected from hydroxyl, hydrogen, halogen, -O(C1-C6 alkyl), -OC(O)(C1-C6 alkyl), -OC(O)C6H5, -OC(O)0(C1-Ce alkyl), -OC(O)OC6H5 and -OSO2(C2- C6 alkyl);
R42 is selected from -CHCHCOOH, -NH(CO)COOH, -COOH, -C2-C6alkenylene-COOH and -C2-C6alkynylene-COOH;
R43 is independently selected at each occurrence from hydrogen, halogen, -CN, -NO2, -C1-C6alkyl and -C1-C6fluoroalkyl; and
R44 is independently selected at each occurrence from hydrogen, halogen, hydroxyl, -C1-C6alkyl, -C1-C6fluoroalkyl, -CN, -O(C1-C6alkyl), and -O(C1-C6fluoroalkyl). Non-limiting examples of SERDS for use in the present invention include:
Figure imgf000204_0001
Figure imgf000205_0001
or a pharmaceutically acceptable salt thereof.
In certain embodiments the SERD is:
Figure imgf000205_0002
or a pharmaceutically acceptable salt thereof.
In certain embodiments the SERD is:
Figure imgf000205_0003
or a pharmaceutically acceptable salt thereof. In certain embodiments the SERD is:
Figure imgf000206_0001
or a pharmaceutically acceptable salt thereof.
Additional chemotherapeutic agents contemplated herein, particularly in the treatment of abnormal tissue of the male reproductive system such as prostate or testicular cancer, include, but are not limited to, an androgen (such as testosterone) inhibitor including but not limited to a selective androgen receptor modulator, a selective androgen receptor degrader, a complete androgen receptor degrader, or another form of partial or complete androgen antagonist. In one embodiment, the prostate or testicular cancer is androgen-resistant. Non- limiting examples of anti-androgen compounds are provided in WO 2011/156518 and US Patent Nos. 8,455,534 and 8,299,112. Additional non-limiting examples of anti-androgen compounds include: chlormadinone acetate, spironolactone, canrenone, drospirenone, ketoconazole, topilutamide, abiraterone acetate, and cimetidine.
The chemotherapeutic agent may include a kinase inhibitor, including but not limited to a phosphoinositide 3-kinase (PI3K) inhibitor, a Bruton’s tyrosine kinase (BTK) inhibitor, or a spleen tyrosine kinase (Syk) inhibitor, or a combination thereof.
PI3k inhibitors are well known. Examples of PI3 kinase inhibitors include but are not limited to Wortmannin, demethoxyviridin, perifosine, idelalisib, pictilisib, Palomid 529, ZSTK474, PWT33597, CUDC-907, and AEZS-136, duvehsib, GS-9820, GDC-0032 (2-[4-[2- (2-Isopropyl-5-methyl-l,2,4-triazol-3-yl)-5,6-dihydroimidazo[l,2-d][l,4]benzoxazepin-9- yl]pyrazol-l-yl]-2-methylpropanamide), MLN-1117 ((2R)-l-Phenoxy-2-butanyl hydrogen (S)-methylphosphonate; or Methyl(oxo) {[(2R)-l-phenoxy-2-butanyl]oxy}phosphonium)), BYL-719 ((2S)-Nl-[4-Methyl-5-[2-(2,2,2-trifluoro-l,l-dimethylethyl)-4-pyridinyl]-2- thiazolyl]-l,2-pyrrolidinedicarboxamide), GSK2126458 (2,4-Difluoro-N-{2-(methyloxy)-5- [4-(4-pyridazinyl)-6-quinolinyl]-3-pyridinyl}benzenesulfonamide), TGX-221 ((±)-7-Methyl- 2-(morpholin-4-yl)-9-(l-phenylaminoethyl)-pyrido[l,2-a]-pyrimidin-4-one), GSK2636771 (2- Methyl-l-(2-methyl-3-(trifluoromethyl)benzyl)-6-morpholino-lH-benzo[d]imidazole-4- carboxylic acid dihydrochloride). KIN-193 ((R)-2-((l-(7-methyl-2-morpholino-4-oxo-4H- pyrido[l,2-a]pyrimidin-9-yl)ethyl)amino)benzoic acid), TGR-1202/RP5264, GS-9820 ((S)- 1- (4-((2-(2-aminopyrimidin-5-yl)-7-methyl-4-mohydroxypropan- 1 -one), GS-1101 (5-fluoro-3- phenyl-2-([S)]-l-[9H-purin-6-ylamino]-propyl)-3H-quinazolin-4-one), AMG-319, GSK- 2269557, SAR245409 (N-(4-(N-(3-((3,5-dimethoxyphenyl)amino)quinoxalin-2- yl)sulfamoyl)phenyl)-3-methoxy-4 methylbenzamide), BAY80-6946 (2-amino-N-(7- methoxy-8-(3-morpholinopropoxy)-2,3-dihydroimidazo[l,2-c]quinaz), AS 252424 (5-[l-[5-(4- Fluoro-2-hydroxy-phenyl)-furan-2-yl] -meth-(Z)-y li dene] -thiazolidine-2,4-di one), CZ 24832 (5-(2-amino-8-fluoro-[l,2,4]triazolo[l,5-a]pyridin-6-yl)-N-tert-butylpyridine-3-sulfonamide), buparlisib (5-[2,6-Di(4-morpholinyl)-4- pyrimidinyl]-4-(trifluoromethyl)-2-pyridinamine), GDC-0941 (2-(lH-Indazol-4-yl)-6-[[4-(methylsulfonyl)-l-piperazmyl]methyl]-4-(4- morpholinyl)thieno[3,2-d]pyrimidine), GDC-0980 ((S)-l-(4-((2-(2-aminopyrimidm-5-yl)-7- methyl-4-morpholinothieno[3,2-d]pyrimidin-6 yl)methyl)piperazin-l-yl)-2-hydroxypropan-l- one (also known as RG7422)), SF1126 ((8S,14S,17S)-14-(carboxymethyl)-8-(3- guani dinopropyl)- 17 -(hy droxy methyl)-3,6,9, 12,15 -pentaoxo- 1 -(4-(4-oxo-8-pheny 1-4H- chromen-2-y l)morpholino-4-ium)-2-oxa-7, 10,13,16-tetraazaoctadecan- 18-oate), PF -
05212384 (N-[4-[[4-(Dimethylamino)-l- piperidinyl]carbonyl]phenyl]-N'-[4-(4,6-di-4- morpholinyl-l,3,5-triazin-2-yl)phenyl]urea), LY3023414, BEZ235 (2-Methyl-2-{4-[3-methyl- 2-oxo-8-(quinolin-3-yl)-2,3-dihydro-lH-imidazo[4,5-c]quinolin-l-yl]phenyl}propanenitrile), XL-765 (N-(3-(N-(3-(3,5-dimethoxyphenylamino)quinoxalin-2-yl)sulfamoyl)phenyl)-3- methoxy-4-methylbenzamide), and GSK1059615 (5-[[4-(4-Pyridinyl)-6- quinolinyl]methylene]-2,4-thiazolidenedione), PX886 ([(3aR,6E,9S,9aR,10R,l laS)-6-
[[bis(prop-2-enyl)amino]methylidene]-5-hydroxy-9-(methoxymethyl)-9a,l la-dimethyl-1,4,7- trioxo-2,3,3a,9,10,ll-hexahydroindeno[4,5h]isochromen- 10-yl] acetate (also known as sonolisib)), and the structure described in W02014/071109 having the formula:
BTK inhibitors are well known. Examples of BTK inhibitors include ibrutinib (also known as PCI-32765)(Imbruvica™) (l-[(3R)-3-[4-amino-3-(4-phenoxy-phenyl)pyrazolo[3,4- d]pyrimidin-l-yl]piperidin-l-yl]prop-2-en-l-one), acalabrutinib (Calquence®), dianilinopyrimidine-based inhibitors such as AVL-101 and AVL-291/292 (N-(3-((5-fluoro-2- ((4-(2-methoxyethoxy)phenyl)amino)pyrimidin-4-yl)amino)phenyl)acrylamide) (Avila Therapeutics) (see US Patent Publication No 2011/0117073, incorporated herein in its entirety), dasatinib ([N-(2-chloro-6-methylphenyl)-2-(6-(4-(2-hydroxyethyl)piperazm-l-yl)-2- methylpyrimidin-4-ylamino)thiazole-5 -carboxamide], LFM-A13 (alpha-cyano-beta-hydroxy- beta-methyl-N-(2,5-ibromophenyl) propenamide), GDC-0834 ([R-N-(3-(6-(4-(l, 4-dimethyl- 3-oxopiperazin-2-yl)phenylamino)-4-methyl-5-oxo-4,5-dihydropyrazin-2-yl)-2- methylphenyl)-4,5,6,7-tetrahydrobenzo[b]thiophene-2-carboxamide], CGI-560 4-(tert-butyl)- N-(3-(8-(phenylamino)imidazo[l,2-a]pyrazin-6-yl)phenyl)benzamide, CGI-1746 (4-(tert- butyl)-N-(2-methyl-3-(4-methyl-6-((4-(morpholine-4-carbonyl)phenyl)amino)-5-oxo-4,5- dihydropyrazin-2-yl)phenyl)benzamide), CNX-774 (4-(4-((4-((3-acrylamidophenyl)amino)-5- fluoropyrimidin-2-yl)amino)phenoxy)-N-methylpicolinamide), CTA056 (7-benzyl-l-(3- (piperidin-l-yl)propyl)-2-(4-(pyridin-4-yl)phenyl)-lH-imidazo[4,5-g]quinoxalin-6(5H)-one), GDC-0834 ((R)-N-(3-(6-((4-(l,4-dimethyl-3-oxopiperazin-2-yl)phenyl)amino)-4-methyl-5- oxo-4, 5-dihydropyrazin-2-yl)-2-methylphenyl)-4, 5,6, 7-tetrahydrobenzo[b]thiophene-2- carboxamide), GDC-0837 ((R)-N-(3-(6-((4-(l ,4-dimethyl-3-oxopiperazin-2- yl)phenyl)amino)-4-methyl-5-oxo-4,5-dihydropyrazin-2-yl)-2-methylphenyl)-4, 5,6,7- tetrahydrobenzo[b]thiophene-2-carboxamide), HM-71224, ACP-196, ONO-4059 (Ono Pharmaceuticals), PRT062607 (4-((3-(2H-l,2,3-triazol-2-yl)phenyl)amino)-2-(((lR,2S)-2- aminocyclohexyl)amino)pyrimidme-5-carboxamide hydrochloride), QL-47 (1-(1- acryloylindolin-6-yl)-9-(l-methyl-lH-pyrazol-4-yl)benzo[h][l,6]naphthyridin-2(lH)-one), and RN486 (6-cyclopropyl-8-fluoro-2-(2-hydroxymethyl-3-{ 1-methyl-5-[5-(4-methyl- piperazin-l-yl)-pyri din-2 -ylamino]-6-oxo-l, 6-dihydro-pyri din-3-yl}-phenyl)-2H-isoquinolin- 1-one), BGB-3111, and other molecules capable of inhibiting BTK activity, for example those BTK inhibitors disclosed in Akinleye et ah, Journal of Hematology & Oncology, 2013, 6:59, the entirety of which is incorporated herein by reference.
Syk inhibitors are well known, and include, for example, Cerdulatinib (4- (cyclopropylamino)-2-((4-(4-(ethylsulfonyl)piperazin-l-yl)phenyl)amino)pyrimidine-5- carboxamide), entospletinib (6-(lH-indazol-6-yl)-N-(4-morpholinophenyl)imidazo[l,2- a]pyrazin-8-amine), fostamatinib ([6-({5-Fluoro-2-[(3,4,5-trimethoxyphenyl)amino]-4- pyrimidmyl}amino)-2,2-dimethyl-3-oxo-2,3-dihydro-4H-pyrido[3,2-b] [l,4]oxazin-4- yl I methyl dihydrogen phosphate), fostamatinib disodium salt (sodium (6-((5-fluoro-2-((3,4,5- trimethoxyphenyl)amino)pyrimidin-4-yl)amino)-2,2-dimethyl-3-oxo-2H-pyrido[3,2- b] [l,4]oxazin-4(3H)-yl)methyl phosphate), BAY 61-3606 (2-(7-(3,4-Dimethoxyphenyl)- imidazo[l,2-c]pyrimidin-5-ylamino)-nicotinamide HC1), RO9021 (6-[(lR,2S)-2-Amino- cyclohexylamino]-4-(5,6-dimethyl-pyridin-2-ylamino)-pyridazine-3-carboxylic acid amide), imatinib (Gleevec; 4-[(4-methylpiperazin-l-yl)methyl]-N-(4-methyl-3-{[4-(pyridm-3- yl)pyrimidin-2-yl]amino}phenyl)benzamide), staurosporine, GSK143 (2-(((3R,4R)-3- aminotetrahydro-2H-pyran-4-yl)amino)-4-(p-tolylamino)pyrimidine-5-carboxamide), PP2 (1- (tert-butyl)-3-(4-chlorophenyl)-lH-pyrazolo[3,4-d]pyrimidin-4-amine), PRT-060318 (2- (((lR,2S)-2-aminocyclohexyl)amino)-4-(m-tolylamino)pyrimidine-5-carboxamide), PRT- 062607 (4-((3-(2H-l,2,3-triazol-2-yl)phenyl)amino)-2-(((lR,2S)-2- aminocyclohexyl)amino)pyrimidme-5-carboxamide hydrochloride), R112 (3,3'-((5- fluoropyrimidine-2,4-diyl)bis(azanediyl))diphenol), R348 (3-Ethyl-4-methylpyridine), R406 (6-((5-fluoro-2-((3,4,5-trimethoxyphenyl)amino)pyrimidin-4-yl)amino)-2,2-dimethyl-2H- pyrido[3,2-b] [l,4]oxazin-3(4H)-one), YM193306(see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J. Med. Chem. 2012, 55, 3614-3643), 7- azaindole, piceatannol, ER-27319 (see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J. Med. Chem. 2012, 55, 3614-3643 incorporated in its entirety herein), Compound D (see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J. Med. Chem. 2012, 55, 3614-3643 incorporated in its entirety herein), PRT060318 (see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J. Med. Chem. 2012, 55, 3614-3643 incorporated in its entirety herein), luteolin (see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J. Med. Chem. 2012, 55, 3614-3643 incorporated in its entirety herein), apigenin (see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J. Med. Chem. 2012, 55, 3614-3643 incorporated in its entirety herein), quercetin (see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J. Med. Chem. 2012, 55, 3614-3643 incorporated in its entirety herein), fisetin (see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J. Med. Chem. 2012, 55, 3614-3643 incorporated in its entirety herein), myricetin (see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J. Med. Chem. 2012, 55, 3614-3643 incorporated in its entirety herein), morin (see Singh et al. Discovery and Development of Spleen Ty rosine Kinase (SYK) Inhibitors, J. Med. Chem. 2012, 55, 3614-3643 incorporated in its entirety herein).
The chemotherapeutic agent can also be a B-cell lymphoma 2 (Bcl-2) protein inhibitor. BCL-2 inhibitors are known in the art, and include, for example, ABT-199 (4-[4-[[2-(4- Chlorophenyl)-4,4-dimethylcyclohex-l-en-l-yl]methyl]piperazin-l-yl]-N-[[3-nitro-4- [[(tetrahydro-2H-pyran-4-yl)methyl]amino]phenyl]sulfonyl]-2-[(lH- pyrrolo[2,3-b]pyridin-5- yl)oxy]benzamide), ABT-737 (4-[4-[[2-(4-chlorophenyl)phenyl]methyl]piperazin-l-yl]-N-[4- [[(2R)-4-(dimethylamino)- 1 -phenylsulfanylbutan-2-yl] amino] -3- nitrophenyl |sulfonylbenzamide). ABT-263 ((R)-4-(4-((4'-chloro-4,4-dimethyl-3, 4,5,6- tetrahydro-[l, 1'-biphenyl]-2-yl)methyl)piperazin-l-yl)-N-((4-((4-morpholino-l-
(phenylthio)butan-2-yl)amino)-3((trifluoromethyl)sulfonyl)phenyl)sulfonyl)benzamide), GX15-070 (obatoclax mesylate, (2Z)-2-[(5Z)-5-[(3,5- dimethyl-lH-pyrrol-2-yl)methylidene]-
4-methoxypyrrol-2-ylidene]indole; methanesulfonic acid))), 2-methoxy-antimycin A3, YC137 (4-(4,9-dioxo-4,9-dihydronaphtho[2,3-d]thiazol-2-ylamino)-phenyl ester), pogosin, ethyl 2- amino-6-bromo-4-(l-cyano-2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate, Nilotinib-d3, TW-37 (N-[4-[[2-(l,l-Dimethylethyl)phenyl]sulfonyl]phenyl]-2,3,4-trihydroxy-5-[[2-(l- methylethyl)phenyl]methyl]benzamide), Apogossypolone (ApoG2), or G3139 (Oblimersen).
Additional chemotherapeutic agents for use in the methods contemplated herein include, but are not limited to, midazolam, MEK inhibitors, RAS inhibitors, ERK inhibitors, ALK inhibitors, HSP inhibitors (for example, HSP70 and HSP 90 inhibitors, or a combination thereof), RAF inhibitors, apoptotic compounds, topoisomerase inhibitors, AKT inhibitors, including but not limited to, MK-2206, GSK690693, Perifosine, (KRX-0401), GDC-0068, Triciribine, AZD5363, Honokiol, PF-04691502, and Miltefosine, or FLT-3 inhibitors, including but not limited to, P406, Dovitinib, Quizartinib (AC220), Amuvatinib (MP-470), Tandutinib (MLN518), ENMD-2076, and KW-2449, or combinations thereof. Examples of MEK inhibitors include but are not limited to trametinib /GSK1120212 (N-(3-{3-Cyclopropyl-
5-[(2-fluoro-4-iodophenyl)amino]-6,8-dimethyl-2,4,7-trioxo-3,4,6,7-tetrahydropyrido[4,3- d]pyrimidin-l(2H-yl}phenyl)acetamide), selumetinib (6-(4-bromo-2-chloroanilino)-7-fluoro- N-(2-hydroxyethoxy)-3-methylbenzimidazole-5-carboxamide),
pimasertib/AS703026/MSC1935369 ((S)-N-(2,3-dihydroxypropyl)-3-((2-fluoro-4- iodophenyl)amino)isonicotinamide), XL-518/GDC-0973 (l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-[(2S)-piperidin-2-yl]azetidin-3-ol),
refametinib/BAY869766/RDEA119 (N-(3,4-difluoro-2-(2-fluoro-4-iodophenylamino)-6- methoxyphenyl)-l-(2,3-dihydroxypropyl)cyclopropane-l -sulfonamide), PD-0325901 (N-
[(2R)-2,3-Dihydroxypropoxy]-3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]-benzamide), TAK733 ((R)-3-(2,3-Dihydroxypropyl)-6-fluoro-5-(2-fluoro-4-iodophenylamino)-8- methylpyrido[2,3d]pyrimidine-4,7(3H,8H)-dione), MEK162/ARRY438162 (5-[(4-Bromo-2- fluorophenyl)amino]-4-fluoro-N-(2-hydroxyethoxy)-l-methyl-lH-benzimidazole-6 carboxamide), R05126766 (3-[[3-Fluoro-2-(methylsulfamoylamino)-4-pyridyl]methyl]-4- methyl-7-pyrimidin-2-yloxychromen-2-one), WX-554, R04987655/CH4987655 (3,4- difluoro-2-((2-fluoro-4-iodophenyl)ammo)-N-(2-hydroxyethoxy)-5-((3-oxo-l,2-oxazinan-2 yl)methyl)benzamide), or AZD8330 (2-((2-fluoro-4-iodophenyl)amino)-N-(2- hydroxy ethoxy )-l,5-dimethyl-6-oxo-l,6-dihydropyridine-3-carboxamide). Examples of RAS inhibitors include but are not limited to Reolysm and siG12D LODER. Examples of ALK inhibitors include but are not limited to Crizotinib, AP26113, and LDK378. HSP inhibitors include but are not limited to Geldanamycin or 17-N-Allylamino-17-demethoxygeldanamycin (17AAG), and Radicicol.
Known ERK inhibitors include SCH772984 (Merck/Schering-Plough), VTX-lle (Vertex), DEL-22379, Ulixertimb (BVD-523, VRT752271), GDC-0994, FR 180204, XMD8- 92, and ERK5-IN-1.
Raf inhibitors are well known, and include, for example, Vemurafmib (N-[3-[[5-(4- Chlorophenyl)-lH-pyrrolo[2,3-b]pyridin-3-yl]carbonyl]-2,4-difluorophenyl]-l- propanesulfonamide), sorafenib tosylate (4-[4-[[4-chloro-3-
(trifluoromethyl)phenyl]carbamoylamino]phenoxy]-N-methylpyridine-2-carboxamide;4- methylbenzenesulfonate), AZ628 (3-(2-cyanopropan-2-yl)-N-(4-methyl-3-(3-methyl-4-oxo- 3,4-dihydroquinazolin-6-ylamino)phenyl)benzamide), NVP-BHG712 (4-methyl-3-(l-methyl- 6-(pyridin-3-yl)-lH-pyrazolo[3,4-d]pyrimidin-4-ylamino)-N-(3-
(trifluoromethyl)phenyl)benzamide), RAF -265 (l-methyl-5-[2-[5-(trifluoromethyl)-lH- imidazol-2-yl]pyridin-4-yl]oxy-N-[4-(trifluoromethyl)phenyl]benzimidazol-2-amme), 2- Bromoaldisine (2-Bromo-6,7-dihydro-lH,5H-pyrrolo[2,3-c]azepine-4,8-dione), Raf Kinase Inhibitor IV (2-chloro-5-(2-phenyl-5-(pyridin-4-yl)-lH-imidazol-4-yl)phenol), and Sorafenib N-Oxide (4-[4-[[[[4-Chloro-3(trifluoroMethyl)phenyl]aMino]carbonyl]aMino]phenoxy]-N- Methyl-2pyridinecarboxaMide 1 -Oxide).
Known topoisomerase I inhibitors useful in the present invention include (S)-IO- [(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-lH-pyrano[3',4':6,7]indolizino[l,2- b] quinoline-3, 14(4H,12H)-di one monohydrochloride (topotecan), (S)-4-ethyl-4-hydroxy-lH- pyrano[3',4':6,7]indolizmo[l,2-b]quinoline-3,14-(4H,12H)-dione (camptothecin), (1S,9S)-1- Amino-9-ethyl-5-fluoro- 1 ,2,3,9, 12, 15 -hexahydro-9-hydroxy -4-methyl- 1 OH, 13H- benzo(de)pyrano(3',4': 6,7)indolizino(l ,2-b)quinoline- 10, 13-dione (exatecan), (7 -(4- methylpiperazinomethylene)-10,l l-ethylenedioxy-20(S)-camptothecin (lurtotecan), or (S)- 4,l l-diethyl-3,4,12,14-tetrahydro-4-hydroxy-3,14-dioxolH-pyrano[3’,4’:6,7]-indolizino[l,2- b]quinolin-9-yl-[l,4’bipiperidine]-r-carboxylate (irinotecan), (R)-5-ethyl-9,10-difluoro-5- hydroxy-4,5-dihydrooxepino[3',4':6,7]indolizino[l,2-b]quinoline-3,15(lH,13H)-dione (diflomotecan), (4S)-11 -((E)-((l , 1 -Dimethylethoxy)imino)methyl)-4-ethyl-4-hy droxy- 1,12- dihydro-14H-pyrano(3',4':6,7)indolizino(l,2-b)quinoline-3,14(4H)-dione (gimatecan), (S)-8- ethyl-8-hydroxy-15-((4-methylpiperazin-l-yl)methyl)-l l,14-dihydro-2H-[l,4]dioxino[2,3- g]pyrano[3',4':6,7]indolizino[l,2-b]quinoline-9,12(3H,8H)-dione (lurtotecan), (4S)-4-Ethyl-4- hydroxy-l l-[2-[(l-methylethyl)amino]ethyl]-lH-pyrano[3,4:6,7]indolizino[l,2-b]quinoline- 3,14(4H,12H)-dione (belotecan), 6-((l,3-dihydroxypropan-2-yl)amino)-2,10-dihydroxy-12- ((2R,3R,4S,5S,6R)-3,4,5-trihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl)-12,13- dihydro-5H-indolo[2,3-a]pyrrolo[3,4-c]carbazole-5,7(6H)-dione (edotecarin), 8,9-dimethoxy- 5-(2-N,N-dimethylaminoethyl)-2,3-methylenedioxy-5H-dibenzo(c,h)(1,6)naphthyridin-6-one (topovale), benzo[6,7]indolizino[l,2-b]quinolin-l l(13H)-one (rosetacin), (S)-4-ethyl-4- hydroxy- 11-(2-(trimethylsilyl)ethyl)-l H-pyrano|3'.4':6.7 |indolizino[l,2-b]quinoline- 3,14(4H,12H)-dione (cositecan), tetrakis{(4S)-9-[([l,4'-bipiperidinyl]- 1'-carbonyl)oxy]-4,l 1- diethyl-3,14-dioxo-3,4,12,14- tetrahydro-lH-pyrano[3',4':6,7]indolizino[l,2-b]quinolin-4-yl} N,N',N",N"'- {methanetetrayltetrakis[methylenepoly(oxyethylene)oxy(l- oxoethylene)]}tetraglycinate tetrahydrochloride (etirinotecan pegol), 10-hydroxy - camptothecin (HOCPT), 9-nitrocamptothecin (rubitecan), SN38 (7-ethyl- 10- hydroxy camptothecin), and 10-hydroxy-9-nitrocamptothecin (CPT109), (R)-9-chloro-5-ethyl- 5-hydroxy-10-methyl-12-((4-methylpiperidin-l-yl)methyl)-4,5- dihydrooxepino[3',4':6,7]indolizino[l,2-b]quinoline-3,15(lH,13H)-dione (elmotecan).
In one embodiment, the chemotherapeutic agent is not an aromatase inhibitor. In one embodiment, the chemotherapeutic agent is not an estrogen or androgen receptor agonist or antagonist.
Growth Factors
In one embodiment, the combination of a CDK4/6 inhibitor, chemotherapeutic agent, and checkpoint inhibitor is further combined with the use of hematopoietic growth factors including, but not limited to, granulocyte colony stimulating factor (G-CSF, for example, sold as Neupogen (filgrastim), Neulasta (peg-filgrastim), or lenograstim), granulocyte-macrophage colony stimulating factor (GM-CSF, for example sold as molgramostim and sargramostim (Leukine)), M-CSF (macrophage colony stimulating factor), Thrombopoietin (megakaryocyte growth development factor (MGDF), for example sold as Romiplostim and Eltrombopag) interleukin (IL)-12, interleukin-3, interleukin-11 (adipogenesis inhibiting factor or oprelvekin), SCF (stem cell factor, steel factor, kit-ligand, or KL) and erythropoietin (EPO), and their derivatives (sold as for example epoetin-a as Darbepoetin, Epocept, Nanokine, Epofit, Epogen, Eprex, and Procrit; epoetin-b sold as for example NeoRecormon, Recormon and Micera), epoetin-delta (sold as for example Dynepo), epoetin- omega (sold as for example Epomax), epoetin zeta (sold as for example Silapo and Retacrit) as well as for example Epocept, Epotrust, Erypro Safe, Repoitin, Vintor, Epofit, Erykine, Wepox, Espogen, Relipoietin, Shanpoietin, Zyrop and EPIAO). CDK4/6 Inhibitors
The present invention also provides advantageous methods to treat a patient with a selective CDK4/6 inhibitor resistant cancer, which include administering an effective amount of a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, or a pharmaceutically acceptable composition, salt, isotopic analog, or prodrug thereof. In one aspect a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, is used to treat a patient with a cancer intrinsically resistant to selective CDK4/6 inhibition. In one aspect, a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, is used to treat a patient with a cancer that has acquired resistance to one or more selective CDK4/6 inhibitors. In one aspect, a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, is administered in combination with a selective CDK4/6 inhibitor to a patient with a selective CDK4/6 inhibition responsive cancer in order to extend the therapeutic effectiveness of the selective CDK4/6 inhibitor. In one aspect, a compound of Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, or XXIV, is administered in combination with a selective CDK4/6 inhibitor to a patient with a selective CDK4/6 inhibition responsive cancer, wherein the patient is selective CDK4/6 inhibitor naive. Selective CDK4/6 inhibitors for use in combination with a compound of the present invention include, but are not limited to palbociclib, abemaciclib, ribociclib, trilaciclib, SHR6390, and lerociclib.
In one embodiment, the selective CDK4/6 inhibitor is Palbociclib:
Figure imgf000213_0001
, or a pharmaceutically acceptable salt therof. In one embodiment, the selective CDK4/6 inhibitor is abemaciclib:
Figure imgf000214_0001
, or a pharmaceutically acceptable salt thereof.
In one embodiment, the selective CDK4/6 inhibitor is ribociclib:
Figure imgf000214_0002
or a pharmaceutically acceptable salt thereof.
In one embodiment, the selective CDK4/6 inhibitor is lerociclib:
Figure imgf000214_0003
or a pharmaceutically acceptable salt thereof.
In one embodiment, the selective CDK4/6 inhibitor is trilaciclib:
Figure imgf000214_0004
, or a pharmaceutically acceptable salt thereof. In one embodiment, the selective CDK4/6 inhibitor is SHR 6390.
In one embodiment, the selective CDK4/6 inhibitor is selected from an inhibitor described in, for example, U.S. Patent Nos. 8,822,683; 8,598,197; 8,598,186; 8,691,830; 8,829,102; 8,822,683; 9,102,682; 9,499,564; 9,481,591; and 9,260,442, filed by Tavares and Strum and assigned to G1 Therapeutics describe a class of N-(heteroaryl)-pyrrolo[3,2- d]pyrimidin-2-amine cyclin dependent kinase inhibitors including those of the formula (with variables as defined therein):
Figure imgf000215_0001
In one embodiment, the selective CDK4/6 inhibitor is selected from an inhibitor described in, for example, U.S. Patent Nos. 9,464,092, 9,487,530, and 9,527,857 which are also assigned to G1 Therapeutics describe the use of the above pyrimidine-based agents in the treatment of cancer.
In one embodiment, the selective CDK4/6 inhibitor is selected from an inhibitor described in, for example, WO 2013/148748 (U.S.S.N. 61/617,657) titled“Lactam Kinase Inhibitors”, WO 2013/163239 (U.S.S.N. 61/638,491) titled“Synthesis of Lactams” and WO 2015/061407 filed by Tavares and also assigned to G1 Therapeutics describes the synthesis of N-(heteroaryl)-pyrrolo[3,2-d]pyrimidin-2-amines and their use as lactam kinase inhibitors.
In one embodiment, the selective CDK4/6 inhibitor is selected from an inhibitor described in, for example. WO 2014/144326 filed by Strum et al. and assigned to G1 Therapeutics describes compounds and methods for protection of normal cells during chemotherapy using pyrimidine-based CDK4/6 inhibitors. WO 2014/144596 filed by Strum et al. and assigned to G1 Therapeutics describes compounds and methods for protection of hematopoietic stem and progenitor cells against ionizing radiation using pyrimidine-based CDK4/6 inhibitors. WO 2014/144847 filed by Strum et al. and assigned to G1 Therapeutics describes HSPC-sparing treatments of abnormal cellular proliferation using pyrimidine-based CDK4/6 inhibitors. WO 2014/144740 filed by Strum et al. and assigned to G1 Therapeutics describes highly active anti-neoplastic and anti-proliferative pyrimidine-based CDK 4/6 inhibitors. WO 2015/161285 filed by Strum et al. and assigned to G1 Therapeutics describes tricyclic pyrimidine-based CDK inhibitors for use in radioprotection. WO 2015/161287 filed by Strum et al. and assigned to G1 Therapeutics describes tricyclic pyrimidine-based CDK inhibitors for the protection of cells during chemotherapy. WO 2015/161283 filed by Strum et al. and assigned to G1 Therapeutics describes tricyclic pyrimidine-based CDK inhibitors for use in HSPC-sparing treatments of RB-positive abnormal cellular proliferation. WO 2015/161288 filed by Strum et al. and assigned to G1 Therapeutics describes tricyclic pyrimidine-based CDK inhibitors for use as anti-neoplastic and anti-proliferative agents. WO 2016/040858 filed by Strum et al. and assigned to G1 Therapeutics describes the use of combinations of pyrimidine-based CDK4/6 inhibitors with other anti-neoplastic agents. WO 2016/040848 filed by Strum et al. and assigned to G1 Therapeutics describes compounds and methods for treating certain Rb-negative cancers with CDK4/6 inhibitors and topoisomerase inhibitors.
VIII. EXAMPLES
Example 1
General Methods:
Compounds of the present invention with stereocenters are drawn racemic for convenience. One skilled in the art will recognize that pure enantiomers can be prepared by methods known in the art. Examples of methods to obtain optically active materials include at least the following.
i) physical separation of crystals— a technique whereby macroscopic crystals of the individual enantiomers are manually separated. This technique can be used if crystals of the separate enantiomers exist, i.e., the material is a conglomerate, and the crystals are visually distinct;
ii) simultaneous crystallization— a technique whereby the individual enantiomers are separately crystallized from a solution of the racemate, possible only if the latter is a conglomerate in the solid state;
iii) enzymatic resolutions— a technique whereby partial or complete separation of a racemate by virtue of differing rates of reaction for the enantiomers with an enzyme;
iv) enzymatic asymmetric synthesis— a synthetic technique whereby at least one step of the synthesis uses an enzymatic reaction to obtain an enantiomerically pure or enriched synthetic precursor of the desired enantiomer;
v) chemical asymmetric synthesis— a synthetic technique whereby the desired enantiomer is synthesized from an achiral precursor under conditions that produce asymmetry (i.e., chirality) in the product, which may be achieved using chiral catalysts or chiral auxiliaries; vi) diastereomer separations— a technique whereby a racemic compound is reacted with an enantiomerically pure reagent (the chiral auxiliary) that converts the individual enantiomers to diastereomers. The resulting diastereomers are then separated by chromatography or crystallization by virtue of their now more distinct structural differences and the chiral auxiliary later removed to obtain the desired enantiomer;
vii) first- and second-order asymmetric transformations— a technique whereby diastereomers from the racemate equilibrate to yield a preponderance in solution of the diastereomer from the desired enantiomer or where preferential crystallization of the diastereomer from the desired enantiomer perturbs the equilibrium such that eventually in principle all the material is converted to the crystalline diastereomer from the desired enantiomer. The desired enantiomer is then released from the diastereomer;
viii) kinetic resolutions— this technique refers to the achievement of partial or complete resolution of a racemate (or of a further resolution of a partially resolved compound) by virtue of unequal reaction rates of the enantiomers with a chiral, non-racemic reagent or catalyst under kinetic conditions;
ix) enantiospecific synthesis from non-racemic precursors— a synthetic technique whereby the desired enantiomer is obtained from non-chiral starting materials and where the stereochemical integrity is not or is only minimally compromised over the course of the synthesis;
x) chiral liquid chromatography— a technique whereby the enantiomers of a racemate are separated in a liquid mobile phase by virtue of their differing interactions with a stationary phase (including via chiral HPLC). The stationary phase can be made of chiral material or the mobile phase can contain an additional chiral material to provoke the differing interactions; xi) chiral gas chromatography— a technique whereby the racemate is volatilized and enantiomers are separated by virtue of their differing interactions in the gaseous mobile phase with a column containing a fixed non-racemic chiral adsorbent phase;
xii) extraction with chiral solvents— a technique whereby the enantiomers are separated by virtue of preferential dissolution of one enantiomer into a particular chiral solvent;
xiii) transport across chiral membranes— a technique whereby a racemate is placed in contact with a thin membrane barrier. The barrier typically separates two miscible fluids, one containing the racemate, and a driving force such as concentration or pressure differential causes preferential transport across the membrane barrier. Separation occurs as a result of the non-racemic chiral nature of the membrane that allows only one enantiomer of the racemate to pass through. Chiral chromatography, including simulated moving bed chromatography, is used in one embodiment. A wide variety of chiral stationary phases are commercially available.
Representative Synthesis
General
Unless otherwise noted, all reagents were used without further purification. 1H NMR spectra were obtained in DMSO-d6 or CD30D at room temperature on a Bruker 300 MHz instrument. When more than one conformer was detected, the chemical shifts for the most abundant one is reported. Chemical shifts of 1H NMR spectra were recorded in parts per million (ppm) on the d scale from an internal standard of residual solvent. Splitting patterns are designed as s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; brs, broad. LC-MS conditions were described below:
General LC/MS method:
Column: Agilent Zorbax XDB C18 4.6x50 mm, 3.5mm
Mobile phase: Solvent A: 0.1 % in formic acid water
Solvent B: MeOH
Flow rate: 1.0 mL/min
Run time/Gradient: 2 min (20%-90% B), then 3 min @90% B
Temperature: 30 °C.
General HPLC Method:
Column: Agilent SB-C18 4.6x 150 mm, 3.5mm
Mobile phase: Solvent A: 0.02% in TFA water
Solvent B: MeOH
Flow rate: 1.0 mL/min
Run time/Gradient: 0.5 min @10% B, 9.5 min gradient @10%-90% B,
then 10 min @90% B,
Temperature: 30 °C.
General preparative HPLC Method: Column: Phenomenex Luna 5u 100A. 21.2x250mm, 5mm
Mobile phase: Solvent A: Water
Solvent B: MeOH
Flow rate: 10 mL/'min
Run time/Gradient: 1 min @20% B, 30 min gradient @20%-80% B,
then 10 min @90% B
Temperature: Ambient
The following abbreviations are used below: PE = petroleum ether, EA = ethyl acetate, DMSO = dimethyl sulfoxide, DMP = Dess-Martin reagent, DMF = N, N-dimethylacetamide, MeOH = methanol, MTBE = Methyl tert-butyl ether, DCM = dichloromethane, TEA = triethylamine, DIPEA = Dhsopropylethylamine, TFA = trifluoroacetic acid, TLC = thin layer chromatography, B2Pin2 = Bis(pinacolato)diboron. Scheme 1. Synthesis of Compound 1
Figure imgf000219_0001
Compound 1-1 (100 g) was converted to 1-2 using LAH (2 eq) in THF at 0 °C. The reaction was heated to 70 °C and stirred for 4 hours. After workup, 80.4 g of crude 1-2 was obtained and the structure was confirmed by HNMR. Compound 1-2 (9 g, 1.1 eq) was converted to 1-3 using 1-a (1 eq), NaHCO3, DMAc at 70 °C overnight. After work-up, the crude product was slurried with EtOAc:PE=l :5 to afford 5.8 g of pure 1-3 (HPLC purity: 98.2%). Compound 1- 3 (125 g) was converted to 1-4 using Dess-Martin in DCM at 0 °C. The reaction was allowed to warm to room temperature and stir for 2.5 hours. Purification afford crude 1-4 (125.5 g). Compound 1-4 (118 g) was converted to 1-5 using 1-b (1.1 eq), PdCl2(PPh3)2 (0.05 eq), Cul (0.12 eq), and Et3N (2 eq) in THF at room temperature overnight. After purification, 150 g of crude 1-5 was obtained. Compound 1-5 (1 g) was converted to 1-6 using TBAF (3 eq) in THF at 60 °C for 2 hours at which point the starting material was consumed. After workup, 990 mg of crude 1-6 was obtained. Compound 1-6 (130 g) was converted to 1-7 using TFA in DCM at room temperature for 1 hour. After purification, 21.3 g of crude 1-7 was obtained. Compound 1-7 (1.5 g) was converted to Compound 1 using 1-c (3 eq)/Cs2CO3(1.5 eq)/ Pd(OAc)2(0.5 eq)/X-phos(0.6 eq)/dioxane (150 mL)/85 °C /3 h.
Scheme 1. Synthesis of Compound 1
Figure imgf000220_0001
Compound 1
Compound 1-7 (60 mg) is converted to Compound 1 using 1-c (3.0 eq), CsC03 (1.5 eq), Pd(OAc)2 (0.15 eq), amd X-phos (0.2 eq) in dioxane at 80 °C overnight followed by cleavage of the PMB groups with TFA, 50 °C for 2 hours.
Scheme 2. Synthesis of Compound 2
Figure imgf000221_0001
Compound 2
Compound 1-7 (1 eq) was converted to Compound 2 using 2-1 (9 grams, 1.2 eq), AcOK (3 eq), Pd(OAc)2 (0.1 eq), amd X-phos (0.4 eq) in DMF at 80 °C overnight followed by cleavage of the PMB groups with TFA, 50 °C for 3 hours.
Scheme 4. Synthesis of Compound 3
Figure imgf000221_0002
Compound 3-1 (430 mg) was converted to 3-2 using 1-methylpiperazine, HOB, and EDC1 in DMF. The reaction was stirred overnight at room temperature. After purification, 350 mg of 3- 2 was obtained. Compound 3-2 (350 mg) was converted to 3-3 using Pd/C/H2 in MeOH at room temperature for 5 hours. After workup, 308 mg of 3-3 was obtained. Compound 3-3 (47.8 mg) was converted to Compound 3 using 3-a, AcOK, Pd(OAc)¾ and X-phos in DMF at 80 °C overnight. After purification, 16.0 mg of Compound 3 was obtained and the structure was confirmed by HNMR, HPLC and LC-MS. Scheme 5. Synthesis of Compound 11
Figure imgf000222_0001
Scheme 6. Synthesis of Compound 18
Figure imgf000223_0001
Compound 18-3 (70 mg) was converted to Compound 18 using DCM and TFA at room temperature for 4 hours. After purification, 7.1 mg of Compound 18 was obtained and the structure was confirmed via HNMR, HPLC and LC-MS.
Scheme 7. Synthesis of Compound 19
Figure imgf000223_0002
Figure imgf000224_0001
Compound 19-2 (614 mg, 1.0 eq) was converted to 19-3 using BnNH2 (3.0 eq) in DMF at 110 °C overnight. After purification, 510 mg of 19-3 was obtained. Compound 19-3 (350 mg, 1.0 eq) was converted to 19-4 using H2SO4 (8 ml) at room temperature. The reaction was stirred for 5 hours. After purification, 95 mg of 19-4 was obtained. Compound 19-4 (42 mg, 1.0 eq) was converted to Compound 19 using 19-b, Pd(OAc)2, X-phos, and AcOK in DMF at 85 °C overnight. After purification, 2.5 mg of Compound 19 was obtained and the structure was confirmed by HNMR, HPLC and LC-MS. Scheme 8. Synthesis of Compound 21
Figure imgf000224_0002
Compound 21-1 (100 mg) was converted to 21-2 using PMB2NH and TEA in DCM at 0 °C for 0.5 hours. To the above reaction mixture, was added NH3 in MeOH at 0 °C for 0.5 hours. After purification, 75 mg of 21-3 was obtained. Compound 21-3 (75 mg) is converted to 21- 4 using 21-a, Pd(OAc)2, X-phos, and AcOH in DMF at 100 °C for 12 hours.
Scheme 9. Synthesis of Compound 22
Figure imgf000225_0001
Compound 22-1 (100 mg) was converted to 22-2 using 22-a in DCM. The reaction stirred at
0 °C for 1 hour. After workup, crude 22-2 was directly used for the next step. Crude 22-2 was converted to 22-3 using NH3 in EtOH at room temperature for 1 hour at which point the product was observed by LC-MS. After purification, 40 mg of 22-3 was obtained and coupled to 22-b to afford Compound 22 using Pd(OAc)2, x-phos, and AcOK in DMF at 100 °C for 6 hours. After purification, 4.2 mg of Compound 22 was obtained and the structure was confirmed by HNMR, HPLC and LC-MS.
Scheme 10. Synthesis of Compound 23
Figure imgf000225_0002
Compound 23-2 (50 mg) was coupled to 23-b to afford Compound 23 using Pd(OAc)2, x- phos, and AcOK in DMF at 80 °C overnight at which point the product was observed by LC- MS. After purification by prep TLC and F1PLC, 2.4 mg of Compound 23 was obtained and the structure was confirmed by LC-MS, HPLC and HNMR. Scheme 11. Synthesis of Compound 24
Figure imgf000226_0001
Compound 24-1 (1 g) was converted to 24-2 using TsOH and NBS in DCM at room temperature for 30 minutes. After purification, 1.1 g of 24-2 was obtained. Compound 24-2 (300 mg) was converted to 24-3 using NaN3 in MeOH at room temperature for 1 hour. After purification, 120 mg of 24-3 was obtained.
Scheme 12. Synthesis of Compound 25
Figure imgf000227_0001
Figure imgf000228_0001
Compound 25-1 (7 g) was converted to 25-2 using AC2O in pyridine at 90 °C overnight. After workup, 5 g of the intermediate 25 1 was obtained. Intermediate 25 1 (1 g) was converted to 25-2 using AC2O, DMAP, and 4-Methylmorpholine in toluene. The reaction stirred at 60 °C overnight. After purification, 210 mg of 25-2 was obtained and the structure was confirmed by HNMR. Compound 25-1 (20 g) was converted to 25 1 using AC2O in MeOH and the reaction was refluxed for 3 hours. After workup, 28 g of 25 1 was obtained and the structure was confirmed by HNMR.
Figure imgf000228_0002
Compound 37-1 (1.2 g) was coupled to 37-a to afford 37-2 using EtiN in DCM at 0° C for 1.5 hours. Work-up afforded 2.0 g of 37-2. Compound 37-2 (2 g) was converted to 37-3 using Pd/C/H2 in MeOH and ethyl acetate. The reaction stirred for 2 hours. After workup, 1.7 g of 37-3 was obtained. Compound 37-3 (74.4 mg) was coupled to 37-b to afford 37-4 using AcOK, Pd(OAc)2, and X-phos in DMF. The reaction stirred at 80 °C overnight. After purification, 25 mg of 37-4 was obtained. Compound 37-4 (25 mg) was converted to Compound 37 using TFA in DCM. The reactions stirred at room temperature for 1 hour at which point the product was observed by LC-MS. After purification, 5.6 mg of Compound 37 was obtained and confirmed by HNMR, HPLC and LC-MS.
Scheme 14. Synthesis of Compound 39 and Compound 45
Figure imgf000229_0001
Compound 39-1 (6 g) was coupled to 39-a to afford 39-2 using pyridine and DMAP in DCM. The reaction stirred for three hours at room temperature for 3 hours. After workup, 12.2 g of crude 39-2 was obtained. Compound 39-2 (1 g) was converted to 39-3 using Hz/Pd/C in MeOH and stirring at room temperature overnight. After purification, 850 mg of 39-3 was obtained. Compound 39-3 (220 mg) was coupled to 39-a to afford 39-4 using Pd(OAc)2, x-phos, KOAc in DMF at 85 °C for 2 hours at which point the product was observed by LC-MS. After purification, 89 mg of 38-4 was obtained. Compound 39-4 (30 mg) was converted to Compound 39 using TFA in DCM. The reaction stirred for 2 hours. After purification, 15.4 mg of Compound 39 was obtained and the structure was confirmed via HNMR, HPLC and LC-MS. Compound 39 is converted to Compound 45 using MsCl in TEA and DCM. The reaction is stirred at room temperature for 3 hours.
Scheme 15. Alternative Synthesis of Compound 45
Figure imgf000230_0001
Compound 45-1 (2 g) was converted to 45-2 using SO2CI2 in 1 ,2-dichloroethane and DMF. The reaction was heated to 85 °C and stirred for 3 hours. Work-up afford compound 45-2 (2.2 g). Compound 45-2 (2.2 g) was coupled to 45-4 to afford 45-3 using DMAP and pyridine in DCM at room temperature for 3 hours. After purification, 1.3 g of 45-3 was obtained. Compound 45-3 (1.3 g) was converted to 45-4 using TFA at 50 °C for 30 minutes. After workup, 0.8 g of 45-4 was obtained. Compound 45-4 (400 mg) was coupled to 45-b to afford 45-5 in DCM and TEA. The reaction stirred overnight at room temperature. After purification, 475 mg of 45-5 was obtained. H-NMR was OK.Compound 45-5 (300 mg) was converted to 45-6 using H2/Pd/C in MeOH and THF. The reaction stirred overnight. After purification, 197 mg of 45-6 was obtained. Compound 45-6 (50 mg) was coupled to 45-c to afford Compound 45 using Pd(OAc)2, x-phos, and CS2CO3 in DMF. The reaction was heated to 85 °C and allowed to stir for 3 hours. After purification, 12.0 mg of Compound 45 was obtained. Scheme 16. Synthesis of Compound 46
Figure imgf000231_0003
Compound 46-1 (500 mg) was converted to 46-2 using 46-a in iPrOH. The reaction stirred for 12 hours at room temperature. After purification, 700 mg of 46-2 was obtained.
Compound 46-2 (50 mg) was coupled to 46-b converted to Compound 46 using Pd(OAc)2, x-phos, and CS2CO3 in DMF at 80 °C for 4 hours. After purification, 3.4 mg of Compound 46 was obtained and the structure was confirmed by HNMR, HPLC and LCMS.
Scheme 17. Synthesis of compound 47
Figure imgf000231_0001
Compound 47-1 (50 mg) was coupled to 47-a to afford Compound 47 using Pd(OAc)2, Cs2CO3, xphos in DMF at 85 °C for 5 hours. After purification, 2.5 mg of Compound 47 was obtained and the structure was confirmed by HNMR, HPLC and LCMS. Scheme 18. Synthesis of Compound 48
Figure imgf000231_0002
Compound 48-1 was coupled to 48-a to afford Compound 48 using Pd(OAc)2, x-phos, CS2CO3 in DMF at 80 °C for 4 hours at which point the product was observed by LC-MS. After purification, 3.6 mg of Compound 48 was obtained and the structure was confirmed by HNMR, HPLC and LCMS.
Scheme 19. Synthesis of 2'-((4-(4-methylpiperazine-l-carbonyl)phenyl)amino)-6'H- spiro[cyclohexane-l,9'-pyrazino[l',2':l,5]pyrrolo[2,3-d]pyrimidin]-6'-one
(COMPOUND 52)
Figure imgf000232_0001
Compound 49-1 (50 mg) was converted to 49-2 using m-CPBA (1.5 eq) in CHCb. The reaction was stirred at 5 ~ 10 °C for 20 minutes at which point the desired product was observed by TLC. After purification, 40 mg of 49-2 was obtained. Compound 49-2 (40 mg) was converted to 49-3 using PCC in CHCb at 0 °C. The reaction was allowed to warm to room temperature over the course of 4 hours at which point a new spot was observed by TLC. After purification, 8 mg of an oxidized product was obtained. An alternative synthesis for Compound 49-3 is shown below:
Figure imgf000233_0001
Compound 49-1 is converted to 49-3 using H5IO6 (1 eq), AC2O (1 eq), and CrO3. An alternative synthesis for Compound 49-3 is shown below:
Figure imgf000233_0002
Compound 49-8 (10 g) was converted to 49-9 using LAH in THF at room temperature overnight. After purification, 8.1 g of 49-9 was obtained. Compound 49-9 is coupled to 49-a (3 g) using NaHCO3 in DMAc at 80 °C overnight. Compound 49-4 (0.5 g) is converted to 49-5 using NH3Ή2O at -10 °C. The reaction is allowed to warm to room temperature and stir for 3 hours. The desired product was observed by GC-MS. Scheme 20. Synthesis of 4-((5'-hydroxy-6'H-spiro [cyclohexane-1, 9'- pyrazino[l',2':l,5]pyrrolo[2,3-d]pyrimidin]-2'-yl)amino)benzenesulfonamide
(COMPOUND 53)
Figure imgf000234_0001
Compound 50-1 (85 mg) was converted to 50-2 using NBS (1.2 eq) in THF. The reaction was stirred at room temperature for 1 hour. After purification, 110 mg of 50-2 was obtained. Compound 50-2 (110 mg) was converted to COMPOUND 53 using LiOH (3 eq), Cu(acac)2 (0.05 eq), and BHMPO (0.05 eq) in DMSO/H2O. The reaction was stirred at 90 °C for 24 hours. The starting material was consumed. After purification by prep HPLC, 0.5 mg of COMPOUND 53 was obtained.
An alternative synthesis for COMPOUND 53 is shown below:
Figure imgf000234_0002
Figure imgf000235_0001
Compound 50-1 (300 mg) was coupled to 50-a using AcOK, Pd(OAc)2, and X-phos in DMF at 80 °C overnight. After purification, 350 mg of 50-3 was obtained. Compound 50-3 (350 mg) was converted to 50-4 using NBS in THF at room temperature for 1 hour. After purification, 248 mg of 50-4 was obtained. Compound 50-4 (100 mg) was converted to 50-5 using KOH in DMSO/H2O at 110 °C for 5 hours. After purification, 24 mg of 50-5 was obtained. Compound 50-5 (24 mg) was converted to COMPOUND 53 using TFA in DCM at room temperature overnight. Scheme 21. Synthesis of 4-((5'-fluoro-6'H-spiro[cyclohexane-l,9'- pyrazino [1 ',2' : 1,5] pyrrolo [2,3-d] pyrimidin] -2'-yl)amino)benzenesulfonamide
(COMPOUND 54)
Figure imgf000235_0002
Compound 51-1 (500 mg) was converted to 51-2 using NBS in MeCN at room temperature for 6 hours. After purification, 200 mg of 51-2 was obtained. Alternatively, Compound 51-1 is converted to 51-2 using SelectFluor.
Scheme 22. Synthesis of 4-((6'-hydroxy-6'H-spiro [cyclohexane-1, 9'- pyrazino [1 ',2' : 1,5] pyrrolo [2,3-d] pyrimidin] -2'-yl)amino)benzenesulfonamide
(COMPOUND 56)
Figure imgf000236_0001
Compound 53-1 (70 mg) was converted to COMPOUND 56 using m-CPBA (0.5 eq) in DCM and THF at 0 °C for 3 hours. The desired product was observed by LC-MS.
Scheme 23. Synthesis of 4-((6'-(difluoromethyl)-6'H-spiro [cyclohexane-1, 9'- pyrazino [1 ',2' : 1,5] pyrrolo [2,3-d] pyrimidin] -2'-yl)amino)benzenesulfonamide
(COMPOUND 57)
Figure imgf000236_0002
Figure imgf000237_0001
Compound 54-1’ (2 g) was converted to 54-2’ using MsCl and Et3N in DCM at -78 °C for 2 hours. After workup, 3 g of 54-2’ was obtained. Compound 54-2’ (1 g) was converted to 54-3’ using NaN3 in DMF at 65 °C for 1 hour. After workup, 700 mg of 54-3’ was obtained. Compound 54-3’ (700 mg) was converted to 54-3 using PPh3 in THF and H2O at 50 °C for 2 hours. After w orkup. 500 mg of 54-3 was obtained.
Alternatively, 54-1 (4.5 g) was coupled to 54-a using tetrabutylammonium iodide and 18-crown-6 in DMF at room temperature overnight to afford 54-2. Compound 54-2 (100 mg) was converted to 54-3 using N2H4 H2O in MeOH. The reaction stirred for 2 hours. After purification, 40 mg of 54-3 was obtained. Crude 54-3 (2.5 g) was converted to 54-4 using (Boc)2O and DMAP in THF. The reaction stirred overnight to afford 934 mg of 54-4 after purification.
An alternative synthesis of Compound 54-5 is shown below:
Figure imgf000238_0001
Compound 54-13 (3 g) was converted to 54-14 using DMF and n-BuLi in Et2O at -78 °C for 5 hours. After purification, 3.2 g of 54-14 was obtained. Compound 54-14 (500 mg) was coupled to 54-b using Ti(OAc)4 in THF. The reaction stirred at room temperature for 2 hours and after purification, 430 mg of 54-15 was obtained.
An alternative synthesis of Compound 54-11 is shown below:
Figure imgf000238_0002
Figure imgf000239_0001
Compound 54-13 (3 g) was coupled to 54-c using n-BuLi in THF at -78 °C overnight to afford 3.7 g of 54-19 after purification. Compound 54-19 (200 mg) was converted to 54-20 using TEA and TiCl4 in Et2O/hex at - 40 °C for 2 hours. After workup by filtration and washing with ethyl ether, 240 mg of 54-20 was obtained. Compound 54-20 (20 mg) was converted to 54-21 using DIBAL-H in toluene at - 30 °C for 2 hours. The desired product was observed by LC-MS and HNMR and 50 mg was converted to 54-22 using TB AF in TF1F at room temperature for 1 hour. After purification, 40 mg of 54-22 was obtained.
An alternative synthesis of Compound 54-11 is shown below:
Figure imgf000239_0002
Compound 54-27 (10 g) was converted to 54-28 using LAH in THF. The reaction was refluxed for 2 hours. After purification, 9.1 g of 54-28 was obtained that was coupled to 54-d using NaHCO3 in DMAc at 70 °C overnight. After purification, 13 g of 54-29 was obtained. Compound 54-9 (7 g) was converted to 54-30 using PCC in DCM at room temperature overnight. After purification, 3.8 g of 54-30 was obtained. Compound 54-22 (100 mg) was coupled to 54-30 using Pd( PPh) 3Cl2. Cul, and TEA in THF at room temperature overnight. After purification, 90 mg of 54-11 was obtained. An alternative synthesis of Compound 54-11 is shown below:
Figure imgf000240_0001
54-28 54-11
Compound 54-22 (100 mg) was coupled to 54-29 using Pd(PPh)3Cl2, Cul, and TEA in THF at room temperature overnight. After purification, 90 mg of 54-33 was obtained. 20 mg of 54-33 was converted to 54-25 using t-BuOK in THF at room temperature for 1 hour. The product was observed by LC-MS.
An alternative synthesis of COMPOUND 57 is shown below:
Figure imgf000240_0002
Figure imgf000241_0002
Compound 54-29 (5 g) was coupled to 54-13 using TEA, Cul, and PdCl2(PPh3)2 in THF at room temperature overnight. After purification, 5.3 g of 54-34 was obtained that was converted to 54-35 using TBAF in THF at 60 °C for 3 hours. After purification, 2.4 g of 54-35 was obtained. Compound 54-35 (2.4 g) was coupled to 54-e using Pd(OAc)2, x-phos, and AcOK in DMF at 50 °C overnight. After purification, 3.7 g of 54-36 was obtained. Compound 54-36 (100 mg) was coupled to 54-f (5 eq.) using t-BuLi (4 eq) in THF at - 78 °C/for 3 hours. The product was confirmed by HNMR.
The following modified approach from the intermediate 54-36 is shown below:
Figure imgf000241_0001
Figure imgf000242_0002
Compound 54-36 (500 mg) was converted to 54-50 using Dess-Martin in THF at 0 °C for 4 hours. The desired product was observed by LC-MS. After purification, 220 mg of 54-40 was obtained that was converted to 54-41 using CH(OMe)3 and cat. p-TsOH in MeOH at room temperature overnight. The temperature was then increased to 65 °C for 7 hours at which point a new spot was observed by TLC. After purification, 310 mg of crude 54-51 was obtained.
The following modified approach from the intermediate 54-37 is shown below:
Figure imgf000242_0001
Scheme 24. Synthesis of 4-((6'-fluoro-8'H-spiro[cyclohexane-l,9'- pyrazino [1 ',2' : 1,5] pyrrolo [2,3-d] pyrimidin] -2'-yl)amino)benzenesulfonamide
(COMPOUND 58)
Figure imgf000243_0001
Compound 55-1 (500 mg) was coupled to 55-a (5 eq) using LDA (5 eq) in THF at - 78
°C for 3 hours. After purification, 260 mg of crude 55-2 was obtained.
An alternative approach to the synthesis of COMPOUND 58 is shown below:
Figure imgf000243_0002
55-5 COMPOUND 58
Compoung 55-3 (20 mg) was converted to 55-4 using TosCl and NaH in DMF. The reaction stirred at room temperature for 30 minutes. The product was observed by LC-MS and after workup, 23 mg of 55-4 was obtained. Compound 55-4 (5 mg) is converted to 55-5 using KF in DMF at 80 °C for 1 hour. The reaction temperature is increased to 100 °C and allowed to stir overnight. Scheme 25. Synthesis of (lr,4r)-4-((4-(l,4-diazaspiro[5.5]undec-4-en-l-yl)pyrimidin-2- yl)amino)cyclohexane- 1-sulfonamide (COMPOUND 59)
Figure imgf000244_0001
Compound 56-1 (5 g) was converted to 56-2 using Br2 in Et2O and 1,4-dioxane at 0 °C for 1.5 hours. After work-up, 8.4 g of crude 56-2 was obtained and the structure was confirmed by H-NMR. Compound 56-2 (950 mg) was coupled to 56-a in toluene at room temperature for 3 hours. The product was observed by LC-MS. After purification, 477 mg of 56-3 was obtained. Compound 56-3 (1.2 g) was coupled to 56-b using 2,4,6-trimethylpyridine in DMAc at 90 °C overnight. After purification, 180 mg of crude 56-4 was obtained.
Scheme 26. Synthesis of (lr,4r)-4-((4-(3-oxo-l,4-diazaspiro[5.5]undec-4-en-l- yl)pyrimidin-2-yl)amino)cyclohexane- 1-sulfonamide (COMPOUND 60)
Figure imgf000244_0002
Figure imgf000245_0001
Compound 57-1 (5 g) was converted to 57-2 using LAH in THF. The reaction was stirred at 0 °C and allowed to warm to 40 °C over 5 hours. After purification, 2 g of 57-2 was obtained. Compound 57-2 (100 mg) is coupled to 57-a using K2CO3 in DMF at room temperature overnight.
An alternative to the synthesis of Compound 57-7 is shown below:
Figure imgf000245_0002
Compound 57-2 (200 mg) is converted to 57-13 using 57-a, HCOOH, and NaBH3CN in MeOH/THF at room temperature for 5 hours.
An alternative to the synthesis of Compound 57-8 is shown below:
Figure imgf000245_0003
Compound 57-10 (250 mg) was converted to 57-11 using 2,4,6-trimethylpyridine in DMAc. The reaction was subjected to microwave conditions of 80 °C for 20 minutes. The product was observed by LC-MS. After purification, 60 mg of 57-11 was obtained.
An alternative to the synthesis of Compound 57-7 is shown below:
Figure imgf000246_0001
57-10 57-17 57-6 57-7
Compound 57-10 (300 mg) was converted to 57-17 using TEA/THF/(Boc)20 at 60 °C overnight. After purification, 450 mg of 57-17 was obtained. Compound 57-17 is converted to 57-6 using NaCIO in MeOH at room temperature overnight and the resulting intermediate is subjected to BBr3 in DCM at room temperature for 1 hour to afford Compound 57-7.
Scheme 27. Synthesis of (lr,4r)-4-((4-(3-oxo-l,4-diazaspiro[5.5]undecan-l-yl)pyrimidin- 2-yl)amino)cyclohexane- 1-sulfonamide (COMPOUND 61)
Figure imgf000247_0001
Compound 58-1 (500 mg) was converted to 58-2 using CbzCl and NaHCCb in FkO- THF at 0 °C. The reaction was allowed to warm to room temperature over 4 hours. After purification. 1 g of 58-2 was obtained. 58-2 (1 g) was converted to 58-2’ using MsCl and Et3N in DCM at 0 °C for 2 hours. After workup, 1.5 g of 58-2’ was obtained. Compound 58-2’ (1.5 g) was converted to 58-5 and 58-5’ using 58-c (3.3 eq) in DMF at 65 °C overnight. After purification, 58 mg of one isomer and 240 mg of the other isomer (58-5 and 58-5’) were obtained. The yield of 58-5 from 58-2 through intermediates 58-2’ was 24%.
Compound 58-5 (150 mg) was converted to 58-6 using NCS in HC1 (2 M) and MeCN at 0 °C for 1 hour. The product was observed by LC-MS and after workup, 160 mg of crude 58-6 was obtained that was used directly m the next step. (Alternatively, 58-5 can be converted to 58-6 using Ck in DCM-H2O at 0°C for 2 hours.) Crude 58-6 (160 mg) was converted to 58- 7 using PMB2NH and Et3N in DCM at room temperature overnight. The product was observed by LC-MS.
Alternatively, Compound 58-2 (100 mg) was coupled to 58-a using DEAD and PPh3 in THF at 60 °C for 2 hours. After purification, 70 mg of 58-3 was obtained. Compound 58-3 (70 mg) was converted to 58-4 using K2CO3 (2 eq) in MeOH at room temperature for 3 hours. After purification, HNMR and LC-MS confirmed the product. Compound 58-4 (200 mg) was converted to 58-4’ using MsCl and TEA in DCM at 0 °C. The reaction was allowed to warm to room temperature for 3 hours. After workup, 250 mg of crude 58-4’ was obtained that was coupled to 58-c in DMF at 65 °C for 2 hours to afford 58-5 and 58-5’. NOESY analysis could not distinguish the two isomers. The yield of 58-5 from 58-2 through intermediates 58-3, 58- 4, and 58-4’ was 5.5%.
Compound 58-9 (300 mg) was converted to 58-10 using 58-b (1 eq) and 2,4,6- trimethylpyridine (1 eq) in DMAc at 120 °C for 48 hours. After purification, 65 mg of 58-10 was obtained.
An alternative to the synthesis of COMPOUND 61 that utilizes a PMBNH2 group is shown below:
Figure imgf000248_0001
Compound 57-6 (20 mg) was converted to 57-12 using PMBNFh and 2, 4, 6-collidine in DCM. The reaction was stirred at room temperature for 1 hour at which point the desired product was observed by LC-MS. Alternatively, 57-6 is converted to 57-12 using PMBNFh and Et3N in DCM at room temperature for 1 hour.
Scheme 28. Synthesis of 3-((6'H-spiro[cyclohexane-l,9'-pyrazino[r,2':l,5]pyrrolo[2,3- d]pyrimidin]-2'-yl)amino)cyclopentane- 1-sulfonamide (COMPOUND 62)
Figure imgf000249_0001
Compound 59-1 (1 g) was converted to 59-2 using phthalimide in MeOH at room temperature overnight. After purification, 300 mg of pure 59-2 was obtained. Compound 59-2 (50 mg) was converted to 59-3 using NaBFD (0.6 eq) in THF at room temperature for 2 hours. The desired product was observed by LC-MS and after purification, 30 mg of 59-3 was obtained. The structure was confirmed by 1HNMR. Compound 59-3 (200 mg) was converted to 59-4 using MsCl and TEA in DCM. The reaction was stirred at 0 °C and allowed to warm to room temperature overnight. The desired product was observed by LC-MS. Compound 59- 4 (500 mg) was converted to 59-5 using 59-a in DMF. The reaction was stirred at 65 °C for 5 hours. The desired product was observed by LC-MS and after purification, 390 mg of 59-5 was obtained. Compound 59-5 (100 mg) was converted to 59-5’ using NCS in the presence of HC1 (2 M) in MeCN. The reaction was conducted at 0 °C and allowed to warm to room temperature. After two hours, a new spot was observed by LC-MS and the starting material was consumed. Following workup, crude 59-5’ was carried forward.
An alternative to the synthesis of COMPOUND 62 that utilizes a PMBNLh group is shown below:
Figure imgf000250_0001
Compound 59-5 (300 mg) was converted to 59-5’ using NCS in HC1 (2 M) and MeCN at 0 °C. The reaction was allowed to warm to room temperature and stir for 2 hours at which point the starting material 59-5 was consumed and a new spot was observed on TLC. After workup, the crude 59-5’ was converted to 59-9 directly using PMBNEL and TEA in EA at room temperature overnight. The desired product was observed by LC-MS. After purification, 75 mg of 59-9 was obtained. Compound 59-9 (70 mg) was converted to 59-10 using N2H4Ή2O in MeOH at 0 °C. The reaction was allowed to warm to room temperature and stir overnight at which point a new spot was observed by TLC and the desired product was observed by LC- MS. After purification, 40 mg of 59-10 was obtained. 59-10 was then converted to 59-11 by reacting it with 59-b in NMP at 140 °C for 2 hours followed by purification.59-11 was converted to Compound 62 by reacting it with TFA in DCM for 5 hours. The crude material was then purified by preparative TLC twice to afford 6 mg of Compound 62 (cis/trans isomers). Scheme 29. Synthesis of 4-((6'H-spiro[cyclohexane-l,9'-pyrazino[r,2':l,5]pyrrolo[2,3- d]pyrimidin]-2'-yl)amino)cycloheptane- 1-sulfonamide (COMPOUND 63)
Figure imgf000251_0001
60-12 COMPOUND 83
Compound 60-1 (100 mg) was converted to 60-2 using TMSCHN2 and n-BuLi in Et20 at -78 °C for 1 hour. The desired product was observed by LC-MS. Following purification by silica column, LC-MS and H-NMR confirmed that Compound 60-2 was converted to Compound 60-3. Compound 60-3 (6.1 g) was converted to 60-4 using NaBFU in THF and MeOH. The reaction was conducted at °C and allowed to warm to room temperature for 1 hour. After purification, 5.27 g of 60-4 was obtained. Compound 60-4 (3 g) was converted to 60-5 using MsCl in the presence of TEA in DCM. The reaction stirred at 0 °C and was allowed to warm to room temperature overnight. The desired product was observed by LC-MS and after purification. 3.05 g of 60-5 was obtained. Compound 60-5 (500 mg) converted to 60-6 using 60-a in DMF. The reaction stirred at 65 °C for 4 hours. The desired product was observed by LC-MS. After purification. 377 mg of 60-6 was obtained. Compound 60-6 (370 mg) was converted to 60-7 using TFA in DCM at room temperature overnight. The desired product was observed by LC-MS. After purification, 180 mg of 60-7 was obtained. 270 mg of crude 60-8 was obtained by reacting 180 mg of 60-7 with Cbz-Cl in the presence of NaHCCh in THF and H2O at room temperature overnight. Compound 60-8 (270 mg) was converted to 60-9 using NCS in MeCN and aqueous HC1 at 0 °C. The reaction was allowed to warm to room temperature and stir for 2 hours. The desired product was observed by LC-MS. After workup, 230 mg of the crude 60-9 was carried forward in the next step. 60-9 was converted to 60-10 by stirring with NFL in water and THF at room temperature overnight. 60-10 was then converted to 60-11 by deprotection with Pd/C and hydrogen gas in MeOH overnight. 60-11 was then converted to Compound 63 by reaction with 60-b in NMP with sodium bicarbonate at 140 °C for 3 hours.
An alternative to the synthesis of COMPOUND 63 that utilizes a PMBNH2 group is shown below:
Figure imgf000252_0001
Scheme 30. Synthesis of 4-((6'-hydroxy-6’H-spiro [cyclohexane-1, 9’- pyrazino [1 ',2' : 1,5] pyrrolo [2,3-d] pyrimidin] -2'-yl)amino)benzenesulfonamide
(COMPOUND 64)
Figure imgf000253_0001
Compound 61-1 (70 mg) was converted to COMPOUND 64 using m-CPBA (0.5 eq) in DCM and THF at 0 °C for 3 hours at which point a new spot was observed on TLC and the desired product was observed by LC-MS. After purification, 2 mg of COMPOUND 64 was obtained which required further purification. Scheme 31. Synthesis of (lr,4r)-4-((4-(3-oxo-l,4-diazaspiro[5.5]undecan-l- yl)pyrimidin- 2-yl)amino)cyclohexane- 1-sulfonamide (COMPOUND 61)
Figure imgf000253_0002
Step 1: Synthesis of benzyl ((lr,4r)-4-hydroxycyclohexyl)carbamate (61-1)
To a solution of (lr,4r)-4-aminocyclohexan-l-ol (14.2 g, 123.29 mmol) in THF (150 mL) was added H2O (120 mL), NaHCCb (20.7 g, 246.40 mmol) and Cbz-Cl (25.3 g, 148.31 mmol) at 0 °C. After stirred at room temperature for 2h, the reaction mixture was quenched with H2O (50 mL) and extracted with DCM:MeOH = 5: 1 (3 x 100 mL). The organic layer was separated and washed with brine, dried over Na2S04, filtered and concentrated in vacuo. The resulting residue was purified by a silica gel column chromatography to give benzyl ((lr,4r)-4- hydroxy cyclohexyl) carbamate 72-1 (27.5 g, 110.30 mmol). LC-MS (ESI+): m/z 272 [M+Na]+.
Step 2: Synthesis of (lr,4r)-4-(((benzyloxy)carbonyl)amino)cyclohexyl methanesulfonate (61-2)
To a solution of benzyl ((lr,4r)-4-hydroxycyclohexyl)carbamate 61-1 (26.6 g, 106.69 mmol) in DCM (300 mL) was added EtiN (16.2 g, 160.09 mmol) and MsCl (14.6 g, 127.46 mmol) at 0 °C. After the addition, the reaction was stirred at 0 °C for lh. After the reaction was completed as indicated by TLC analysis, the reaction mixture was neutralized with saturated aqueous Na2CCb (100 mL) and extracted with DCM (100 mL x 3). The organic layer was separated and washed with brine, dried over NaiSOi. filtered and concentrated in vacuum to provide (lr,4r)-4-(((benzyloxy)carbonyl)amino)cyclohexyl methanesulfonate 61-2 (34.8 g, 106.29 mmol) as a yellow solid. LC-MS (ESI+): m/z 350 [M+Na]+.
Step 3: Synthesis of S-((lr,4r)-4-(((benzyloxy)carbonyl)amino)cyclohexyl) ethanethioate (61-3)
Under N2 atmosphere, to a solution of (lr,4r)-4-(((benzyloxy)carbonyl)amino) cyclohexyl methanesulfonate 61-2 (20.7 g, 63.23 mmol) in DMSO (300 mL) was added potassium ethanethioate (10.8 g, 94.57 mmol). After stirred at 50 °C overnight, the reaction mixture was cooled to room temperature, quenched with water (100 mL) and extracted with EtOAc (200 mL x 3). The combined organic phase was washed with brine (10 mL x 2), dried over Na2SOr, filtered and concentrated in vacuo. The resulting residue was purified by a silica gel column chromatography to give S-((lr,4r)-4-(((benzyloxy)carbonyl)amino) cyclohexyl)ethanethioate (11.2 g, 36.43 mmol) and S-((ls,4s)-4- (((benzyloxy)carbonyl)amino)cyclohexyl) ethanethioate (5.6 g of impure cis-isomer) 61-3. LC-MS (ESI+): m/z 330 [M+Na]+. Step 4: Synthesis of benzyl ((lr,4r)-4-(chlorosulfonyl)cyclohexyl)carbamate (61-4)
To a solution of NCS (11.7 g, 87.62 mmol) in MeCN (100 mL) was added aq. HC1 (2M, 90 mL). After the addition, the reaction was stirred at room temperature for 15 min. A solution of S-((lr,4r)-4-(((benzyloxy)carbonyl)amino) cyclohexyl)ethanethioate 61-3 (9 g, 29.28 mmol) in MeCN (100 mL) was added at 0 °C. The reaction was stirred at 0 °C for 2h, quenched with water (100 mL) and extracted with DCM (200 mL x 3). The combined organic phase was washed with brine (100 mL x 2), dried over Na2S04. The organic phase was concentrated under vacuum to provide crude benzyl ((lr,4r)-4-(chlorosulfonyl) cyclohexyl)carbamate 61-4 (10 g). LC-MS (ESI+): m/z 354 [M + Na]+.
Step 5: Synthesis of benzyl ((lr,4r)-4-(N-(4-methoxybenzyl)sulfamoyl)cyclohexyl) carbamate (61-5)
To a solution of crude benzyl ((lr,4r)-4-(chlorosulfonyl)cyclohexyl)carbamate 61-4 (10 g) in DCM (100 mL) was added Et3N (6.1 g, 60.28 mmol) and (4-methoxyphenyl)methanamine (4.95 g, 36.08 mmol). After stirred at room temperature for 30min, the reaction was quenched with water (100 mL) and extracted with DCM (100 mL x 3). The combined organic phase was washed with brine (100 mL x 2), dried over Na2S04, filtered and concentrated in vacuo. The resulting residue was purified by a silica gel column chromatography to give benzyl ((lr,4r)- 4-(N-(4-methoxybenzyl)sulfamoyl)cyclohexyl)carbamate 61-5 (9 g, 20.81 mmol). LC-MS (ESI+): m/z 455 [M+Na]+.
Step 6: Synthesis of (lr,4r)-4-amino-N-(4-methoxybenzyl)cyclohexane-l- sulfonamide (61-6)
To a solution of benzyl ((lr,4r)-4-(N-(4-methoxybenzyl)sulfamoyl)cyclohexyl) carbamate 61-5 (750 mg, 1.73 mmol) in MeOH (10 mL) and THF (10 mL) was added Pd/C (100 mg). The reaction solution was degassed and backfilled with hydrogen three times. After stirred under atmospheric pressure of hydrogen overnight, the reaction mixture was filtered. The filtrate was concentrated under vacuum to provide (lr,4r)-4-amino-N-(4- methoxybenzyl)cyclohexane -1 -sulfonamide 61-6 (360 mg, 1.21 mmol) as a white solid. LC- MS (ESI+): m/z 299 [M+H]+.
Step 7: Synthesis of l-(2-chloropyrimidin-4-yl)-l,4-diazaspiro[5.5]undecan-3-one (61-8)
To a solution of l,4-diazaspiro[5.5]undecan-3-one 61-7 (338.2 mg, 2.01 mmol) in DMAc (8 mL) w as added 2,4-dichloropyrimidine (300 mg, 2.01 mmol) and 2,4,6- trimethylpyridine (255.8 mg, 2.11 mmol). After stirred at 120 °C for 48h, the reaction was cooled to room temperature, quenched with water (10 mL) and extracted with DCM:MeOH = 5: 1 (10 mL x 3). The combined organic phase was washed with brine (10 mL x 2), dried over Na2S04, filtered and concentrated in vacuo. The resulting residue was purified by a silica gel column chromatography to give l-(2-chloropyrimidin-4-yl)-l,4-diazaspiro[5.5] undecan-3- one 61-8 (67 mg, 0.24 mmol). LC-MS (ESI+): m/z 281 [M+H]+.
Step 8: Synthesis of (lr,4r)-N-(4-methoxybenzyl)-4-((4-(3-oxo-l,4-diazaspiro
[5.5] undecan-l-yl)pyrimidin-2-yl)amino)cyclohexane- 1-sulfonamide (61-9)
Under N2 atmosphere, to a solution of l-(2-chloropyrimidin-4-yl)-l,4-diazaspiro
[5.5]undecan-3-one 61-8 (60 mg, 0.21 mmol) in DMAc (6 mL) was added (lr,4r)-4-amino-N-
(4- methoxybenzyl)cyclohexane-l -sulfonamide 61-6 (70 mg, 0.23 mmol) and NaHCCh (72 mg, 0.86 mmol). After stirred at 130 °C for 48h, the reaction mixture was cooled to room temperature, quenched with water (10 mL) and extracted with DCM:MeOH = 5: 1 (10 mL x 3). The combined organic phase was washed with brine (10 mL x 2), dried over Na2S04, filtered and concentrated in vacuo. The resulting residue was purified by a silica gel column chromatography to give (lr,4r)-N-(4-methoxybenzyl)-4-((4-(3-oxo-l,4- diazaspiro[5.5]undecan-l-yl) py rimi din-2 -yl)amino)cyclohexane-l -sulfonamide 61-9 (23 mg, 0.04 mmol). LC-MS (ESI+): m/z 543 [M+H]+.
Step 9: Synthesis of (lr,4r)-4-((4-(3-oxo-l,4-diazaspiro[5.5]undecan-l-yl) pyrimidin-2- yl)amino)cyclohexane- 1-sulfonamide (COMPOUND 61)
To a solution of (lr,4r)-N-(4-methoxybenzyl)-4-((4-(3-oxo-l,4-diazaspiro[5.5] undecan-l-yl)pyrimidin-2-yl)amino)cyclohexane-l-sulfonamide 61-9 (23 mg, 0.04 mmol) in DCM (2 mL) was added TFA (0.4 mL). After stirred at room temperature overnight, the reaction mixture was neutralized with saturated aqueous Na2C03 (5 mL) and extracted with DCM:MeOH = 5: 1 (15 mL x 3). The organic layer was separated and washed with brine, dried over Na2S04, filtered and concentrated in vacuo. The resulting residue was purified by preparative TLC to give (lr,4r)-4-((4-(3-oxo-l,4-diazaspiro[5.5]undecan-l-yl) pyrimidin-2- yl)amino) cyclohexane- 1 -sulfonamide COMPOUND 61 (3.6 mg, 0.008 mmol). LC-MS (ESI+): m/z 423 [M+H]+; ' HNMR (300MHZ, CD3OD) d 7.68 (d, J = 6Hz, 1H), 5.82 (d, J = 6Hz, 1H), 4.28 (s, 2H), 4.03 - 3.81 (m, 1H), 3.52 (s, 2H), 3.21 - 3.13 (m, 2H), 3.03 - 2.88 (m, 1H), 2.39 - 2.27 (m, 2H), 2.22 - 2.15 (m, 2H), 1.78 - 1.67 (m, 4H), 1.60 - 1.34 (m, 8H). Scheme 32. Synthesis of (lr,4r)-4-((6'H-spiro[cyclohexane-l,9'- pyrazino [1 ',2' : 1,5] pyrrolo [2,3-d] pyrimidin] -2'-yl)amino)cyclohexane- 1-sulfonamide (COMPOUND 71) and (lr,4r)-4-amino-N-(6'H-spiro [cyclohexane-1, 9'- pyrazino[l',2':l,5]pyrrolo[2,3-d]pyrimidin]-2'-yl)cyclohexane-l-sulfonamide
(COMPOUND 72)
Figure imgf000257_0001
Under N2 atmosphere, to a solution of 2'-chloro-6'H-spiro[cyclohexane-l,9'- pyrazino[r,2': l,5]pyrrolo[2,3-d]pyrimidine] (200 mg, 0.73 mmol) in DMAc (5 mL) was added (lr,4r)-4-aminocyclohexane-l-sulfonamide (130 mg, 0.73 mmol) and NaHC03 (367 mg, 4.37 mmol). After stirred at 125 °C overnight, the reaction mixture was cooled to room temperature, quenched with water (10 mL) and extracted with EtOAc:MeOH = 5: 1 (10 mL x 3). The combined organic phase was washed with brine (10 mL x 2), dried over Na2S04, filtered and concentrated in vacuo. The resulting residue was purified by preparative TLC to give (lr,4r)- 4-amino-N-(6'H-spiro[cyclohexane-l,9'-pyrazino[r,2': l,5]pyrrolo[2,3-d]pyrimidin]-2'- yl)cyclohexane-l -sulfonamide (1.9 mg, 0.005 mmol). LC-MS (ESI+): m/z 417 [M+H]+; ' H NMR (300 MHz, CD30D) </ 8.58 (s, 1H), 8.31 (s, 1H), 6.79 (s, 1H), 4.12 - 4.05 (m, 1H), 4.02 (s, 2H), 3.09 - 3 01 (m, 1H), 2.95 - 2.85 (m, 2H), 2.40 - 2.29 (m, 1H), 2.25 - 2.17 (m, 2H), 2.13 - 2.01 (m, 3H), 1.99 - 1.89 (m, 2H), 1.85 - 1.77 (m, 6H), 1.63 - 1.57 (m, 2H) COMPOUND 72 and (lr,4r)-4-((6'H-spiro[cyclohexane- l,9'-pyrazino[T,2': l,5]pyrrolo[2,3-d]pyrimidin]-2'- yl)amino)cyclohexane-l -sulfonamide (2.1 mg, 0.005 mmol). LC-MS (ESI+): m/z 417 [M+H]+; ¾ NMR (300 MHz, CD3OD/CDCI3) d 8.96 (s, 1H), 8.56 (s, 1H), 6.77 (s, 1H), 4.02 (s, 2H), 3.80 - 3.70 (m, 1H), 3.01 - 2.86 (m, 3H), 2.39 - 2.30 (m, 4H), 1.88 - 1.77 (m, 6H), 1.70 - 1.59 (m, 4H), 1.50 - 1.40 (m, 2H) COMPOUND 71. Scheme 33. Synthesis of 4-((6'H-spiro[cyclohexane-l,9'-pyrazino [l',2':l,5] pyrrolo[2,3- d]pyrimidin]-2'-yl)amino)-3-hydroxybenzenesulfonamide (Compound 73)
Figure imgf000258_0001
COMPOUND 73
Under N2 atmosphere, to a solution of 2'-chloro-6'H-spiro[cyclohexane -l,9'-pyrazino [G,2': 1,5] pyrrolo[2.3-d]pyrimidine] (200 mg, 0.73 mmol) in DMF (4 mL) was added 4-amino-
3-hydroxybenzenesulfonamide (152 mg, 0.81 mmol), Pd(OAc)2 (18 mg, 0.08 mmol), X-Phos (140 mg, 0.29 mmol) and AcOK (216 mg, 2.20 mmol). The reaction mixture was stirred at 80 °C for 3 h. The reaction mixture was cooled to rt, fdtered and the filtrate was concentrated in vacuo. The resulting residue was purified by preparative TLC to provide 4-((6'H- spiro [cy cl ohexane-l,9'-pyrazino [T,2': l,5] pyrrolo[2,3-d]pyrimidin] -2'-yl)amino)-3- hydroxybenzenesulfonamide COMPOUND 73 (2 mg, 0.005 mmol). LC-MS (ESI+): m/z 427 [M+H]+; ¾ NMR (300 MHz, DMSO -d6) d 10.79 (s, 1H), 8.89 (s, 1H), 8.50 - 8.47 (m, 2H), 8.15 (s, 1H), 7.34 (s, 1H), 7.31 - 7.28 (m, 1H), 7.21 (s, 2H), 6.88 (s, 1H), 4.04 (s, 2H), 2.76 - 2.70 (m, 2H), 1.81 - 1.71 (m, 5H), 1.65 - 1.57 (m, 2H), 1.39 - 1.35 (m, 1H).
Scheme 34. Synthesis of 4-((6'-methyl-6’H-spiro[cyclohexane-l,9'- pyrazino [1 ',2' : 1,5] pyrrolo [2, 3-d] pyrimidin] -2'-yl)amino)benzenesulfonamide
(COMPOUND 74)
Figure imgf000259_0001
Step 1: Synthesis of tert-butyl (4-(2-chloro-4-((l-(hydroxymethyl)cyclohexyl) amino)pyrimidin-5-yl)but-3-yn-2-yl)carbamate (74- 1)
Under N2 atmosphere, to a solution of (l-((2-chloro-5-iodopyrimidin-4-yl) amino)cyclohexyl)methanol (460.5 mg, 2.72 mmol) in THF (20 mL) was added tert-butyl but- 3-yn-2- ylcarbamate (1 g, 2.72 mmol), Cul (67.3 mg, 0.35 mmol), Pd(PPh3)2Ch (95.6 mg, 0.05 mmol) and TEA (550 mg, 5.45 mmol). After stirred at room temperature overnight, the reaction was quenched with water (20 mL) and extracted with EtOAc (15 mL x 3). The combined organic phase was washed with brine (10 mL x 2), dried over Na2S04, filtered and concentrated in vacuo. The resulting residue was purified by silica gel column chromatography to give tert- butyl (4-(2-chloro-4-((l-(hydroxymethyl)cyclohexyl)amino)pyrimidin-5-yl)but-3-yn-2-yl) carbamate 74-1 (750 mg, 1.83 mmol). Step 2: Synthesis of tert-butyl (4-(2-chloro-4-((l-formylcyclohexyl)amino) pyrimidin-5- yl) but-3-yn-2-yl)carbamate (74-2)
To a solution of tert-butyl (4-(2-chloro-4-((l-(hydroxymethyl)cyclohexyl) amino)pyrimidin-5-yl)but-3-yn-2-yl)carbamate 74-1 (1 g, 2.45 mmol) in DCM (20 mL) was added DMP (1.25 g, 2.94 mmol) at 0 °C. The reaction was stirred for 2 h. The reaction mixture was quenched with saturated aqueous sodium bicarbonate solution (20 mL) and saturated aqueous sodium thiosulfate solution (20 mL), extracted with DCM (20 mL x 3). The combined organic phase was dried over Na2S04, fdtered and concentrated in vacuo. The resulting residue was purified by silica gel column chromatography to provide tert-butyl (4-(2-chloro-4-((l- formylcyclohexyl) amino)pyrimidin-5-yl) but-3-yn-2-yl)carbamate 74-2 (850 mg, 2.09 mmol). LC-MS (ESI+): m/z 407 [M+H]+.
Step 3: Synthesis of tert-butyl 2'-chloro-8'-hydroxy-6'-methyl-6'H-spiro [cyclohexane- 1, 9'-pyrazino[r,2':l,5]pyrrolo[2,3-d]pyrimidine]-7'(8'H)- carboxylate (74-3)
Under N2 atmosphere, to a solution of tert-butyl (4-(2-chloro-4-((l- formylcyclohexyl)amino)pyrimidin-5-yl)but-3-yn-2-yl)carbamate 74-2 (550 mg, 1.35 mmol) in THF (5 mL) was added TBAF (4.1 mL, 1 M in THF) at 60 °C. After stirred at 60 °C for 3 h, the reaction was quenched with water (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic phase was washed with brine (10 mL x 2), dried over Na2S04, filtered and concentrated in vacuo. The resulting residue was purified by silica gel column chromatography to give tert-butyl 2'-chloro-8'- hydroxy-6'-methyl-6'H-spiro[cyclohexane-l,9'-pyrazino [l',2': l,5]pyrrolo[2,3-d] pyrimidine]-7'(8'H)-carboxylate 74-3 (100 mg, 0.25 mmol). LC-MS (ESI+): m/z 407 [M+H]+.
Step 4: Synthesis of tert-butyl 2'-((4-(N,N-bis(4-methoxybenzyl)sulfamoyl) phenyl)amino)-8'-hydroxy-6'-methyl-6'H-spiro[cyclohexane- L9'- pyrazino[l',2':l,5]pyrrolo[2,3-d]pyrimidine]-7'(8'H)-carboxylate (74-4)
Under N2 atmosphere, to a solution of tert-butyl 2'-chloro-8'-hydroxy- 6'- methyl-6'H- spiro[cyclohexane-l,9'-pyrazino[l',2': l,5]pyrrolo[2,3-d]pyrimidine]-7'(8'H)- carboxylate 74-3 (30 mg, 0.07 mmol) in DMF (5 mL) was added 4-amino-N,N-bis(4-methoxybenzyl) benzenesulfonamide (30.5 mg, 0.07 mmol), AcOK (21.7 mg, 0.22 mmol), Pd(OAc)2 (1.6 mg, 0.007 mmol) and X-phos (14 mg, 0.03 mmol). After the addition, the reaction mixture was stirred at 85 °C for 3 h. The reaction mixture was cooled to room temperature, quenched with water (15 mL) and extracted with EtOAC (10 mL x 3). The combined organic phase was washed with brine (10 mL x 2), dried over Na2S04 and concentrated in vacuo. The resulting residue was purified by preparative TLC to provide tert-butyl 2'-((4-(N,N-bis(4- methoxybenzyl) sulfamoyl)phenyl)amino)-8'-hydroxy-6'-methyl-6'H-spiro [cyclohexane- 1,9'- pyrazino[r,2,: l,5]pyrrolo[2,3-d]pyrimidine]-7'(8'H)- carboxylate 74-4 (6 mg, 0.008 mmol). LC-MS (ESI +): m/z 784 [M+H]+.
Step 5: Synthesis of 4-((6'-methyl-6'H-spiro[cyclohexane-l,9'-pyrazino [l',2':l,5]pyrrolo
[2,3-d]pyrimidin]-2'-yl)amino)benzenesulfonamide (COMPOUND 74)
To a solution of tert-butyl 2'-((4-(N,N-bis(4-methoxybenzyl)sulfamoyl) phenyl)amino)-8'-hydroxy-6'-methyl-6'H-spiro[cyclohexane-l,9'- pyrazino[r,2': l,5]pyrrolo[2,3-d]pyrimidine]-7'(8'H)-carboxylate 74-4 (30 mg, 0.04 mmol) in DCM (6 mL) was added TFA (2 mL) at rt. The reaction was stirred at rt overnight. The reaction mixture was neutralized with saturated aqueous Na2C03 (10 mL) and extracted with DCM:MeOH = 5: 1 (10 mL x 3). The combined organic phase was dried over Na2S04 and concentrated in vacuo. The resulting residue was purified by preparative TLC to provide 4-((6'- methyl-6'H-spiro[cyclohexane-l,9'-pyrazino [r,2': l,5]pyrrolo[2,3-d]pyrimidin]-2'- yl)amino)benzenesulfonamide COMPOUND 74 (3.2 mg, 0.01 mmol) as a yellow solid. LC- MS (ESI+): m/z 425 [M+H]+; 1HNMR (300 MHz, CD3OD) d 8.64 (s,lH), 8.46 - 8.45 (m, 1H), 7.98 (d, J = 9Hz, 2H), 7.86 (d, J = 9Hz, 2H), 6.34 (s, 1H), 4.98 - 4.91 (m, 1H), 3.56 - 3.50 (m, 1H), 3.23 - 3.19 (m, 1H), 2.09 - 1.82 (m, 8H), 1.68 ( d, J = 6.9Hz, 3H ).
Scheme 35. Synthesis of 4-((6'-methyl-8'H-spiro[cyclohexane-l,9'-pyrazino [l',2':l,5]pyrrolo[2,3-d]pyrimidin]-2'-yl)amino)benzenesulfonamide (COMPOUND 75)
Figure imgf000261_0001
Under N2 atmosphere, to a solution of 2'-chloro-6'-methyl-8'H-spiro[cyclohexane- 1,9'- pyrazino[r,2': l,5]pyrrolo[2,3-d]pyrimidine] (100 mg, 0.35 mmol) and 4- aminobenzenesulfonamide (71.7 mg, 0.42 mmol) in anhydrous DMF (5 mL) was added Pd(OAc)2 (8.8 mg, 0.04 mmol), X-phos (66.2 mg, 0.14 mmol) and KOAc (102.1 mg, 1.04 mmol) . The reaction mixture was stirred at 90 °C for 4h. After cooled to room temperature, the reaction mixture was quenched with water (10 mL) and extracted with MeOH/DCM=l :5 (10 mL x 3). The organic layer was separated and concentrated in vacuo to give the residue, which was purified by preparative TLC to provide 4-((6'-methyl-8'H-spiro[cyclohexane-l,9'- pyrazino[r,2': l,5]pyrrolo [2,3-d]pyrimidin]-2'-yl)amino)benzenesulfonamide COMPOUND 75 (16.3 mg, 0.038 mmol). LC-MS (ESI +): mJz 425 [M+H]+; ¾ NMR (300 MHz, DMSO -d6) d 8.90 (s, 1H), 7.97 (d, J = 9Hz, 2H), 7.74 (d, J = 9Hz, 2H), 7.22 (s, 2H), 6.98 (s, 1H), 3.98 (s, 2H), 2.89 - 2.78 (m, 2H), 2.34 (s, 3H), 1.88 - 1.74 (m, 5H), 1.68 - 1.55 (m, 2H), 1.45 - 1.33 (m, 1H).
Scheme 36. Synthesis of (R)-4-((6'-methyl-6'H-spiro [cyclohexane-1, 9'- pyrazino [1 ',2' : 1,5] pyrrolo [2,3-d] pyrimidin] -2'-yl)amino)benzenesulfonamide
(COMPOUND 76)
Figure imgf000262_0001
Figure imgf000263_0001
Step 1: Synthesis of tert-butyl (R)-(4-(2-chloro-4-((l-(hydroxymethyl)cyclohexyl)amino) pyrimidin-5-yl)but-3-yn-2-yl)carbamate (76- 1)
Under N2 atmosphere, to a solution of (l-((2-chloro-5-iodopyrimidin-4-yl)amino) cyclohexyl)methanol (2.6 g, 6.95 mmol) in DMF (20 mL) was added tert-butyl (R)-but-3-yn- 2-ylcarbamate (2 g, 11.89 mmol), Cul (165.51 mg, 0.87 mmol), Pd(PPh3)2Cl2 (243.99 mg, 0.35 mmol) and DIEA (3 g, 23.21 mmol). After stirred at room temperature overnight, the reaction was quenched with water (20 mL) and extracted with EtOAc (10 mL x 3). The combined organic phase was washed with brine (10 mL x 4), dried over Na2S04, filtered and concentrated in vacuo. The resulting residue was purified by silica gel column chromatography to give tert- butyl (R)-(4-(2-chloro-4-((l-(hydroxymethyl) cyclohexyl)amino)pyrimidin-5-yl)but-3-yn-2- yl) carbamate 76-1 (2.2 g, 5.38 mmol). LC-MS (ESI+): m/z 409 [M+H]+.
Step 2: Synthesis of tert-butyl (R)-(4-(2-chloro-4-((l-formylcyclohexyl)amino) pyrimidin -5-yl)but-3-yn-2-yl)carbamate (76-2)
To a solution of tert-butyl (R)-(4-(2-chloro-4-((l-(hydroxymethyl)cyclohexyl) amino)pyrimidin-5-yl)but-3-yn-2-yl)carbamate 76-1 (2.2 g, 5.38 mmol) in DCM (15 mL) was added DMP (2.7 g, 6.46 mmol) at 0 °C. After the addition, the reaction was stirred at rt for 2 h. The reaction mixture was quenched with saturated aqueous sodium bicarbonate solution (10 mL) and saturated aqueous sodium thiosulfate solution (10 mL), extracted with DCM (10 mL x 3). The combined organic phase was dried over Na2S04 and concentrated in vacuo. The resulting residue was purified by silica gel column chromatography to provide tert-butyl (R)- (4-(2-chloro-4- (( l-formylcyclohexyl)amino)pyrimi din-5 -yl)but-3-yn-2-yl) carbamate 76-2 (2.08 g, 5.11 mmol). LC-MS (ESI+): m/z 407 [M+H]+. Step 3: Synthesis of tert-butyl (6'R)-2'-chloro-8'-hydroxy-6'-methyl-6'H- spiro [cyclohexane -l,9'-pyrazino[ ,2':l,5]pyrrolo[2,3-d]pyrimi(line]-7'(8'H)- carboxylate (76-3)
Under N2 atmosphere, to a solution of tert-butyl (R)-(4-(2-chloro-4-((l- formylcyclohexyl)amino)pyrimidin-5-yl)but-3-yn-2-yl)carbamate 76-2 (300 mg, 0.74 mmol) in THF (5 mL) was added TBAF (2.2 mL, 1 M in THF) at 60 °C. After stirred at 60 °C for 3 h, the reaction was cooled to rt and quenched with water (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic phase was washed with brine (10 mL x 2), dried over Na2S04, filtered and concentrated in vacuo. The resulting residue was purified by silica gel column chromatography to give tert-butyl (6'R)-2'-chloro-8'- hydroxy-6'-methyl-6'H- spiro[cyclohexane-l,9'-pyrazino[l',2': l,5]pyrrolo[2,3-d] pyrimidine] -7'(8'H)-carboxylate 76-3 (55.8 mg, 0.14 mmol). LC-MS (ESI+): m/z 407 [M+H]+.
Step 4: Synthesis of tert-butyl (6'R)-2'-((4-(N,N-bis(4-methoxybenzyl)sulfamoyl)phenyl) amino)-8,-hydroxy-6,-methyl-6'H-spiro[cyclohexane-l,9,-pyrazino[l,,2,:l,5]pyrrolo[2,3- d]pyrimidine]-7'(8'H)-carboxylate (76-4)
Under N2 atmosphere, to a solution of tert-butyl (6'R)-2'-chloro-8'-hydroxy- 6'- methyl- 6'H-spiro[cyclohexane-l,9'-pyrazino[r,2': l,5]pyrrolo[2,3-d]pyrimidine]-7'(8'H)- carboxylate 76-3 (55.8 mg, 0.14 mmol) in DMF (5 mL) was added 4-amino-N,N-bis(4-methoxybenzyl) benzenesulfonamide (56.5 mg, 0.14 mmol), AcOK (40.3 mg, 0.41 mmol), Pd(OAc)2 (3.1 mg, 0.01 mmol) and X-phos (26.2 mg, 0.05 mmol). After stirred at 80 °C for 3 h, the reaction mixture was cooled to room temperature, quenched with water (10 mL) and extracted with EtOAC (10 mL x 3). The combined organic phase was washed with brine (10 mL x 2), dried over NaiSOi and concentrated in vacuo. The resulting residue was purified by silica gel column chromatography to provide tert-butyl (6'R)-2'-((4-(N,N-bis(4- methoxybenzyl) sulfamoyl)phenyl)amino)-8'-hydroxy-6'-methyl-6'H-spiro [cyclohexane-l,9'-pyrazino
[r,2': l,5]pynOlo[2,3-d]pyrimidine]-7'(8'H)- carboxylate 76-4 (70 mg, 0.09 mmol). LC-MS (ESI +): m/z [M+H]+.
Step 5: Synthesis of (R)-4-((6'-methyl-6'H-spiro[cyclohexane-l,9'-pyrazino [G,2':1,5] pyrrolo[2,3-d]pyrimidin]-2'-yl)amino)benzenesulfonamide (COMPOUND 76)
To a solution of tert-butyl (6'R)-2'-((4-(N,N-bis(4-methoxybenzyl)sulfamoyl) phenyl)amino)-8'-hydroxy-6'-methyl-6'H-spiro[cyclohexane-l,9'- pyrazino[r,2': l,5]pyrrolo[2,3-d]pyrimidine]-7'(8'H)-carboxylate 76-4 (70 mg, 0.09 mmol) in DCM (6 mL) was added TFA (2 mL). After the addition, the reaction was stirred at room temperature overnight. The reaction mixture was neutralized with saturated aqueous Na2C03 (10 mL) and extracted with DCM (10 mL x 2). The combined organic phase was dried over Na2S04 and concentrated in vacuo. The resulting residue was purified by preparative TLC to provide (R)-4-((6'-methyl-6'H-spiro[cyclohexane-l,9'- pyrazino[l',2': l,5]pyrrolo[2,3- d]pyrimidin]-2'-yl)amino)benzenesulfonaniide COMPOUND 76 (9.4 mg, 0.02 mmol) as a yellow solid. LC-MS (ESI+): m/z 425 [M+H]+; lHNMR (300 MHz, CD3OD/CDCI3) d 8.64 (s, 1H), 8.46 (s, 1H), 7.99 (d, J = 8.4Hz, 2H), 7.85 (d, J = 9.3Hz, 2H), 6.34 (s, 1H), 5.02 - 4.95 (m, 1H), 3.58 - 3.45 (m, 1H), 3.22 - 3.12 (m, 1H), 2.17 - 1.85 (m, 8H), 1.67 (d, J = 6.9Hz, 3H).
Scheme 37. Synthesis of (S)-4-((6'-methyl-6'H-spiro [cyclohexane-1, 9'- pyrazino [1 ',2' : 1,5] pyrrolo [2,3-d] pyrimidin] -2'-yl)amino)benzenesulfonamide
(COMPOUND 77)
Figure imgf000265_0001
Step 1: Synthesis of tert-butyl (S)-(4-(2-chloro-4-((l-(hydroxymethyl)cyclohexyl) amino)pyrimidin-5-yl)but-3-yn-2-yl)carbamate (77-1)
Under N2 atmosphere, to a solution of (l-((2-chloro-5-iodopyrimidin-4-yl)amino) cyclohexyl)methanol (2.6 g, 6.95 mmol) in DMF (20 mL) was added tert-butyl (S)-but-3-yn- 2-ylcarbamate (2 g, 6.95 mmol), Cul (165.51 mg, 0.87 mmol), Pd(PPh3)2Ck (243.99 mg, 0.35 mmol) and DIEA (3 g, 23.21 mmol). After stirred at room temperature overnight, the reaction mixture was quenched with water (30 mL) and extracted with EtOAc (30 mL x 3). The combined organic phase was washed with brine (10 mL x 2), dried over Na2S04, filtered and concentrated in vacuo. The resulting residue was purified by silica gel column chromatography to give tert-butyl (S)-(4-(2-chloro-4-((l-(hydroxymethyl)cyclohexyl)amino)pyrimi din-5- yl)but-3-yn-2-yl) carbamate 77-1 (2.8 g, 6.85 mmol). LC-MS (ESI+): m!z 409 [M+H]+.
Step 2: Synthesis of tert-butyl (S)-(4-(2-chloro-4-((l-foraiylcyclohexyl)amino) pyrimidin- 5-yl)but-3-yn-2-yl)carbamate (77-2)
To a solution of tert-butyl (S)-(4-(2-chloro-4-((l-(hydroxymethyl)cyclohexyl) amino)pyrimidin-5-yl)but-3-yn-2-yl)carbamate 77-1 (2.8 g, 6.85 mmol) in DCM (30 mL) was added DMP (3.2 g, 7.53 mmol) at 0 °C. After the addition, the reaction was stirred at room temperature for 2 h. The reaction mixture was quenched with saturated aqueous sodium bicarbonate solution (20 mL) and saturated aqueous sodium thiosulfate solution (20 mL), extracted with DCM (30 mL x 3). The combined organic phase was dried over Na2S04 and concentrated in vacuo. The resulting residue was purified by silica gel column chromatography to provide tert-butyl (S)-(4-(2-chloro-4-((l-formylcyclohexyl)amino)pyrimidin-5-yl)but-3-yn- 2-yl) carbamate 77-2 (2.08 g, 5.11 mmol). LC-MS (ESI+): m/z 407 [M+H]+
Step 3: Synthesis of tert-butyl (6'S)-2'-chloro-8'-hydroxy-6'-methyl-6'H-spiro [cyclohexane- 1, 9'-pyrazino[l',2':l,5]pyrrolo[2,3-d]pyrimidine]-7'(8'H)- carboxylate
(77-3)
Under N2 atmosphere, to a solution of tert-butyl (S)-(4-(2-chloro-4-((l- formylcyclohexyl)amino)pyrimidin-5-yl)but-3-yn-2-yl)carbamate 77-2 (500 mg, 1.23 mmol) in THF (5 mL) was added TBAF (3.7 mL, 1 M in THF) at 60 °C. After stirred at 60 °C for 3 h, the reaction mixture was quenched with water (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic phases were washed with brine (10 mL x 2), dried over Na2S04, filtered and concentrated m vacuo. The resulting residue was purified by silica gel column chromatography to give tert-butyl i6 S)-2 -chloro-8 -hydro\y-6 -methyl-6 H- spiro[cyclohexane-l,9’-pyrazino[r,2’: l,5] pyrrolo[2,3-d]pyrhmdine]-7’(8’H)-carboxylate 77-3 (62 mg, 0.15 mmol). LC-MS (ESI+): m/z 407 [M+H]+.
Step 4: Synthesis of tert-butyl (6’S)-2’-((4-(N,N-bis(4-methoxybenzyl)sulfamoyl) phenyl)amino)-8,-hydroxy-6,-methyl-6,H-spiro[cyclohexane-l,9,- pyrazino[l,,2,:l,5]pyrrolo[2,3-d]pyrimidine]-7,(8,H)-carboxylate (77-4)
Under N2 atmosphere, to a solution of tert-butyl (6'S)-2'-chloro-8'-hydroxy- 6'- methyl-6'H-spiro[cyclohexane-l,9'-pyrazino[r,2': l,5]pyrrolo[2,3-d]pyrimidine]-7'(8'H)- carboxylate 77-3 (60 mg, 0.15 mmol) in DMF (3 mL) was added 4-amino- N,N-bis(4- methoxybenzyl)benzenesulfonamide (67 mg, 0.16 mmol), AcOK(43.4 mg, 0.44 mmol), Pd(OAc)2 (3.3 mg, 0.01 mmol) and X-phos (28 mg, 0.06 mmol). After stirred at 85 °C for 3 h, the reaction mixture was cooled to room temperature, quenched with water (15 mL) and extracted with EtOAc (15 mL x 3). The combined organic phase was washed with brine (10 mL x 2), dried over Na2S04 and concentrated in vacuo. The resulting residue was purified by silica gel column chromatography to provide tert-butyl (6'S)-2'-((4-(N,N-bis(4- methoxybenzyl) sulfamoyl)phenyl)amino)-8'-hydroxy-6'-methyl-6'H-spiro[cyclohexane-l,9'- pyrazino [r,2': l,5]pyrrolo[2,3-d]pyrimidine]-7'(8'H)- carboxylate 77-4 (92 mg, 0.11 mmol). LC-MS (ESI +): m/z 784 [M+H]+.
Step 5: Synthesis of (S)-4-((6'-methyl-6'H-spiro [cyclohexane-1, 9'-pyrazino [r,2,:l,5]pyrrolo[2,3-d]pyrimidin]-2'-yl)amino)benzenesulfonamide (COMPOUND 77)
To a solution of tert-butyl (6'S)-2'-((4-(N,N-bis(4-methoxybenzyl)sulfamoyl) phenyl)amino)-8'-hydroxy-6'-methyl-6'H-spiro[cyclohexane-l,9'- pyrazino[r,2': l,5]pyrrolo[2,3-d]pyrimidine]-7'(8'H)-carboxylate 77-4 (92 mg, 0.11 mmol) in DCM (3 mL) was added TFA (1 mL). After the addition, the reaction was stirred at room temperature overnight. The reaction mixture was neutralized with saturated aqueous Na2C03 (10 mL) and extracted with DCM:MeOH = 5: 1 (10 mL x 3). The combined organic phase was dried over Na2S04 and concentrated in vacuo. The resulting residue was purified by preparative TLC to provide (S)-4-((6'-methyl-6'H-spiro [cyclohexane-l,9'-pyrazino[r,2': l,5]pyrrolo[2,3- d]pyrimidin]-2'-yl)amino) benzenesulfonamide COMPOUND 77 (30.4 mg, 0.07 mmol) as a yellow solid. LC-MS (ESI+): m/z 425 [M+H]+; !HNMR (300 MHz, CD3OD) d 8.64 (s, 1H), 8.46 (s, 1H), 7.99 (d, J = 7.8Hz, 2H), 7.83 (d, J = 8.1Hz, 2H), 6.35 (s, 1H), 5.02 - 4.95 (m, 1H), 3.57 - 3.45 (m, 1H), 3.22 - 3.08 (m, 1H), 2.20 - 1.79 (m, 8H), 1.66 (d, J = 6.6Hz, 3H). Example 2. Representative Compounds of the Present Invention
Table 1
Figure imgf000268_0001
Figure imgf000269_0001
Figure imgf000270_0001
Figure imgf000271_0001
Figure imgf000272_0001
Figure imgf000273_0001
Figure imgf000274_0001
Figure imgf000275_0001
Figure imgf000276_0001
Figure imgf000277_0001
Figure imgf000278_0001
Figure imgf000279_0002
Table IB
Figure imgf000279_0001
Figure imgf000280_0001
Figure imgf000281_0001
Figure imgf000282_0001
Figure imgf000283_0001
Table 1C
Figure imgf000283_0002
Figure imgf000284_0001
Figure imgf000285_0001
Table ID
Figure imgf000286_0001
Figure imgf000287_0001
Figure imgf000288_0001
Example 3: CDK Inhibition In Vitro Assays
Selected compounds disclosed herein were tested in kinase assays by Nanosyn (Santa Clara, CA) to determine their inhibitory effect on these CDKs. The assays were performed using microfluidic kinase detection technology (Caliper Assay Platform). The compounds were tested in 12-point dose-response format in singlicate at Km for ATP. Specifics of each assay are as described below:
CDKl/Cyclin Bl: Enzyme concentration: 0.08 nM; ATP concentration: 40 mM; Incubation time: 3 hr.
CDK2/Cyclin A: Enzyme concentration: 0.1 nM; ATP concentration: 50 mM;
Incubation time: 3 hr.
CDK2/Cyclin E: Enzyme concentration: 0.15 nM; ATP concentration: 100 mM; Incubation time: 3 hr.
CDK4/Cyclin Dl : Enzyme concentration: 1 nM; ATP concentration: 200 mM; Incubation time: 3 hr.
CDK6/Cyclin D3: Enzyme concentration: 2 nM; ATP concentration: 300 mM; Incubation time: 3 hr.
CDK9/Cyclin Tl : Enzyme concentration: 5 nM; ATP concentration: 10 mM; Incubation time: 17 hr.
Figure imgf000289_0001
Figure imgf000290_0001
Table 2B: Biological Data
Figure imgf000290_0002
Table 2C: Biological Data
Figure imgf000291_0001
Table 3: Biological Data
Figure imgf000291_0002
Figure imgf000292_0001
Figure imgf000293_0001
Table 3B: Biological Data
Figure imgf000293_0002
Table 3C: Biological Data
Figure imgf000293_0003
Example 4. Small Molecule Inhibitors are Potent against CDK2/Cyclin E
The activity of select compounds against a panel of CDKs and their respective binding partners is shown in Table 4. Assays were completed in a 12 point dose-response format by Nanosyn, Inc. Results are shown as nanomolar IC50 concentrations against each target. Compounds 1, 2, 3, and 74 have sub-nanomolar potencies against CDK2 when complexed to Cyclin E or Cyclin A.
Table 4. Biochemical Profile of Select Compounds
Figure imgf000294_0001
*PF = PF-0687300
Compounds 1, 2, and 3 were also tested in the NanoBRET™ Target Engagement Intracellular Kinase Assay (Figure 1). As shown in Figure 1, as the concentration of all three compounds increased, the acceptor : donor emission ratio (the BRET ratio) decreased. The IGo values for Compounds 1, 2, and 3 for binding to CDK2/Cyclin E as determined by the NanoBRET™ Target Engagement Intracellular Kinase Assay are shown in Table 5. All three compounds exhibited potent binding of CDK2/Cyclin E in cells. Pfizer’s CDK2 Inhibitor, PF- 0687300, was used as a reference compound in both experiments of Example 4.
Table 5. Activity of Select Compounds in the Target Engagement Intracellular Kinase Assay
Figure imgf000294_0002
Example 5. Selectivity of Select Compounds against Kinome
Kinome screens of Compounds 1, 2, and 3 against a panel of 485 kinases were performed by SelectScreen Kinase Profiling Services at Thermo Fisher Scientific. Figure 2A, Figure 2B, and Figure 2C are kinome trees of kinases inhibited 95% by Compound 1, Compound 2, Compound 3, respectively (Cell Signaling Technology, Inc). CDK2 is the labelled circle. Table 6 corresponds to Figure 2A and provides the kinase name and percent inhibition of kinases inhibited greater than 95% by Compound 1. Compound 1 was assessed at 200x the biochemical ICso of CDK2/CyclinE. Table 7 corresponds to Figure 2B and provides the kinase name and percent inhibition kinases inhibited greater than 95% by Compound 2. Compound 2 was assessed at lOOOx the biochemical IC50 of CDK2/CyclinE. Table 8 corresponds to Figure 2C and provides the kinase name and percent inhibition kinases inhibited greater than 95% of Compound 3. Compound 3 was assessed at 250x the biochemical IC50 of CDK2/CyclinE. All three compounds exhibit significant binding to the target protein. Table 6. Kinases Inhibited Greater than 95% by Compound 1
Figure imgf000295_0001
Table 7. Kinases Inhibited Greater than 95% by Compound 2
Figure imgf000296_0001
Table 8. Kinases Inhibited Greater than 95% by Compound 3
Figure imgf000296_0002
Example 6. Inhibitors Prevent Incorporation of EdU in S-Phase of Cell Cycle
MCF7 Parental cells (ATCC, ER+ Her2- BC, CDK4/6 dependent) were maintained in culture for four months in complete media (EMEM/10% FBS/glutamax/ insulin) as a control. MCF7 palbociclib-resistant (Palbo-R) cells were maintained in complete media plus palbociclib for three months at ~IC9o (750nM) followed by one month at lpM. Pictures of the cells in culture depict the similar morphology and growth of the parental and resistant cell lines (Figure 3 A). When MCF7 parental cells (Figure 3B) and Palbo-R cells (Figure 3C) were treated with palbociclib, the ICso was 87x higher in the Palbo-R cells compared to the control as measured using the 6-Day CellTiter Glo Assay. The ICso of palbociclib in the MCF7 cell line was 33 nM and the IC50 in the MCF7 Palbo-R cell line was 2890 nM.
Whole transcriptome profding was performed on MCF7 parental cells and Palbo-R MCF7 cells by RNA-Seq. Libraries were prepared using the Illumina TruSeq Stranded mRNA assay and paired-end sequenced (2x50bp) on the Illumina HiSeq platform. Figure 4A is the pairwise comparison of transcript levels in MCF7 Palbo-R vs. MCF7 parental. 6,039 genes out of 17,383 detectable genes were differentially expressed with adjusted p-value <0.05 (genes above the horizontal dashed line in Figure 4A). Figure 4B is the fold change (expressed as Log2) of the transcript levels of specific genes in MCF7 palbo-R vs. MCF7 parental. In Figure 4B, the transcript levels of CCNE1, CCNE2, and CDK6 were significantly upregulated in the MCF7 Palbo-R cells compared to the MCF7 parental cells. The Western blot analysis of MCF7 parental and MCF7 palbo-R cells demonstrated an increase in the ratio of Cyclin E to Rb levels in Palbo-R cells compared to the MCF7 parental cells, with quantitation of normalized signal to loading control, GAPDH, located at the bottom of the Western blot (Figure 4C).
HCC1806 (TNBC, CDK4/6 independent) (Figure 5A), MCF7 (Figure 5B), MCF7 Palbo-R (Figure 5C), and Hs68 (normal human fibroblasts) (Figure 5D) cell lines were treated with Compound 1, Compound 2, Compound 3, and PF-0687300 for 24 hours (PF-0687300 was a reference compound). Cell cycle profiles were evaluated following treatment via Flow Cytometry using FlowJo (vlO.O) software. All four cell lines showed a dose-dependent decrease in the incorporation of EdU during the S-phase of the cell cycle and a decrease in phosphorylated phospho-histone H3 (SerlO) following treatment with Compound 1, Compound 2, or Compound 3. Representative flow gating for FxCycle DNA stain, Click-iTTM EdU Flow Cytometry Assay Kit (Figure 5E and Figure 5F) and Phospho-Histone H3 conjugated antibody (Figure 5G and Figure 5H) from HCC1806 samples treated with vehicle (Figure 5E and Figure 5G) and 1000 nM of Compound 3 (Figure 5F and Figure 5H) were performed.
Example 7. Inhibitors Decrease Phosphorylation of Rb
HCC1806 (Figure 6A), MCF7 Palbo-R (Figure 6B), and Hs68 (Figure 6C) cell lines were treated for 24 hours with Compound 1, Compound 2, Compound 3, and PF-0687300 at IOOhM, 300nM, or lOOOnM (PF-0687300 was a reference compound). Compound 1, Compound 2, and Compound 3 showed a dose-dependent decrease in phosphorylated-Rb (Ser807/811). The corresponding graphs shown below each Western blot in Figure 6A, Figure 6B, and Figure 6C depict changes in p-Rb and p21 signals normalized to loading control alpha- tubulin and relative to vehicle.
Example 8. Inhibitors Prevent Proliferation of TNBC, MCF7 Palbo-R and Normal Cells
Two triple-negative breast cancer cell lines, HCC1806 (Figure 7A) and BT549 (Figure 7B), in addition to cell lines MCF7 (Figure 7C), MCF7 Palbo-R (Figure 7D) and Hs68 (Figure 7E), were treated with Compound 1, Compound 2, Compound 3, and PF-0687300 at concentrations ranging from 1 nM to 10 mM in the 6-Day CellTiter Glo Asay (PF-0687300 was a reference compound). Table 6 provides the corresponding I vCal5u0es for each cell line. This data demonstrates the potency of CDK2 inhibitors and the correlation between increased biochemical potency on CDK2/CyclinE and increased potency in vitro.
Table 6. Activity of Compounds 1, 2, and 3 in Various Cell Lines
Figure imgf000298_0001
Example 9. Inhibitors do not Elicit Caspase 3/7 Activation at 24 Hours
Compounds 1 and 3 did not elicit caspase 3/7 in either the HCC 1806 (Figure 8 A) or the Hs68 (Figure 8B) cell line. The cell lines were incubated with concentrations of Compound 1, Compound 3, Pfizer’s PF-06873600 or control (staurosporine) at concentrations ranging from InM to 3 mIM for 24 hours. Staurosporine clearly induced activation of caspase 3/7, while Compound 1 and Compound 3 did not.
Example 10. Evaluation of Compound 2 in In Vivo PDX Human Breast Cancer Model
To evaluate the in vivo activity of Compound 2, palbociclib-resistant human PDX breast tumors (CTG-2891) were established in female athymic nude mice aged 6 to 8 weeks of age. When tumors reached an average tumor volume of 150-300mm3 animals, the tumor in each animal was measured, and the mice where matched by tumor volume into treatment or control groups used for dosing, with dosing initiating on Day 0. Mice were dosed with a control vehicle, 100 mg/kg once daily, or 75 mg/kg twice a day for 14 days. Tumor dimensions were measured twice weekly by digital caliper and data including individual and mean estimated tumor volumes (Mean TV ± SEM) recorded for each group; tumor volume was calculated using the formula (1): TV= width2 x length x 0.52. The results are provided in Fig. 9, and show that Compound 2 at both dosing levels significantly reduced the growth of the tumor. In addition, no notably toxicity in either dosing group was observed, and no significant weight loss was noted in the two treatment groups.
Example 11. Inhibitors Induce Cell Cycle Arrest
Hs68 (normal human fibroblasts) (Figure 10) cell lines were treated with Compound 74 for 24 hours. DMSO was used as a control. Cell cycle profiles were evaluated following treatment via Flow Cytometry using FlowJo (vlO.O) software. Hs68 cells showed a dose- dependent cell cycle arrest. The results are provided in Fig. 10, and show a prominent cell cycle arrest at 100 nM of Compound 74.
Hs68 (normal human fibroblasts) (Figure 11) cell lines were treated with Compound 75for 24 hours. A vehicle control, Compound 2 at 300 nM, and G1T38, a potent CDK4/6 inhibitor, was used as a comparative. Cell cycle profiles were evaluated following treatment via Flow Cytometry using FlowJo (vlO.O) software. Hs68 cells showed a dose-dependent cell cycle arrest when exposed to Compound 75.
All publications and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference.
The descriptions herein are described by way of illustration and example for purposes of clarity of understanding for embodiments only. It will be readily apparent to one of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the invention as defined in the appended claims.

Claims

Claims I Claim:
1. A compound of Formula:
Figure imgf000300_0001
Figure imgf000301_0001
Figure imgf000302_0001
or a pharmaceutically acceptable salt thereof;
wherein
y is 0, 1, 2, 3, or 4;
x is 0, 1, 2, 3, or 4;
L is selected from -S(O)-, -S(O)2-, -C(O)-, and -C(S)-; X1, X2, X3, X4, and X5 are independently selected from N, CH, CR2, and CR4; wherein at least one of X1, X2, X3, X4, and X5 is CR2; and wherein no more than 2 of X1, X2, X3, X4, and X5 are selected to be N;
X6, X7. X8, X9, and X10 are independently selected from N, CH, CR2, and CR4; and wherein no more than 2 of X6, X7. X8, X9, and X10 are selected to be N;
each R1 is independently halogen, -OR7, hydrogen, alky l, aryl, cycloalkyl, haloalkyl, heteroaryl, alkyl-hydroxyl, or heterocycle, wherein two R3s on adjacent ring atom(s) or on the same ring atom may come together with the ring atom(s) to which they are attached to optionally constitute a 3, 4, 5, 6, 7, or 8-membered cycloalkyl or 4, 5, 6, 7, or 8-membered heterocycle that has 1, 2, or 3 heteroatoms selected from N, O, and S; wherein the cycloalkyl or heterocycle formed by combining two R 1 s with the atom(s) to which they are attached can be optionally substituted with 1 or 2 substituents independently selected from R50;
each R2 is independently -NR14C(O)R6, -NR14S(O)R6, -NR14S(O)2R6, -NR14C(S)R6, - 0C(O)R6, -OS(O)R6, -OS(O)2R6, -OC(S)R6, -C(O)R6; -C(S)R6, -S(O)R6, or -S(O)2R6;
R3 is selected from:
Figure imgf000303_0001
each R4 is independently hydrogen, alky l, aryl, cycloalkyl, haloalkyl, heteroaryl, heterocycle, halogen, cyano, -OR14, -NR14R15, -NR14C(O)R6, -NR14S(O)R6, -NR14S(O)2R6, -NR14C(S)R6, -0C(O)R6, -OS(O)R6, -OS(O)2R6, -OC(S)R6, -C(O)R6; -C(S)R6, -S(O)R6, or
-S(O)2R6;
R5 is hydrogen, alkyl, aryl cycloalkyl, haloalkyl, heteroaryl, heterocycle, -C(O)R6, -C(S)R6, -S(O)R6, -S(O)2R6, -C(O)NR7R7, -C(S)NR7R7, -S(O)NR7R7, -S(O)2NR7R7, -C(O)0R7, -C(S)OR7, -S(O)0R7, or -S(O)20R7; each R6 is selected independently from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocy cle, aryl, heteroaryl, NR'R7, and OR7 each of which R6 except hydrogen, NR7R7, and OR7 is optionally substituted with 1, 2, 3, or 4 R8 groups;
each R7 is independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycle, aryl, alkyl-aryl, alkyl-heteroaryl, and heteroaryl each of which R7 except hydrogen is optionally substituted with 1, 2, 3, or 4 R8 groups;
each R8 is independently selected from hydrogen, halogen, haloalkyl, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycle, aryl, heteroaryl, -S(O)2alkyl, NR12R13, alkyl-heteroaryl, alkyl-aryl, and OR12;
R11 is selected from halogen, haloalkyl, hydrogen, alkyl, -NR12R13, -S(O)alkyl, - SO2alkyl, cycloalkyl, heterocycle, aryl, heteroaryl, alkyl-aryl, and alkyl-heteroaryl;
each R12 and R13 are independently selected from hydrogen, alkyl, alkenyl, alkynyl, -C(O)alkyl, -C(S)alkyl, aryl, -SO2alkyl, -SOalkyl, heteroaryl, alkyl-aryl, cycloalkyl, heterocycle, and alkyl-heteroaryl;
each R14 and R15 are independently selected from hydrogen, alkyl, alkenyl, alkynyl, -C(O)R6, -C(O)alkyl, -C(S)alkyl, aryl, -SO2alkyl, heteroaryl, heterocycle, -alkyl-aryl, and -alkyl-heteroaryl;
R16 is selected from hydrogen, halogen, alkyl, hydroxyl, alkoxy, and haloalkyl;
R17 is selected from:
Figure imgf000304_0001
R18 is selected from: cycloalkyl, heterocycle, and aryl, each of which R18 is substituted with at least one R2 group, and wherein each R18 is optionally substituted with 1, 2, 3, or 4 substituents independently selected from R4 and R5; R19 is selected from
Figure imgf000305_0001
R20 is selected from alkyl, alkenyl, alkynyl, -C(O)R6, -C(O)alkyl, -C(S)alkyl, aryl, -S02alkyl, heteroaryl, heterocycle, -alkyl-aryl, and -alkyl-heteroaryl;
R21 is selected from hydrogen, alkyl, -S(O)alkyl, -SO2alkyl, cycloalkyl, heterocycle, aryl, heteroaryl, alkyl-aryl, and alkyl-heteroaryl;
R50 is selected from hydrogen, amino, -NHR14, -NR14R15, hydroxyl, OR14, and R4; and y2 is 0, 1, 2, 3, or 4;
each m is independently 0 or 1;
each n is independently 0, 1, or 2;
each R31 is independently alkyl, aryl, cycloalkyl or haloalkyl, wherein each of said alkyl, cycloalkyl and haloalkyl groups optionally includes heteroatoms O, N, or S in place of a carbon in the chain and two R31s on adjacent ring atoms or on the same ring atom together with the ring atom(s) to which they are attached optionally form a 3-8-membered cycle or two R31s on adjacent ring atoms together with the ring atoms to which they are attached optionally form a 6-membered aryl ring;
R32 is -(alkylene)m-heterocyclo, -(alkylene)m-heteroaryl, -(alkylene)m-NR33R34, -(alkylene)m-C(O)-NR33R34; -(alkylene)m-C(O)-O-alkyl; -(alkylene)m-O-R35, -(alkylene)m-S(O)n-R35, or -(alkylene)m-S(O)n-NR33R34 any of which may be optionally independently substituted with one or more Rx groups as allowed by valance, and wherein two Rx groups bound to the same or adjacent atom may optionally combine to form a ring;
R32* is -alkylene-heterocyclo, -(alkylene)m-heteroaryl, -alkylene-NR33R34, -(alkylene)m-C(O)-NR33R34; -(alkylene)m-C(O)-O-alkyl; -alkylene-S(O)n-R35, or -(alkylene)m-S(O)n-NR33R34 any of which may be optionally independently substituted with one or more Rx groups as allowed by valance, and wherein two Rx groups bound to the same or adjacent atom may optionally combine to form a ring;
R33 and R34 at each occurrence are independently:
(v) hydrogen or
(vi) alkyl, cycloalkyl, heterocyclo, aryl, heteroaryl, cycloalkylalkyl, heterocycloalkyl, arylalkyl, or heteroarylalkyl; or R33 and R34 together with the nitrogen atom to which they are attached may combine to form a heterocyclo ring;
R35 is independently:
(i) hydrogen or
(ii) alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclo, aryl, heteroaryl, cycloalkylalkyl, heterocycloalkyl, arylalkyl, or heteroarylalkyl;
Rx at each occurrence is independently selected from halo, cyano, nitro, oxo, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclo, ary l heteroaryl, arylalky l, heteroarylalkyl, cycloalkylalkyl, heterocycloalkyl, -(alkylene)m-OR35, -(alkylene)m-O-alkylene-OR35, -(alkylene)m-S(O)n-R35, -(alkylene)m-NR33R34, -(alkylene)m-CN, -(alkylene)m-C(O)-R35, -(alkylene)m-C(S)-R35, -(alkylene)m-C(O)-OR35, -(alkylene)m-O-C(O)-R35,
-(alkylene)m-C(S)-OR35, -(alkylene)m-C(O)-(alkylene)m-NR33R34,
-(alkylene)m-C(S)-NR33R34, -(alkylene)m-N(R33)-C(O)-NR33R34,
-(alkylene)m-N(R33)-C(S)-NR33R34, -(alkylene)m-N(R33)-C(O)-R35,
-(alkylene)m-N(R33)-C(S)-R35, -(alkylene)m-O-C(O)-NR33R34,
-(alkylene)m-O-C(S)-NR33R34, -(alkylene)m-S02-NR33R34, -(alkylene)m-N(R33)-SO2-R35, -(alkylene)m-N(R33)-SO2-NR33R34, -(alkylene)m-N(R33)-C(O)-OR35,
-(alkylene)m-N(R33)-C(S)-OR35, or -(alkylene)m-N(R33)-SO2-R35.
2. The compound of claim 1 of formula:
Figure imgf000306_0001
or a pharmaceutically acceptable salt thereof
3. The compound of claim 2, wherein R21 is hydrogen.
4. The compound of claim 2, wherein R21 is alkyl.
5. The compound of claim 2, wherein R21 is halogen.
6. The compound of claim 1 of formula:
Figure imgf000307_0001
or a pharmaceutically acceptable salt thereof.
7. The compound of claim 6, wherein R 20 is alkyl.
8. The compound of claim 6, wherein R20 is methyl.
9. The compound of any one of claims 1-8, wherein
Figure imgf000307_0002
10. The compound of any one of claims 1-8, wherein
Figure imgf000307_0003
11. The compound of any one of claims 1-8, wherein
Figure imgf000307_0004
12. A compound of Formula:
Figure imgf000307_0005
Figure imgf000308_0001
Figure imgf000309_0001
or a pharmaceutically acceptable salt thereof;
wherein
y is 0, 1, 2, 3, or 4;
x is 0, 1, 2, 3, or 4;
L is selected from -S(O)-, -S(O)2-, -C(O)-, and -C(S)-; X1, X2, X3, X4, and X5 are independently selected from N, CH, CR2, and CR4; wherein at least one of X1, X2, X3, X4, and X5 is CR2; and wherein no more than 2 of X1, X2, X3, X4, and X5 are selected to be N;
X6, X7. X8, X9, and X10 are independently selected from N, CH, CR2, and CR4; and wherein no more than 2 of X6, X7. X8, X9, and X10 are selected to be N;
each R1 is independently halogen, -OR7, hydrogen, alkyl aryl, cycloalkyl, haloalkyl, heteroaryl, alkyl-hydroxyl, or heterocycle, wherein two R1s on adjacent ring atom(s) or on the same ring atom may come together with the ring atom(s) to which they are attached to optionally constitute a 3, 4, 5, 6, 7, or 8-membered cycloalkyl or 4, 5, 6, 7, or 8-membered heterocycle that has 1, 2, or 3 heteroatoms selected from N, 0, and S; wherein the cycloalkyl or heterocycle formed by combining two R 1 s with the atom(s) to which they are attached can be optionally substituted with 1 or 2 substituents independently selected from R50;
each R2 is independently -NR14C(O)R6, -NR14S(O)R6, -NR14S(O)2R6, -NR14C(S)R6, - 0C(O)R6, -OS(O)R6, -OS(O)2R6, -OC(S)R6, -C(O)R6; -C(S)R6, -S(O)R6, or -S(O)2R6;
R3 is selected from:
Figure imgf000310_0001
each R4 is independently hydrogen, alky l, aryl cycloalkyl, haloalkyl, heteroaryl, heterocycle, halogen, cyano, -OR14, -NR14R15, -NR14C(O)R6, -NR14S(O)R6, -NR14S(O)2R6, -NR14C(S)R6, -0C(O)R6, -OS(O)R6, -OS(O)2R6, -OC(S)R6, -C(O)R6; -C(S)R6, -S(O)R6, or -S(O)2R6;
R5 is hydrogen, alkyl, aryl cycloalkyl, haloalkyl, heteroaryl, heterocycle, -C(O)R6, -C(S)R6, -S(O)R6, -S(O)2R6, -C(O)NR6R6, -C(S)NR6R6, -S(O)NR6R6, -S(O)2NR6R6, -C(O)0R6, -C(S)OR6, -S(O)0R6, or -S(O)20R6;
each R6 is selected independently from hydrogen, alkyl, alkenyl, alkynyl. cycloalkyl, heterocycle, aryl, heteroaryl, NR R . and OR7 each of which R6 except hydrogen, NR7R7, and OR7 is optionally substituted with 1, 2, 3, or 4 R8 groups; each R7 is independently selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycle, aryl, alkyl-aryl, alkyl-heteroaryl, and heteroaryl each of which R7 except hydrogen is optionally substituted with 1, 2, 3, or 4 R8 groups;
each R8 is independently selected from hydrogen, halogen, haloalkyl, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycle, aryl, heteroaryl, -S(O)2alkyl, NR12R13, alkyl-heteroaryl, alkyl-aryl, and OR12;
R11 is selected from halogen, haloalkyl, hydrogen, alkyl, -NR12R13, -S(O)alkyl, - SO2alkyl, cycloalkyl, heterocycle, aryl, heteroaryl, alkyl-aryl, and alkyl-heteroaryl:
each R12 and R13 are independently selected from hydrogen, alkyl, alkenyl, alkynyl, -C(O)alkyl, -C(S)alkyl, aryl, -SO2alkyl, -SOalkyl, heteroaryl, alkyl-aryl, cycloalkyl, heterocycle, and alkyl-heteroaryl;
each R14 and R15 are independently selected from hydrogen, alkyl, alkenyl, alkynyl, -C(O)R6, -C(O)alkyl, -C(S)alkyl, aryl, -SO2alkyl, heteroaryl, heterocycle, -alkyl-aryl, and -alkyl-heteroaryl;
R16 is selected from hydrogen, halogen, alkyl, hydroxyl, alkoxy, and haloalkyl;
R17 is selected from:
Figure imgf000311_0001
R18 is selected from: cycloalkyl, heterocycle, and aryl, each of which R18 is substituted with at least one R2 group, and wherein each R18 is optionally substituted with 1, 2, 3, or 4 substituents independently selected from R4 and R5; and
R50 is selected from hydrogen, amino, -NHR14, -NR14R15, hydroxyl, OR14, and R4.
13. The compound of claim 12, wherein the compound is of Formula:
Figure imgf000312_0001
or a pharmaceutically acceptable salt thereof;
wherein:
each R1 is independently hydrogen, alkyl, aryl cycloalkyl, haloalkyl, heteroaryl, alkyl-hydroxyl, or heterocycle, wherein two R1s on adjacent ring atom(s) or on the same ring atom may come together with the ring atom(s) to which they are attached to optionally constitute a 3, 4, 5, 6, 7, or 8-membered cycloalkyl or 4, 5, 6, 7, or 8-membered heterocycle that has 1 , 2, or 3 heteroatoms selected from N, 0, and S; wherein the cycloalkyl or heterocycle formed by combining two R1s with the atom(s) to which they are attached can be optionally substituted with 1 or 2 substituents independently selected from R50; R3 is selected from:
Figure imgf000313_0001
R11 is selected from hydrogen, alkyl, -NR12R13, -S(O)alkyl, -SO2alkyl, cycloalkyl, heterocycle, aryl, heteroaryl, alkyl-aryl, and alkyl-heteroaryl.
14. The compound of any one of claims 1-13, wherein two R3s together with the carbon to which they are attached form a 3-8 membered cycle.
15. The compound of any one of claims 1-13, wherein two R3s together with the carbon to which they are attached form a 6-membered carbocycle.
16. The compound of any one of claims 1-15, wherein y is 3.
17. The compound of any one of claims 1-15, wherein y is 4.
18. The compound of any one of claims 1-17, wherein one R1 is halogen.
19. The compound of any one of claims 1-17, wherein one R1 is haloalkyl.
20. The compound of any one of claims 1-17, wherein one R1 is hydroxyl.
21. The compound of any one of claims 1-20, wherein R1 is selected from alkyl, aryl, cycloalkyl, and haloalkyl.
22. The compound of any one of claims 1-21, wherein R2 is -C(O)R6; -C(S)R6, -S(O)R6, or -S(O)2R6.
23. The compound of any one of claims 1-21, wherein R2 is -C(O)R6.
24. The compound of any one of claims 1-21, wherein R2 is -S(O)2R6.
25. The compound of any one of claims 1-24, wherein each R6 is selected independently from hydrogen, alky l, alkenyl, alkynyl, cycloalky l, heterocycle, aryl, and heteroaryl.
26. The compound of any one of claims 1-24, wherein each R6 is selected independently from NR7R7 and OR7.
27. The compound of any one of claims 1-21, wherein R2 is -S(O)2NH2.
28. The compound of any one of claims 1-27, wherein R11 is hydrogen.
29. The compound of any one of claims 1-27, wherein R11 is halogen.
30. The compound of any one of claims 1-27, wherein R11 is haloalkyl.
31. The compound of any one of claims 1-30, wherein the compound is of Formula:
Figure imgf000314_0001
or a pharmaceutically acceptable salt thereof.
32. The compound of any one of claims 1-30, wherein the compound is of Formula:
Figure imgf000314_0002
or a pharmaceutically acceptable salt thereof.
33. The compound of any one of claims 1-30, wherein the compound is of Formula:
Figure imgf000315_0001
or a pharmaceutically acceptable salt thereof.
34. The compound of any one of claims 1-30, wherein the compound is of Formula:
Figure imgf000315_0002
or a pharmaceutically acceptable salt thereof.
35. The compound of any one of claims 1-30, wherein the compound is of Formula:
Figure imgf000316_0001
or a pharmaceutically acceptable salt thereof
36. The compound of any one of claims 1-30, wherein the compound is of Formula:
Figure imgf000316_0002
or a pharmaceutically acceptable salt thereof.
37. The compound of any one of claims 1-30, wherein the compound is of Formula:
Figure imgf000316_0003
Figure imgf000317_0001
or a pharmaceutically acceptable salt thereof.
38. The compound of any one of claims 34-37, wherein R16 is hydroxyl.
39. The compound of any one of claims 34-37, wherein R16 is fluorine.
40. The compound of any one of claims 1-39, wherein
Figure imgf000317_0002
41. The compound of any one of claims 1-39, wherein
Figure imgf000317_0003
42. The compound of any one of claims 1-39, wherein X1 is CH.
43. The compound of any one of claims 1-39, wherein X1 is N.
44. The compound of any one of claims 1-39, wherein X1 is CR2.
45. The compound of any one of claims 1-39, wherein X1 is CR4.
46. The compound of any one of claims 1-45, wherein X2 is CH.
47. The compound of any one of claims 1-45, wherein X2 is N.
48. The compound of any one of claims 1-45, wherein X2 is CR2.
49. The compound of any one of claims 1-45, wherein X2 is CR4.
50. The compound of any one of claims 1-49, wherein X3 is CH.
51. The compound of any one of claims 1-49, wherein X3 is N.
52. The compound of any one of claims 1-49, wherein X3 is CR2.
53. The compound of any one of claims 1-49, wherein X3 is CR4.
54. The compound of any one of claims 1-53, wherein X4 is CH.
55. The compound of any one of claims 1-53, wherein X4 is N.
56. The compound of any one of claims 1-53, wherein X4 is CR2.
57. The compound of any one of claims 1-53, wherein X4 is CR4.
58. The compound of any one of claims 1-57, wherein X1 is CH.
59. The compound of any one of claims 1-57, wherein X1 is N.
60. The compound of any one of claims 1-57, wherein X1 is CR2.
61. The compound of any one of claims 1-57, wherein X1 is CR4.
62. The compound of any one of claims 1-39, wherein R3 is
Figure imgf000318_0001
63. The compound of any one of claims 1-39, wherein R3 is
Figure imgf000318_0002
64. The compound of any one of claims 1-39, wherein R3 is
Figure imgf000319_0001
65. The compound of any one of claims 1-64, wherein R4 is OR14.
66. The compound of any one of claims 1-64, wherein R4 is halogen.
67. The compound of any one of claims 1-64, wherein R4 is alkyl.
68. The compound of any one of claims 1-39, wherein R3 is alkyl.
69. The compound of any one of claims 62-64, wherein x is 0.
70. The compound of any one of claims 62-67, wherein x is 1.
71. The compound of any one of claims 62-67, wherein x is 2.
72. The compound of any one of claims 62-67, wherein x is 3.
73. The compound of any one of claims 1-30, wherein the compound is of Formula:
Figure imgf000319_0002
or a pharmaceutically acceptable salt thereof.
74. The compound of any one of claim 73, wherein the compound is of Formula:
Figure imgf000320_0001
or a pharmaceutically acceptable salt thereof.
75. The compound of any one of claim 73, wherein the compound is of Formula:
Figure imgf000320_0002
or a pharmaceutically acceptable salt thereof.
76. The compound of any one of claims 73-75, wherein R17 is selected from:
Figure imgf000320_0003
77. The compound of any one of claims 73-75, wherein R is selected from:
Figure imgf000320_0004
78. The compound of any one of claims 73-75, wherein R17 is selected from:
Figure imgf000320_0005
79. The compound of any one of claims 73-75, wherein R17 is selected from:
Figure imgf000321_0001
80. The compound of any one of claim 73, wherein the compound is of Formula:
Figure imgf000321_0002
or a pharmaceutically acceptable salt thereof.
81. The compound of any one of claim 73, wherein the compound is of Formula:
Figure imgf000321_0003
or a pharmaceutically acceptable salt thereof.
82. The compound of claim 80 or 81, wherein R18 is cycloalkyl, each of which R18 is substituted with at least one R2 group, and wherein each R18 is optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R4 and R5.
83. The compound of claim 80 or 81 , wherein R18 is heterocycle, each of which R18 is substituted with at least one R2 group, and wherein each R18 is optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R4 and R5.
84. The compound of claim 80 or 81 , wherein R18 is aryl, each of which R18 is substituted with at least one R2 group, and wherein each R18 is optionally substituted with 1 , 2, 3, or 4 substituents independently selected from R4 and R5.
85. A compound selected from:
Figure imgf000322_0001
Figure imgf000323_0001
Figure imgf000324_0001
Figure imgf000325_0001
or a pharmaceutically acceptable salt thereof.
86. A compound selected from:
Figure imgf000325_0002
Figure imgf000326_0001
or a pharmaceutically acceptable salt thereof.
87. A compound selected from:
Figure imgf000327_0001
or a pharmaceutically acceptable salt thereof.
88. The compound of claim 87 of structure:
Figure imgf000327_0002
or a pharmaceutically acceptable salt thereof.
89. The compound of claim 87 of structure:
Figure imgf000328_0001
or a pharmaceutically acceptable salt thereof
90. The compound of claim 87 of structure:
Figure imgf000328_0002
or a pharmaceutically acceptable salt thereof.
91. The compound of claim 87 of structure:
Figure imgf000328_0003
or a pharmaceutically acceptable salt thereof.
92. The compound of claim 87 of structure:
Figure imgf000329_0001
or a pharmaceutically acceptable salt thereof
93. The compound of claim 87 of structure:
Figure imgf000329_0002
or a pharmaceutically acceptable salt thereof.
94. The compound of claim 87 of structure:
Figure imgf000329_0003
or a pharmaceutically acceptable salt thereof.
95. The compound of claim 87 of structure:
Figure imgf000330_0001
or a pharmaceutically acceptable salt thereof.
96. The compound of claim 87 of structure:
Figure imgf000330_0002
or a pharmaceutically acceptable salt thereof
97. The compound of claim 87 of structure:
Figure imgf000330_0003
or a pharmaceutically acceptable salt thereof
98. The compound of claim 87 of structure:
Figure imgf000331_0001
or a pharmaceutically acceptable salt thereof
99. A method for the treatment of a disorder associated with abnormal cellular proliferation comprising administering an effective amount to a host in need thereof of a compound of any one of claims 1-98, optionally in a pharmaceutically acceptable carrier.
100. The method of claim 99, wherein the host is a human.
101. The method of claim 99 or 100, wherein the disorder is an inflammatory disorder.
102. The method of claim 99 or 100, wherein the disorder is a fibrotic disorder.
103. The method of claim 99 or 100, wherein the disorder is an autoimmune disorder.
104. The method of claim 99 or 100, wherein the disorder is a tumor.
105. The method of claim 99 or 100, wherein the disorder is a cancer.
106. The method of claim 99 or 100, wherein the disorder is rheumatoid arthritis.
107. A method of reducing the effect of chemotherapy on healthy cells in a human being treated for cancer or abnormal cell proliferation, wherein said healthy cells are hematopoietic stem cells or hematopoietic progenitor cells, the method comprising administering to the human an effective amount of a compound of any one of claims 1- 98, optionally in a pharmaceutically acceptable carrier.
108. A pharmaceutical composition comprising a compound of any one of claims 1- 98 or pharmaceutically acceptable salt thereof in a pharmaceutically acceptable carrier.
109. The pharmaceutical composition of claim 108 for treating a disorder associated with abnormal cellular proliferation.
110. The pharmaceutical composition of claim 109. wherein the disorder is an inflammatory disorder.
111. The pharmaceutical composition of claim 109, wherein the disorder is a fibrotic disorder.
112. The pharmaceutical composition of claim 109, wherein the disorder is an autoimmune disorder.
113. The pharmaceutical composition of claim 109, wherein the disorder is a tumor.
114. The pharmaceutical composition of claim 109, wherein the disorder is a cancer.
115. The pharmaceutical composition of claim 109, wherein the disorder is rheumatoid arthritis.
116. The pharmaceutical composition of claim 108 for reducing the effect of chemotherapy on healthy cells in a human being treated for cancer or abnormal cell proliferation, wherein said healthy cells are hematopoietic stem cells or hematopoietic progenitor cells.
117. A compound for use in the manufacture of a medicament to treat a disorder associated with abnormal cellular proliferation, wherein the compound is selected from any of claims 1-98 or a pharmaceutically acceptable salt thereof.
118. The compound of claim 117, wherein the disorder is an inflammatory disorder.
119. The compound of claim 117, wherein the disorder is a fibrotic disorder.
120. The compound of claim 117, wherein the disorder is an autoimmune disorder.
121 The compound of claim 117, wherein the disorder is a tumor.
122 The compound of claim 117, wherein the disorder is a cancer.
123. The compound of claim 117, wherein the disorder is rheumatoid arthritis.
124. A compound for use in the manufacture of a medicament to reduce the effect of chemotherapy on healthy cells in a human being treated for cancer or abnormal cell proliferation, wherein said healthy cells are hematopoietic stem cells or hematopoietic progenitor cells, wherein the compound is selected from any of claims 1-98 or a pharmaceutically acceptable salt thereof.
125. Use of a compound in the treatment of a disorder associated with abnormal cellular proliferation, wherein the compound is selected from any of claims 1-98 or a pharmaceutically acceptable salt thereof.
126. The use of claim 125, wherein the disorder is an inflammatory disorder.
127. The use of claim 125, wherein the disorder is a fibrotic disorder.
128. The use of claim 125, wherein the disorder is an autoimmune disorder.
129. The use of claim 125, wherein the disorder is a tumor.
130. The use of claim 125, wherein the disorder is a cancer.
131. The use of claim 125, wherein the disorder is rheumatoid arthritis.
132. Use of a compound in reducing the effect of chemotherapy on healthy cells in a human being treated for cancer or abnormal cell proliferation, wherein said healthy cells are hematopoietic stem cells or hematopoietic progenitor cells, wherein the compound is selected from any of claims 1-98 or a pharmaceutically acceptable salt thereof.
PCT/US2020/026262 2019-04-01 2020-04-01 Cell cycle inhibiting compounds for the treatment of medical disorders WO2020206034A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201962827703P 2019-04-01 2019-04-01
US201962827692P 2019-04-01 2019-04-01
US62/827,692 2019-04-01
US62/827,703 2019-04-01
US201962870351P 2019-07-03 2019-07-03
US62/870,351 2019-07-03

Publications (1)

Publication Number Publication Date
WO2020206034A1 true WO2020206034A1 (en) 2020-10-08

Family

ID=72667500

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2020/026262 WO2020206034A1 (en) 2019-04-01 2020-04-01 Cell cycle inhibiting compounds for the treatment of medical disorders

Country Status (1)

Country Link
WO (1) WO2020206034A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150018348A1 (en) * 2012-03-29 2015-01-15 G1 Therapeutics, Inc. Lactam Kinase Inhibitors
WO2018005860A1 (en) * 2016-07-01 2018-01-04 G1 Therapeutics, Inc. Pyrimidine-based antiproliferative agents
WO2018005863A1 (en) * 2016-07-01 2018-01-04 G1 Therapeutics, Inc. Pyrimidine-based compounds for the treatment of cancer

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150018348A1 (en) * 2012-03-29 2015-01-15 G1 Therapeutics, Inc. Lactam Kinase Inhibitors
WO2018005860A1 (en) * 2016-07-01 2018-01-04 G1 Therapeutics, Inc. Pyrimidine-based antiproliferative agents
WO2018005863A1 (en) * 2016-07-01 2018-01-04 G1 Therapeutics, Inc. Pyrimidine-based compounds for the treatment of cancer

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BISI ET AL.: "Preclinical Characterization of G1T28: A Novel CDK4/6 Inhibitor for Reduction of Chemotherapy-Induced Myelosuppression", MOL CANCER THER, vol. 15, no. 5, 2016, pages 783 - 793, XP055457141, DOI: 10.1158/1535-7163.MCT-15-0775 *

Similar Documents

Publication Publication Date Title
US10654831B2 (en) Antiproliferative pyrimidine-based compounds
US10618905B2 (en) Pyrimidine-based compounds for the treatment of cancer
US20190135811A1 (en) Pyrimidine-based antiproliferative agents
US20210198256A1 (en) Compounds for the degradation of brd9 or mth1
US11357779B2 (en) G1T38 superior dosage regimes
EP3735247A1 (en) Heterocyclic compounds for the treatment of abnormal cellular proliferation
US11643416B2 (en) Substituted 1′,2′-dihydro-3′H-spiro[cyclohexane-1,4′-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,2,4]triazin]-3′-ones as cyclin-dependent kinase inhibitors
WO2019222521A1 (en) Cdk inhibitors for the treatment of neoplastic disorders
JP2020526492A (en) Form of G1T38 and its manufacturing method
US20240018118A1 (en) Tricyclic compounds to degrade neosubstrates for medical therapy
CA3124574A1 (en) Cyclin-dependent kinase inhibitors
WO2020206035A1 (en) Treatment of cdk4/6 inhibitor resistant neoplastic disorders
WO2020206034A1 (en) Cell cycle inhibiting compounds for the treatment of medical disorders
WO2023249974A2 (en) Cyclin-dependent kinase 2 inhibitors for medical treatment

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 20784154

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20784154

Country of ref document: EP

Kind code of ref document: A1