WO2010045451A1 - Pyrrolopyrimidine compounds - Google Patents

Pyrrolopyrimidine compounds Download PDF

Info

Publication number
WO2010045451A1
WO2010045451A1 PCT/US2009/060830 US2009060830W WO2010045451A1 WO 2010045451 A1 WO2010045451 A1 WO 2010045451A1 US 2009060830 W US2009060830 W US 2009060830W WO 2010045451 A1 WO2010045451 A1 WO 2010045451A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
mmol
methyloxy
alkyl
dihydro
Prior art date
Application number
PCT/US2009/060830
Other languages
French (fr)
Inventor
Kirk Stevens
John Brad Shotwell
Aniko Redman
Joseph Wendell Wilson
Huangshu Lei
Roseanne Gerding
Samarjit Patnaik
Original Assignee
Glaxosmithkline Llc
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 Glaxosmithkline Llc filed Critical Glaxosmithkline Llc
Publication of WO2010045451A1 publication Critical patent/WO2010045451A1/en

Links

Classifications

    • 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
    • C07D487/02Heterocyclic 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 two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the receptor is activated by the ligands IGF-1 and IGF-2, which are mitogenic proteins that signal through the IGF-1 R and IR in an endocrine, paracrine or autocrine manner. Activation of the IGF-1 receptor elicits cellular responses which include cellular proliferation and protection of cells from apoptosis.
  • IGF-1 R Overexpression of IGF-1 R leads to malignant transformation of cultured cells, while downregulation can reverse the transformed phenotype of tumor cells and potentially render them susceptible to apoptosis.
  • IGF-1 R is a hetero-tetrameric, transmembrane, cell surface receptor.
  • An IGF-1 binding domain is part of the extracellular alpha- chain of IGF-1 R, whereas the intracellular beta-chain contains the tyrosine kinase domain.
  • Three tyrosine residues represent autophosphorylation sites, specifically Tyr1 131 , Tyr1135, and Tyr1136, within the activation loop of the IGF-1 R beta catalytic domain (Li, W et al., J. Biol. Chem. (2006) 281 (33):23785-23791 ).
  • lymphoblastic T cell leukemia including lymphoblastic T cell leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, hairy-cell leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic neutrophilic leukemia, acute lymphoblastic T cell leukemia, plasmacytoma, immunoblastic large cell leukemia, mantle cell leukemia, multiple myeloma, megakaryoblastic leukemia, acute megakaryocyte leukemia, promyelocytic leukemia, erythroleukemia, malignant lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, lymphoblastic T cell lymphoma, Burkitt's lymphoma, follicular lymphoma, myelogenous leukemia, chronic lymphocytic leukemia, hairy-cell leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia
  • the present invention provides a compound of formula (I):
  • -alkylene C i-c6-C(O)NH 2 one or two of X 1 , X 2 , and X 3 are independently selected from N, O, and S, while the remaining one or two of X 1 , X 2 , and X 3 are C; m is 0 or 1 ; R 1 is selected from H, halo, alkyl C i-c6, -alkyleneci-cs-haloi-s, and -O-alkyl C i-c6when X 1 is
  • R 1 is absent;
  • R 2 is selected from H, halo, alkyl C i-c6, -alkyleneci-cs-haloi-s, and -O-alkyl C i-c6when X 2 is
  • R 7 is selected from alkyl C i-c6, -O-alkyl C i-c6, halo, -NR 22 R 23 , and -O-alkylene C i-c6-halo 1-3 ;
  • each R 14 is independently selected from H, OH, halo, alkyl C i-c6, -0-alkyl C i-c6, -cyclopropyl, -C(O)-alkyl C i-c6, SO 2 -alkyl C i-c6, -(CH 2 )i-4-halo ,and -(CH 2 ) 1-4 -SO 2 -alkylci-c 6 ; -C(O )-alkylene C0 -c 6 -N R 22 R 23 , -NR 22 R 23 ; or
  • each R 23 is selected from H and alkyl C i-c6; or
  • R 22 and R 23 combine to form a four, five, or six membered, heterocyclic ring containing the N atom to which they are attached and optionally an additional heteroatom selected from N and O, wherein the ring is optionally substituted with -OH or -alkyld-ce-
  • a compound of formula I is provided as described in any one of the examples.
  • a pharmaceutically acceptable derivative of the compound of formula I described in any one of the examples is provided.
  • the invention provides a method for treating a susceptible neoplasm in a mammal in need thereof, comprising: administering to the mammal, a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable derivative thereof.
  • the invention provides a method for treating a condition selected from breast cancer, sarcomas, lung cancer (including non-small cell lung carcinoma), prostate cancer, colorectal cancer, renal cancer, pancreatic cancer, hematologic cancers (including multiple myeloma), neuroblastomas, gliomas, head and neck cancer, thyroid cancer, hepatocarcinoma, ovarian cancer, vulval cancer, cervical cancer, endometrial cancer, testicular cancer, bladder cancer, esophageal cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, gastrointestinal stromal tumor and skin cancer (including melanoma) in a mammal in need thereof, comprising: administering to the mammal, a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable derivative thereof.
  • the invention provides a method for treating a condition selected from breast cancer, sarcoma, lung cancer, non-small cell lung carcinoma, prostate cancer, colorectal cancer, pancreatic cancer, hematologic cancers, multiple myeloma, head and neck cancer or ovarian cancer in a mammal in need thereof, comprising: administering to the mammal, a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable derivative thereof.
  • the invention provides a compound of
  • Formula I or a pharmaceutically acceptable derivative thereof for use in the treatment of a condition mediated by inappropriate activity of at least one IGF-1 R family receptor.
  • the invention provides a compound of Formula I, or a pharmaceutically acceptable derivative thereof for use in the treatment of a susceptible neoplasm in a mammal in need thereof.
  • the invention provides a compound of Formula I, or a pharmaceutically acceptable derivative thereof for use in the treatment of a condition selected from breast cancer, sarcomas, lung cancer (including non-small cell lung carcinoma), prostate cancer, colorectal cancer, renal cancer, pancreatic cancer, hematologic cancers (including multiple myeloma), neuroblastomas, gliomas, head and neck cancer, thyroid cancer, hepatocarcinoma, ovarian cancer, vulval cancer, cervical cancer, endometrial cancer, testicular cancer, bladder cancer, esophageal cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, gastrointestinal stromal tumor and skin cancer (including melanoma) in a mammal in need thereof.
  • the invention provides a compound of Formula I, or a pharmaceutically acceptable derivative thereof for use in the treatment of a condition selected from breast cancer, sarcoma, lung cancer, non-small cell lung carcinoma, prostate cancer, colorectal cancer, pancreatic cancer, hematologic cancers, multiple myeloma, head and neck cancer or ovarian cancer in a mammal in need thereof.
  • the invention provides the use of a compound of formula I, or pharmaceutically acceptable derivative thereof in the manufacture of a medicament for use in the treatment of a condition mediated by inappropriate activity of at least one IGF-1 R family receptor.
  • the invention provides the use of a compound of formula I, or pharmaceutically acceptable derivative thereof in the manufacture of a medicament for use in the treatment of a susceptible neoplasm in a mammal in need thereof.
  • the invention provides the use of a compound of formula I, or pharmaceutically acceptable derivative thereof in the manufacture of a medicament for use in the treatment of a condition selected from breast cancer, sarcomas, lung cancer (including non-small cell lung carcinoma), prostate cancer, colorectal cancer, renal cancer, pancreatic cancer, hematologic cancers (including multiple myeloma), neuroblastomas, gliomas, head and neck cancer, thyroid cancer, hepatocarcinoma, ovarian cancer, vulval cancer, cervical cancer, endometrial cancer, testicular cancer, bladder cancer, esophageal cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, gastrointestinal stromal tumor and skin cancer (including melanoma) in a mammal in need thereof.
  • alkyl refers to straight or branched hydrocarbon chains containing from 1 to 6 carbon atoms, unless a different number of atoms is specified.
  • alkyl as used herein include, but are not limited to, methyl, ethyl, n-propyl, n-butyl, n-pentyl, isobutyl, isopropyl, and tert-butyl.
  • alkylene as used herein include, but are not limited to, methylene, ethylene, propylene, butylene, and isobutylene.
  • Alkyl also includes substituted alkyl.
  • alkyl (and alkylene) groups may be optionally substituted one or more times with a halogen or hydroxyl.
  • alkyl may include for example, trifluoromethyl and trifluoroethyl, among other halogenated alkyls, and hydroxymethyl and other hydroxylated alkyls when specified.
  • alkenyl refers to straight or branched hydrocarbon chains containing from 2 to 6 carbon atoms, unless a different number of atoms is specified, and at least one and up to three carbon-carbon double bonds.
  • alkenyl as used herein include, but are not limited to ethenyl and propenyl.
  • alkenylene as used herein include, but are not limited to, ethenylene, propenylene and butenylene.
  • Alkenyl (and “alkenylene”) also may include substituted alkenyl. The alkenyl groups may optionally be substituted one or more times with a halogen or hydroxyl, as specified.
  • alkynyl refers to straight or branched hydrocarbon chains containing from 2 to 6 carbon atoms, unless a different number of atoms is specified, and at least one and up to three carbon-carbon triple bonds.
  • alkynyl as used herein include, but are not limited to ethynyl and propynyl.
  • Alkynyl may also include substituted alkynyl. The alkynyl groups may optionally be substituted one or more times with a halogen or hydroxyl, as specified.
  • cycloalkyl refers to a saturated monocyclic carbocyclic ring having from 3 to 6 carbon atoms, unless a different number of atoms is specified.
  • Cycloalkyl includes by way of example cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
  • Cycloalkyl also includes substituted cycloalkyl.
  • the cycloalkyl may optionally be substituted on any available carbon with one or more substituents selected from the group consisting of alkoxy, halo, and haloalkyl, e.g., perfluoroalkyl.
  • halo or “halogen” refers to fluoro, chloro, bromo and iodo.
  • alkoxy refers to the group -O-alkyl, where alkyl is as defined above. Examples of “alkoxy” as used herein include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, and t-butoxy. "Alkoxy” also includes substituted alkoxy. The alkoxy groups may be optionally substituted one or more times with a halogen.
  • aryl refers to monocyclic carbocyclic groups and fused bicyclic carbocyclic groups having from 6 to 10 carbon atoms, unless a different number of atoms is specified, and having at least one aromatic ring.
  • aryl groups include but are not limited to phenyl and naphthyl.
  • One particular aryl group according to the invention is phenyl.
  • heterocycle refers to a mono- or poly-cyclic ring system containing optionally one or more degrees of unsaturation and also containing one or more heteroatoms.
  • Heteroatoms may include N, O, and/or S. Preferred heteroatoms are N and O, particularly N.
  • the heterocycle is three to ten- membered and is either saturated or has one or more degrees of unsaturation.
  • Heterocycles may be optionally fused to one or more of another heterocyclic ring, heteroaryl ring, aryl ring, or cycloalkyl ring.
  • heterocyclic groups include, e.g. indole, indoline, isoquinoline.
  • pharmaceutically acceptable derivative refers to salts and solvates of the selected compound.
  • R 4a and R 4b are each independently selected from H, alkyl C i-c 6 , -OH, -alkyleneci-c ⁇ -OH, -alkylene C i-c 6 -phenyl (optionally substituted with a halo), and -alkylene C i-c6-C(O)NH 2 ; one or two of X 1 , X 2 , and X 3 are independently selected from N, O, and S, while the remaining one or two of X 1 , X 2 , and X 3 are C; m is O or 1 ; R 1 is selected from H, halo, alkyl C i-c 6 , -alkyleneci-cs-halOi-s, and -O-alkyl C i-c 6 when X 1 is
  • R 2 is selected from H, halo, alkyl C i-c 6 , -alkyleneci-cs-halOi-s, and -O-alkyl C i-c 6 when X 2 is C, otherwise R 2 is absent;
  • R 3 is selected from H, halo, alkyl C i-c 6 , -alkyleneci-cs-haloi-s, and -O-alkyl C i-c 6 when X 3 is
  • R 7 is selected from alkyl C i-c6, -0-alkyl C i-c6, halo, -NR 22 R 23 , and -O-alkylene C i-c6-halo 1-3 ;
  • R 8 is selected from H, halo, and alkyl C i-c6; one of R 9 and R 10 is selected from -alk ⁇ yylleenneci-c6-SO 2 -alkyl C i-c6,
  • Het1 and Het2 are each independently a five or six membered heterocyclic ring having an N atom and optionally one or two additional heteroatoms selected from N and O, and each R 14 is independently selected from H, OH, halo, alkyl C i-c6, -0-alkyl C i-c6, -cyclopropyl, -C(O)-alkyl C i-c6, SO 2 -alkyl C i-c6, -(CH 2 )i-4-halo ,and -(CH 2 ) 1-4 -SO 2 -alkylci-c 6 ; -C(O )-alkylene C o-c 6 -N R 22 R 23 , -NR 22 R 23 ; or R 9 and R 10 , together with the atoms to which they are attached form a five, six, or seven- membered heterocyclic ring containing one or two N atom and the remainder C atoms, wherein at least one N N
  • R 15 is selected from H, -alkyl C i-c4, -alkylene C i-c4-halo,
  • each R 19 is independently selected from H and alkyl C i-c6; each R 22 is selected from H, alkyl C i-c6, -O-alkyl C i-c6, -alkyleneci-c ⁇ -O-alkylci-ce; and,
  • each R 23 is selected from H and alkyl C i-c6; or
  • R 22 and R 23 combine to form a four, five, or six membered, heterocyclic ring containing the N atom to which they are attached and optionally an additional heteroatom selected from N and O, wherein the ring is optionally substituted with -OH or -alkyb-c ⁇ .
  • R 7 is -0-alkyl C i-c6 and the remaining substituents are as described above in the main embodiment.
  • R 7 is -O-alkyl C i-c6, R 10 is H, and the remaining substituents are as described above in the main embodiment.
  • R 7 is -O-alkyl C i-c6, m is 0, X1 is S, X2 and X3 are C, and the remaining substituents are as described above in the main embodiment.
  • R 7 is -O-alkyl C i-c6; m is 0, X1 is O, X2 and X3 are C, and the remaining substituents are as described above in the main embodiment.
  • R 7 is -O-alkyl C i-c6; m is 0, X1 is C, X2 is S, X3 is C, and the remaining substituents are as described above in the main embodiment.
  • R 7 is -O-alkyl C i-c6; m is 1 , X1 is N, X2 and X3 are C, and the remaining substituents are as described above in the main embodiment.
  • the substituents are as described in the above embodiments, respectively, wherein R 7 is -O-methyl.
  • the compound is of formula (Ia):
  • the compound is of formula (Ib): wherein the substituents are as described above in the main embodiment.
  • the compound is of formula (Ic):
  • the compound is of formula (Id):
  • R 10 is H and R 9 is selected from
  • R 9 and R 10 together with the atoms to which they are attached form a five, six, or seven-membered heterocyclic ring containing one or two N atom and the remainder C atoms, wherein at least one N atom is substituted with R 15 , and the C atoms of the heterocyclic ring are optionally substituted with one or more groups selected from (R 19 )i- 2 , and the remaining substituents are as described above in the main embodiment.
  • R 9 and R 10 together with the atoms to which they are attached form a five, six, or seven-membered heterocyclic ring containing one or two N atom and the remainder C atoms, wherein at least one N atom is substituted with R 15 , and the C atoms of the heterocyclic ring are optionally substituted with one or more groups selected from (R 19 )i -2 , R 7 is -O-alkyl C i-c6, and the remaining substituents are as described above in the main embodiment.
  • R 9 and R 10 together with the atoms to which they are attached form a five or six-membered heterocyclic ring selected from
  • R 9 and R 10 together with the atoms to which they are attached form a five or six-membered heterocyclic ring selected from , R 7 is -O-alkyld-ce, and the remaining substituents are as described above in the main embodiment.
  • substitution at the R 7 position is favorable and has generally been found to boost IGF1 R potency in comparison to otherwise similar compounds that are unsubstituted at the R 7 position. Further, substitution at the R 7 position has been found to increase selectivity over other kinases, for instance the Aurora A and Aurora B kinase, which is often desirable.
  • the pharmaceutically acceptable salts of the compounds of formula (I) include conventional salts formed from pharmaceutically acceptable (i.e., non-toxic) inorganic or organic acids or bases as well as quaternary ammonium salts.
  • Representative salts include the following: acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, calcium edetate, camsylate, carbonate, chloride, clavulanate, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, monopotassium maleate, mucate, napsylate, nitrate, N-methylglucamine, oxalate,
  • Certain compounds of formula (I) may exist in stereoisomeric forms (e.g. they may contain one or more asymmetric carbon atoms or may exhibit cis-trans isomerism).
  • the individual stereoisomers (enantiomers and diastereomers) and mixtures of these are included within the scope of the present invention.
  • the present invention also covers the individual isomers of the compounds represented by formula (I) as mixtures with isomers thereof in which one or more chiral centres are inverted. It is understood that compounds of formula (I) may exist in tautomeric forms other than that shown in the formula and these are also included within the scope of the present invention.
  • the compounds of formula (I) are intended for use in pharmaceutical compositions it will readily be understood that they are each preferably provided in substantially pure form, for example at least 60% pure, more suitably at least 75% pure and preferably at least 85%, especially at least 98% pure (% are on a weight for weight basis). Impure preparations of the compounds may be used for preparing the more pure forms used in the pharmaceutical compositions.
  • the compounds of the present invention are inhibitors of one or more IGFR family receptors.
  • IGFR inhibitor is meant a compound which exhibits a pICso of greater than 5.5 against at least one IGFR family receptor in the IGFR inhibition enzyme assay described below (TR-FRET) and/or an IC 5 O of below about 1.0 ⁇ M potency against IGFR cellular autophosphorylation and/or in cell proliferation of a cell line that is dependent upon IGF signaling (e.g. Colo205, NCI-H929) in at least one of the cellular assay described below.
  • IGF signaling e.g. Colo205, NCI-H929
  • IGFR inhibitor refers to a compound which exhibits a plC 50 of greater than 7.6 against at least one IGFR family receptor in the IGFR inhibition enzyme assay described below.
  • IGFR inhibitor refers to a compound which exhibits an IC 50 of below about 250 nM potency against IGFR cellular autophosphorylation and/or in cell proliferation of a cell line that is dependent upon IGF signaling (e.g. Colo205, NCI-H929) in at least one of the cellular assay described below.
  • the present invention is not limited to compounds of formula (I) which are selective for IGFR family receptor kinases; rather, the present invention expressly contemplates compounds of formula (I) which may also possess activity against receptors in addition to the IGFR family receptors.
  • the compounds of formula (I) are selective for insulin receptor (IR) family kinases.
  • IR insulin receptor
  • Several compounds of the present invention also possess activity against, for instance, one or more of the Jnk1 , Jnk2, and Jnk3, anaplastic lymphoma kinase (ALK), and insulin related receptor (IRR).
  • the present invention further provides compounds of formula (I) for use in medical therapy in a mammal, e.g. a human.
  • the present invention provides compounds of formula (I) for use in the treatment of a condition mediated by at least one IGFR family receptor in a mammal, and, advantageously, conditions mediated by inappropriate activity of one or more IGFR family receptor in a mammal.
  • the inappropriate IGFR family receptor activity referred to herein is any IGFR receptor activity that deviates from the normal IGFR family receptor activity expected in a particular mammalian subject.
  • Inappropriate IGFR family receptor activity may take the form of, for instance, an abnormal increase in activity, or an aberration in the timing and/or control of IGFR family receptor activity.
  • Such inappropriate activity may result then, for example, from overexpression or mutation of the protein kinase or ligand leading to inappropriate or uncontrolled activation of the receptor.
  • unwanted IGFR family receptor activity may reside in an abnormal source, such as a malignancy. That is, the level of IGFR family activity does not have to be abnormal to be considered inappropriate, rather the activity derives from an abnormal source.
  • the compounds of formula (I) and salts and solvates thereof are believed to have anticancer and antitumor activity as a result of inhibition of one or more IGFR family receptor and its effect on selected cell lines whose growth is dependent on IGFR family activity.
  • the present invention provides compounds of formula (I) for use in the treatment of a susceptible neoplasm.
  • "Susceptible neoplasm” as used herein refers to neoplasms which are susceptible to treatment with a IGFR inhibitor. Neoplasms which have been associated with inappropriate activity of one or more IGFR family receptors and are therefor susceptible to treatment with a IGFR inhibitor are known in the art, and include both primary and metastatic tumors and cancers.
  • susceptible neoplasms within the scope of the present invention include but are not limited to breast cancer, sarcomas, lung cancer (including non-small cell lung carcinoma), prostate cancer, colorectal cancer, renal cancer, pancreatic cancer, hematologic cancers (including multiple myeloma), neuroblastomas, gliomas, head and neck cancer, thyroid cancer, hepatocarcinoma, ovarian cancer, vulval cancer, cervical cancer, endometrial cancer, testicular cancer, bladder cancer, esophageal cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, gastrointestinal stromal tumor, and skin cancer (including melanoma).
  • susceptible neoplasms may be selected from breast cancer, sarcoma, lung cancer, non-small cell lung carcinoma, prostate cancer, colorectal cancer, pancreatic cancer, hematologic cancers, multiple myeloma, head and neck cancer, and ovarian cancer.
  • the present invention provides methods for the treatment of several conditions in a mammal in need thereof, all of which comprise the step of administering a therapeutically effective amount of a compound of formula (I).
  • the mammal in need of treatment with a compound of the present invention is advantageously a human.
  • treatment refers to alleviating the specified condition, eliminating or reducing the symptoms of the condition, slowing or eliminating the progression of the condition and preventing or delaying the reoccurrance of the condition in a previously afflicted subject.
  • the term "therapeutically effective amount” means an amount of a compound of formula (I) which is sufficient, in the subject to which it is administered, to elicit the biological or medical response of a cell culture, tissue, system, mammal (including human) that is being sought, for instance, by a researcher or clinician.
  • the term also includes within its scope amounts effective to enhance normal physiological function.
  • a therapeutically effective amount of a compound of formula (I) for the treatment of a condition mediated by at least one IGFR family receptor is an amount sufficient to treat the condition in the subject.
  • a therapeutically effective amount of a compound of formula (I) for the treatment of a susceptible neoplasm is an amount sufficient to treat the susceptible neoplasm in the subject.
  • a therapeutically effective amount of a compound of formula (I) is an amount sufficient to regulate, modulate, bind or inhibit at least one IGFR family receptor.
  • the precise therapeutically effective amount of the compounds of formula (I) will depend on a number of factors including, but not limited to, the age and weight of the subject being treated, the precise condition requiring treatment and its severity, the nature of the formulation, and the route of administration, and will ultimately be at the discretion of the attendant physcian or veternarian.
  • the compound of formula (I) will be given for treatment in the range of 0.1 to 200 mg/kg body weight of recipient (mammal) per day and more usually in the range of 1 to 100 mg/kg body weight per day.
  • Acceptable daily dosages may be from about 0.1 to about 2000 mg/day, and preferably from about 0.1 to about 100 mg/day.
  • the actual amount per day would usually be from 5 to 700 mg and this amount may be given in a single dose per day or more usually in a number (such as two, three, four, five or six) of sub-doses per day or, alternatively, on an alternative dosing schedule such as weekly or monthly, such that the total daily dose is the same.
  • a therapeutically effective amount of a salt or solvate may be determined as a proportion of the therapeutically effective amount of the compound of formula (I) per se. It is envisaged that similar dosages would be appropriate for treatment of the other conditions referred to above.
  • the present invention also provides the use of a compound of formula (I) for the preparation of a medicament for the treatment of condition mediated by at least one IGFR family receptor in a mammal (e.g., a human) in need thereof.
  • the present invention further provides the use of a compound of formula (I) for the preparation of a medicament for the treatment of a susceptible neoplasm in a mammal.
  • a therapeutically effective amount of a compound of formula (I) may be administered as the raw chemical, it is typically presented as the active ingredient of a pharmaceutical composition or formulation.
  • the invention further provides a pharmaceutical composition comprising a compound of the formula (I).
  • the pharmaceutical composition may further comprise one or more pharmaceutically acceptable carriers, diluents, and/or excipients.
  • the carrier(s), diluent(s) and/or excipient(s) must be acceptable in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • a process for the preparation of a pharmaceutical formulation including admixing a compound of the formula (I) with one or more pharmaceutically acceptable carriers, diluents and/or excipients.
  • Pharmaceutical formulations may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) route.
  • Such formulations may be prepared by any method known in the art of pharmacy, for example by bringing into association the active ingredient with the carrier(s) or excipient(s).
  • compositions adapted for oral administration may be presented as discrete units such as capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in-oil liquid emulsions.
  • the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like.
  • Powders are prepared by comminuting the compound to a suitable fine size and mixing with a similarly comminuted pharmaceutical carrier such as an edible carbohydrate, as, for example, starch or mannitol. Flavoring, preservative, dispersing and coloring agent can also be present.
  • Capsules are made by preparing a powder mixture, as described above, and filling formed gelatin sheaths.
  • Glidants and lubricants such as colloidal silica, talc, magnesium stearate, calcium stearate or solid polyethylene glycol can be added to the powder mixture before the filling operation.
  • a disintegrating or solubilizing agent such as agar-agar, calcium carbonate or sodium carbonate can also be added to improve the availability of the medicament when the capsule is ingested.
  • 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, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, 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. Tablets are formulated, for example, by preparing a powder mixture, granulating or slugging, adding a lubricant and disintegrant and pressing into tablets.
  • a powder mixture is prepared by mixing the compound, suitably comminuted, with a diluent or base as described above, and optionally, with a binder such as carboxymethylcellulose, an aliginate, gelatin, or polyvinyl pyrrolidone, a solution retardant such as paraffin, a resorption accelerator such as a quaternary salt and/or an absorption agent such as bentonite, kaolin or dicalcium phosphate.
  • the powder mixture can be granulated by wetting with a binder such as syrup, starch paste, acadia mucilage or solutions of cellulosic or polymeric materials and forcing through a screen.
  • the powder mixture can be run through the tablet machine and the result is imperfectly formed slugs broken into granules.
  • the granules can be lubricated to prevent sticking to the tablet forming dies by means of the addition of stearic acid, a stearate salt, talc or mineral oil.
  • the lubricated mixture is then compressed into tablets.
  • the compounds of the present invention can also be combined with a free flowing inert carrier and compressed into tablets directly without going through the granulating or slugging steps.
  • a clear or opaque protective coating consisting of a sealing coat of shellac, a coating of sugar or polymeric material and a polish coating of wax can be provided. Dyestuffs can be added to these coatings to distinguish different unit dosages.
  • Oral fluids such as solution, syrups and elixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of the compound.
  • Syrups can be prepared by dissolving the compound in a suitably flavored aqueous solution, while elixirs are prepared through the use of a non-toxic alcoholic vehicle.
  • Suspensions can be formulated by dispersing the compound in a non-toxic vehicle.
  • Solubilizers and emulsifiers such as ethoxylated isostearyl alcohols and polyoxy ethylene sorbitol ethers, preservatives, flavor additive such as peppermint oil or natural sweeteners or saccharin or other artificial sweeteners, and the like can also be added.
  • dosage unit formulations for oral administration can be microencapsulated.
  • the formulation can also be prepared to prolong or sustain the release as for example by coating or embedding particulate material in polymers, wax or the like.
  • the compounds of formula (I) may be administered by inhalation, such as by metered dose pressurised aerosols, metered dose inhalers, dry powder inhalers, nebulizers or insufflators.
  • the compound is provided in the form of a dry powder composition.
  • the composition is suitable for inhaled administration and may be incorporated into a plurality of sealed dose containers (e.g. containing the dry powder composition) mounted in a strip or ribbon inside a suitable inhalation device.
  • the container is rupturable or peel-openable on demand and the dose of the dry powder composition may be administered by inhalation via a device such as the DISKUSTM device, marketed by GlaxoSmithKline.
  • the DISKUSTM inhalation device is, for example, described in GB2242134A.
  • the compounds of formula (I) may be formulated into spray compositions for inhalation which may, for example, be formulated as aqueous solutions or suspensions or as aerosols delivered from pressurised packs, such as a metered dose inhaler (MDI), with the use of a suitable liquefied propellant.
  • spray compositions for inhalation which may, for example, be formulated as aqueous solutions or suspensions or as aerosols delivered from pressurised packs, such as a metered dose inhaler (MDI), with the use of a suitable liquefied propellant.
  • MDI metered dose inhaler
  • Aerosol compositions suitable for inhalation can be either a suspension or a solution and generally contain a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof and a suitable propellant such as a fluorocarbon or hydrogen-containing chlorofluorocarbon or mixtures thereof, particularly hydrofluoroalkanes, especially 1 ,1 ,1 ,2-tetrafluoroethane, 1 ,1 ,1 ,2,3,3,3-heptafluoro-n-propane or a mixture thereof.
  • the aerosol composition may optionally contain additional formulation excipients well known in the art such as surfactants e.g. oleic acid, lecithin or an oligolactic acid derivative e.g.
  • the compounds of formula (I) can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles.
  • Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines.
  • the compounds of formula (I) may also be delivered by the use of monoclonal antibodies as individual carriers to which the compound molecules are coupled. The compounds may also be coupled with soluble polymers as targetable drug carriers.
  • Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide -phenol, polyhydroxyethylaspartamidephenol, or polyethyleneoxidepolylysine substituted with palmitoyl residues.
  • the compounds may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels.
  • compositions adapted for transdermal administration may be presented as discrete patches intended to remain in intimate contact with the epidermis of the recipient for a prolonged period of time.
  • the active ingredient may be delivered from the patch by iontophoresis as generally described in Pharmaceutical Research, 3(6), 318 (1986).
  • Pharmaceutical formulations adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils.
  • the formulations are preferably applied as a topical ointment or cream.
  • the active ingredient may be employed with either a paraffinic or a water- miscible ointment base.
  • the active ingredient may be formulated in a cream with an oil-in-water cream base or a water-in-oil base.
  • compositions adapted for topical administrations to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent.
  • compositions adapted for topical administration in the mouth include lozenges, pastilles and mouth washes.
  • Pharmaceutical formulations adapted for rectal administration may be presented as suppositories or as enemas.
  • compositions adapted for nasal administration wherein the carrier is a solid include a coarse powder having a particle size for example in the range 20 to 500 microns which is administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close up to the nose.
  • Suitable formulations wherein the carrier is a liquid, for administration as a nasal spray or as nasal drops, include aqueous or oil solutions of the active ingredient.
  • Fine particle dusts or mists which may be generated by means of various types of metered, dose pressurised aerosols, nebulizers or insufflators.
  • compositions adapted for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations.
  • Pharmaceutical formulations adapted for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation near isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • sterile liquid carrier for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
  • formulations may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavouring agents.
  • a compound of the invention may be employed alone, in combination with one or more other compounds of the invention, or in combination with other therapeutic methods or agents.
  • the compound of the invention may be used alone or in combination with one or more of a chemotherapeutic, a hormonal and/or antibody agent, surgical therapy, and radiotherapy.
  • chemotherapeutic refers to any chemical agent having a therapeutic effect on the subject to which it is administered.
  • “Chemotherapeutic” agents include but are not limited to anti-neoplastic agents, analgesics and anti-emetics.
  • anti-neoplastic agents include both cytostatic and cytotoxic agents.
  • Anti-emetics include but are not limited to 5HT 3 antagonists such as ondansetron, granisetron, and the like; metaclopromide; dexamethasone and neurokinin-1 antagonists.
  • the present invention provides the methods of treatment and uses as described above, which comprise administering a compound of the invention together with at least one chemotherapeutic agent.
  • the chemotherapeutic agent is an anti-neoplastic agent.
  • the present invention provides a pharmaceutical composition as described above further comprising at least one other chemotherapeutic agent, more particularly, the chemotherapeutic agent is an anti-neoplastic agent.
  • the compounds of the invention and at least one additional anti-neoplastic therapy may be employed in combination concomitantly or sequentially in any therapeutically appropriate combination.
  • the administration of a compound of the invention with one or more other anti-neoplastic agents may be in combination in accordance with the invention by administration concomitantly in (1 ) a unitary pharmaceutical composition including both or all compounds or (2) separate pharmaceutical compositions each including one of the compounds.
  • the combination may be administered separately in a sequential manner wherein one anti-neoplastic agent is administered first and the other(s) second or vice versa. Such sequential administration may be close in time or remote in time.
  • chemotherapeutic agents When a compound of the invention is used in combination with a chemotherapeutic agent, the dose of each compound may differ from that when the compound is used alone. Appropriate doses will be readily appreciated by those skilled in the art. The appropriate dose of the compound(s) of the invention and the other therapeutically active agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect, and are within the expertise and discretion of the attendent clinician.
  • chemotherapeutic agents which may be used in combination with a compound of the present invention, are anti-proliferative/anti-neoplastic agents.
  • Anti-neoplastic agents may induce anti-neoplastic effects in a cell-cycle specific manner, i.e., are phase specific and act at a specific phase of the cell cycle, or bind DNA and act in a non cell-cycle specific manner, i.e., are non-cell cycle specific and operate by other mechanisms. Both types of anti-neoplastic agents may be employed in combination with the compounds of the present invention.
  • any chemotherapeutic agent that has activity against a susceptible neoplasm being treated may be utilized in combination with the compounds the invention, provided that the particular agent is clinically compatible with therapy employing a compound of the invention.
  • Typical anti-neoplastic agents useful in the present invention include, but are not limited to: alkylating agents, anti-metabolites, antitumor antibiotics, antimitotic agents, topoisomerase I and Il inhibitors, hormones and hormonal analogues, matrix metalloprotease inhibitors; signal transduction pathway inhibitors including inhibitors of cell growth or growth factor function, angiogenesis inhibitors, and serine/threonine inhibitors; cyclin dependent kinase inhibitors; antisense therapies and immunotherapeutic agents.
  • alkylating agents include but are not limited to: nitrogen mustards such as cyclophosphamides, temozolamide, melphalan, and chlorambucil; oxazaphosphor-ines; alkyl sulfonates such as busulfan; nitrosoureas such as carmustine; triazenes such as dacarbazine; and platinum coordination complexes such as cisplatin, oxapliplatin and carboplatin.
  • nitrogen mustards such as cyclophosphamides, temozolamide, melphalan, and chlorambucil
  • oxazaphosphor-ines alkyl sulfonates
  • alkyl sulfonates such as busulfan
  • nitrosoureas such as carmustine
  • triazenes such as dacarbazine
  • platinum coordination complexes such as cisplatin, oxapliplatin and carboplatin.
  • antimetabolite anti-neoplastic agents include purine and pyrimidine analogues and anti-folate compounds, and more specifically, hydroxyurea, cytosine, arabinoside, raltitrexed, tegafur, fluorouracil (e.g., 5FU), methotrexate, cytarabine, mecaptopurine and thioguanine.
  • purine and pyrimidine analogues and anti-folate compounds and more specifically, hydroxyurea, cytosine, arabinoside, raltitrexed, tegafur, fluorouracil (e.g., 5FU), methotrexate, cytarabine, mecaptopurine and thioguanine.
  • antitumor antibiotic agents include, but are not limited to, actinomycins such as dactinomycin, mitomycin-C, anthracyclins such as daunorubicin, doxorubicin, idarubicin, epirubicin; daunomycin, adriamycin and bleomycins.
  • antimitotic agents include, but are not limited to, diterpenoids, vinca alkaloids, polo-like kinase (PLK) inhibitors and CenpE inhibitors.
  • diterpenoids include, but are not limited to, taxol, taxotere, paclitaxel and its analog docetaxel.
  • vinca alkaloids include, but are not limited to, vinblastine, vincristine, vindesine and vinorelbine. PLK inhibitors are discussed further below.
  • topoisomerase I inhibitors examples include camptothecins, such as amsacrine, irinotecan, topotecan, and the various optical forms of 7-(4-methylpiperazino- methylene)-10,11-ethylenedioxy-20-camptothecin.
  • topoisomerase Il inhibitors include epipodophyllotoxins, such as etoposide and teniposide.
  • hormones and hormonal analogues believed to be useful in the treatment of neoplasms include, but are not limited to: antiestrogens, such as tamoxifen, toremifene, raloxifene, fulvestrant, iodoxyfene and droloxifene; anti-androgens; such as flutamide, nilutamide, bicalutamide and cyproterone acetate; adrenocorticosteroids such as prednisone and prednisolone; aminoglutethimide and other aromatase inhibitors such as anastrozole, letrazole, vorazole, and exemestane; progestrins such as megestrol acetate; 5 ⁇ -reductase inhibitors such as finasteride and dutasteride; and gonadotropin-releasing hormones (GnRH) and analogues thereof, such as Leutinizing Hormone-releasing hormone
  • MMP matrix metalloproteinases
  • Signal transduction pathway inhibitors useful in the present invention include, but are not limited to, inhibitors of receptor tyrosine kinases, non-receptor tyrosine kinases, SH2/SH3 domain blockers, serine/threonine kinases, phosphatidyl inositol-3-OH kinases, myo-inositol signaling, and Ras oncogenes (e.g. farnesyltransferase, geranyl- geranyl transferase, and CAAX proteases as well as anti-sense oligonucleotides, ribozymes and immunotherapy).
  • Ras oncogenes e.g. farnesyltransferase, geranyl- geranyl transferase, and CAAX proteases as well as anti-sense oligonucleotides, ribozymes and immunotherapy.
  • protein tyrosine kinases catalyse the phosphorylation of specific tyrosyl residues in various proteins involved in the regulation of cell growth.
  • protein tyrosine kinases can be broadly classified as receptor or non-receptor kinases.
  • receptor tyrosine kinases also known as "growth factor receptor inhibitors", in addition to IGF-1 R inhibitors, include but are not limited to inhibitors of: epidermal growth factor family receptors (EGFR, ErbB2, and ErbB4); platelet derived growth factor receptors (PDGFRs), vascular endothelial growth factor receptors (VEGFRs), tyrosine kinase with immunoglobulin-like and epidermal growth factor homology domains (TIE-2), macrophage colony stimulating factor (c-fms), c-kit, c-met, fibroblast growth factor receptors (FGFRs), hepatocyte growth factor receptors (HGFRs), Trk receptors (TrkA, TrkB, and TrkC), ephrin (Eph) receptors, the RET protooncogene, and Akt kinases.
  • EGFR epidermal growth factor family receptors
  • PDGFRs platelet derived growth factor receptors
  • inhibitors of growth factor receptors include ligand antagonists, antibodies, tyrosine kinase inhibitors, anti-sense oligonucleotides and aptamers. Any of these growth factor receptor inhibitors may be employed in combination with the compounds of the present invention in any of the compositions and methods/uses described herein.
  • Trastuzumab (Herceptin®) is an example of an anti- erbB2 antibody inhibitor of growth factor function.
  • an anti-erbB1 antibody inhibitor of growth factor function is cetuximab (ErbituxTM, C225).
  • small molecule inhibitors of epidermal growth factor receptors include but are not limited to lapatinib (TykerbTM), erlotinib (TARCEVA®), gefitinib (IRESSA®), canetinib or CU 033.
  • lmitanib is one example of a PDGFR inhibitor.
  • VEGFR inhibitors include pazopanib, ZD6474, AZD2171 , PTK787, SU1 1248 and sunitinib.
  • Tyrosine kinases that are not growth factor receptor kinases are termed non- receptor tyrosine kinases.
  • Inhibitors of non-receptor tyrosine kinases are sometimes referred to as "anti-metastatic agents" and are useful in the present invention.
  • Targets or potential targets of anti-metastatic agents include, but are not limited to, c-Src, Lck, Fyn, Yes, Jak, abl kinase (c-Abl and Bcr-Abl), FAK (focal adhesion kinase) and Bruton's tyrosine kinase (BTK).
  • Non-receptor kinases and agents, which inhibit non-receptor tyrosine kinase function are described in Sinha, S. and Corey, S. J., (1999) J.
  • SH2/SH3 domain blockers are agents that disrupt SH2 or SH3 domain binding in a variety of enzymes or adaptor proteins including, but not limited to, PI3-K p85 subunit, Src family kinases, adaptor molecules (She, Crk, Nek, Grb2) and Ras-GAP.
  • Src inhibitors include but are not limited to dasatinib and BMS-354825 (J.Med.Chem (2004) 47:6658-6661 ).
  • serine/threonine kinase inhibitors include, but are not limited to polo- like kinase inhibitors (PIk family e.g., PIkI , Plk2, and Plk3),such as 5- ⁇ 6-[(4- Methylpiperazin-1-yl)methyl]-1 H-benzimidazol-1-yl ⁇ -3- ⁇ (1 R)-1-[2-
  • PIk family e.g., PIkI , Plk2, and Plk3
  • MAP kinase cascade blockers which include Ras/Raf kinase inhibitors, mitogen or extracellular regulated kinases (MEKs), and extracellular regulated kinases (ERKs); Aurora kinase inhibitors (including inhibitors of Aurora A and Aurora B); protein kinase C (PKC) family member blockers; inhibitors of kappa-B (IkB) kinase family (IKK-alpha, IKK-beta); PKB/Akt kinase family inhibitors; and inhibitors of TGF-beta receptor kinases.
  • Ras/Raf kinase inhibitors mitogen or extracellular regulated kinases (MEKs), and extracellular regulated kinases (ERKs)
  • Aurora kinase inhibitors including inhibitors of Aurora A and Aurora B
  • PKC protein kinase C family member blockers
  • IkB inhibitors of kappa-B
  • PIk inhibitors examples include PIk inhibitors, also referred to as urokinase-type Plasminogen Activator
  • uPA expression may be used in combination with the compounds of the present invention in the compositions and methods described above.
  • Inhibitors of kinases involved in the IGF signalling axis may also be useful in combination with the compounds of the present invention.
  • Such inhibitors include but are not limited to inhibitors of JNK1/2/3, PI3K, AKT and MEK, and 14.3.3 signalling inhibitors.
  • Cell cycle signaling inhibitors including inhibitors of cyclin dependent kinases (CDKs) are also useful in combination with the compounds of the invention in the compositions and methods described above.
  • CDKs cyclin dependent kinases
  • Examples of cyclin dependent kinases, including CDK2, CDK4, and CDK6 and inhibitors for the same are described in, for instance, Rosania G. R., et al., Exp. Opin. Ther. Patents (2000) 10:215-230.
  • Receptor kinase angiogenesis inhibitors may also find use in the present invention.
  • Inhibitors of angiogenesis related to VEGFR and TIE-2 are discussed above in regard to signal transduction inhibitors (both are receptor tyrosine kinases).
  • Other inhibitors may be used in combination with the compounds of the present invention.
  • anti-VEGF antibodies which do not recognize VEGFR (the receptor tyrosine kinase), but bind to the ligand; small molecule inhibitors of integrin (alpha v beta 3 ) that inhibit angiogenesis; endostatin and angiostatin (non-RTK) may also prove useful in combination with the compounds of the invention.
  • VEGFR antibody is bevacizumab (AvastinTM).
  • Inhibitors of phosphotidyl inositol-3-OH kinase family members including blockers of PI3-kinase, ATM, DNA-PK, and Ku may also be useful in combination with the present invention.
  • myo-inositol signaling inhibitors such as phospholipase C blockers and myoinositol analogues.
  • antisense therapies include those directed towards the targets described above such as ISIS 2503 and gene therapy approaches such as those using thymidine kinase or cytosine deaminase.
  • Immunotherapeutic regimens include ex-vivo and in-vivo approaches to increasing immunogenicity of patient tumor cells such as transfection with cytokines (IL-2, IL-4, GMCFS and MCFS), approaches to increase T- cell activity, approaches with transfected immune cells and approaches with anti- idiotypic antibodies.
  • Agents used in proapoptotic regimens e.g., Bcl-2 antisense oligonucleotides
  • Bcl-2 antisense oligonucleotides may also be used in combination with the compounds of the invention.
  • Members of the Bcl-2 family of proteins block apoptosis. Upregulation of Bcl-2 has therefore been linked to chemoresistance.
  • the invention also provides a process for preparing the compound of formula (I) and pharmaceutically acceptable derivatives thereof.
  • compounds of formula (I) are prepared by reacting the three main components of the compounds, referred to herein as the head, core, and tail of the compounds.
  • Each compound of formula (I) may be conveniently prepared by separately preparing three constituents of the compound and subsequently combining those constituents to form the compound (I).
  • the three constituents are referred to herein as the head (II), the core (III), and the tail (IV).
  • head, core, and tail nomenclature is used throughout to refer to each constituent when referred to individually, and also to refer to the corresponding moiety when described in the context of head/core, tail/core, and/or head/core/tail combinations.
  • the head component of the invented compounds is an o-amino carboxamide represented by formula (II):
  • each of substituents L 1 and L 2 represent a leaving group, e.g. a halogen, preferably chlorine, or OTF, and X represents a protecting group, e.g. sulfonamide or alcholated alkyl, e.g. allyl, benzyl, or SEM.
  • a leaving group e.g. a halogen, preferably chlorine, or OTF
  • X represents a protecting group, e.g. sulfonamide or alcholated alkyl, e.g. allyl, benzyl, or SEM.
  • the tail component of the invented compounds is a substituted aniline represented by formula (IV):
  • R 7 , R 8 , R 9 , and R 10 are as defined above.
  • the head (II), core (III), and tail (IV) of the compounds may be synthesized and combined, for instance, with the synthetic routes shown below:
  • a solvent i.e. dichloromethane or dichloroethane
  • a nitrating agent i.e. nitric acid
  • a variety of substituted anilines can be prepared via direct SNAR displacement of 4-fluoro nitrobenzenes I-3, I-4, I-5, I-7, and 11-1 (commercially available through Oakwood Products) by addition of base (for example potassium carbonate), solvent (DMF), and a primary or secondary amine (including exemplary amines 11-10 through 11-14.)
  • base for example potassium carbonate
  • solvent DMF
  • a primary or secondary amine including exemplary amines 11-10 through 11-14.
  • Subsequent selective nitro reduction using a catalyst (iron (III) chloride or palladium on carbon), additive (activated carbon), alcoholic or ethereal solvent (MeOH, EtOH, or THF), and reducing agent (hydrogen gas or hydrazine) affords the desired anilines II-8.
  • SNAR displacement with a versatile multifunctional amine affords Boc protected nitrobenzene II-2.
  • Boc deprotection with acid i.e. hydrochloric or trifluoroacetic acid
  • alkylation at nitrogen with base and an electrophile potassium carbonate, iso-butyl iodide
  • electrophile potassium carbonate, iso-butyl iodide
  • acylation at nitrogen with a base and an electrophile followed by displacement with an amine triethyl amine, ⁇ -CI acetyl bromide, then dimethyl amine
  • SNAR displacement with amino-alcohol 11-5 followed by alcohol oxidation using an oxidant affords ketone 11-6.
  • Reductive amination with a buffered proton source for example acetic acid with triethyl amine), solvent (dichloroethane), and amine (for example exemplary amine 11-17) followed by nitro reduction as described above for intermediate II-8 or with a hydride source (for example sodium borohydride) with added metal (for example NiC ⁇ ) in alcoholic solvent (MeOH) affords the corresponding functionalized aniline II-9.
  • Reduction of this intermediate typically is carried out in two steps, first via reduction of pyridinium by addition of a hydride source (sodium borohydride or sodium cyanoborohyride) in solvent and second via reduction of the in- situ generated olefin and nitro-group using catalyst (palladium on carbon) in an alcoholic or ethereal solvent (THF, MeOH, EtOH) under pressurized hydrogen gas (typically 30-50 psi) affords piperidine analog III-7.
  • a poisoned hydrogenation catalyst for example Pt-C with sulphide
  • acetamide III-8 Protection of this anline with a suitable protecting group (i.e. trifluoroacetic anhydride, triethyl amine) affords acetamide III-8.
  • a suitable protecting group i.e. trifluoroacetic anhydride, triethyl amine
  • One-step exhaustive pyridine reduction can be effected with a catalyst (Pt-carbon) under hydrogen gas (30-50 psi) with alcoholic or ethereal solvent (typically MeOH, EtOH, THF) to give the corresponding piperidine.
  • bicyclic indoline tails may be prepared using 5-methoxy indole (Aldrich) as a starting material.
  • the 5-methoxy indole is first reduced using a hydride source, e.g. Na(CN)BH 3 , and acid, e.g. AcOH, then protected with an acylating agent, e.g. Ac 2 O, in solvent, e.g. AcOH, optionally with a base.
  • the protection provides for subsequent selective nitration with nitrating agent, e.g. HNO 3 /AC 2 O, followed by deacylation with acid, e.g. HCI in solvent, e.g. MeOH to provide intermediate IV-2
  • Intermediate IV-2 is subjected to subsequent acylation with an acylating agent, e.g. ⁇ -bromoacetyl chloride or an amino acid, with coupling reagents, e.g. HATU, DMAP, and base, e.g. PS-DIPEA, and solvent, e.g. THF.
  • the ⁇ -bromoacetyl product may be reacted with an appropriate amine base, e.g. Me 2 NH (with solvent, e.g. THF), and the product then reduced (see reduction of II-4, Scheme 2) to provide anilino indoline tails IV-3.
  • an acylating agent e.g. ⁇ -bromoacetyl chloride or an amino acid
  • coupling reagents e.g. HATU, DMAP, and base, e.g. PS-DIPEA
  • solvent e.g. THF
  • the set of reaction described above for Scheme 4 can be reordered to allow for the elimination of the acylation/deacylation sequence.
  • indoline IV-6 commercially available as the HCI salt from ChemPacific
  • an acylating agent ⁇ -bromo-acetyl chloride
  • base optionally a trialkyl amine base such as triethyl amine or an inorganic base such as potassium carbonate
  • a nucleophile for example dimethyl amine
  • Chloro aniline V-3 is readily prepared via acylation of 6-nitro indoline with an acylating agent, e.g. ⁇ -bromoacetyl chloride, subsequent displacement with an amine base, and nitro-reduction (hydrogenation). Subsequent reaction with an electrophilic halogen source, e.g. NCS, affords V-3.
  • an acylating agent e.g. ⁇ -bromoacetyl chloride
  • an electrophilic halogen source e.g. NCS
  • aniline V-5 is produced via nitration and deprotection of amide V-4 (see International Patent Publication WO 2001023374), acylation, e.g. ⁇ -bromoacetyl chloride and a suitable base (i.e. K 2 CO 3 ), subsequent displacement with an amine base (for example dimethyl amine or pyrrolidine), and nitro-reduction via hydrogenation (with a palladium source, i.e. Pd/C, and hydrogen gas).
  • a palladium source i.e. Pd/C, and hydrogen gas
  • Aniline V-9 is prepared starting with nitro-indoline V-7 according to Scheme 5.
  • Acylation with an acylating agent for example here acroyl chloride
  • a triakylamine base i.e. Hunig's base
  • solvent dichloromethane
  • 1 ,4- conjugate addition of a nucleophile for example dimethyl amine
  • a nucleophile for example dimethyl amine
  • nitro reduction as described for Intermediate II-4 Scheme 2
  • Further reduction of the amide can be effected via treatment of V-8 with a hydride reducing agent (lithium aluminum hydride) in ethereal solvent (for example tetrahydrofuran or diethyl ether) to afford V-9.
  • a hydride reducing agent lithium aluminum hydride
  • ethereal solvent for example tetrahydrofuran or diethyl ether
  • tetrahydroquinoline tails may be prepared as follows.
  • Intermediate (VI-1 ) prepared according to Chem. Pharm. Bull. (2001 ) p.822, may be alkylated with an alkylating agent and base, e.g. MeI and K 2 CO3, to provide VI-2, then amide reduced, e.g. borane, treated with an acylating agent, e.g. ⁇ -chloro acetyl choride, an amine base, e.g. dimethyl amine or other exemplary amines, and nitro-reduced (as with Intermediate II-4, Scheme 2) to provide (VI-3).
  • an alkylating agent and base e.g. MeI and K 2 CO3
  • amide reduced e.g. borane
  • an acylating agent e.g. ⁇ -chloro acetyl choride
  • an amine base e.g. dimethyl amine or other exemplary amines
  • Furan-containing carboxamide VIII-8 can be generated from 3-furanoic acid VIII- 6.
  • a Curtius rearrangement may be effected via activation of VIII-6 (oxalyl chloride or DPPA) and an azide source (sodium azide or DPPA) which when carried out in the presence of base (triethyl amine) in t-BuOH solvent affords the corresponding 3-N-boc furan.
  • Subsequent exposure to 2 equivalents of a strong base (for example tertiary butyl lithium) and then an electrophile (using methylchloroformate above) affords functionalized furan VIII-7.
  • Boc-deprotection of VIII-7 can be carried out via direct treatement with acid (TFA or HCI) to afford free aniline VIII-8.
  • the head (IX-8 & IX-9, IX-10) and core (IX-1 ) can be combined as illustrated in Scheme 9a-b.
  • 2,4-dichloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidine is combined with an appropriate amine or aniline (for example IX-8 or IX-9) and a suitable solvent (for example trifluoroethanol) with acid (TFA) to give the corresponding 4- substituted pyrrolo[2,3-d] pyrimidines (IX-2 through IX-4) or with a suitable amine (i.e. IX- 10) in solvent (iPrOH) with base (Hunig's base).
  • the carboxylate esters present on IX-2 through IX-4 may be hydrolyzed under standard aqueous basic conditions (LiOH, water, tetrahydrofuran) and the derived acids (including IX-11 obtained directly) may be activated by conversion to the corresponding acid chlorides by treatment with a chloride source/activating agent (oxalyl chloride) and catalyst (DMF).
  • a chloride source/activating agent oxalyl chloride
  • DMF catalyst
  • the in situ generated acid chlorides react intramolecularly with the 5-nitrogen group to give the corresponding tetracyclic species IX-5 through IX-7 and IX-12.
  • Tetracyclic species IX-5 through IX-7 activate the C(7) chloride toward further displacement (see Scheme 10).
  • Removal of the tosyl protection group proceeds upon treatment with base (KOH, NaOH, NaOMe) in a polar protic solvent (H 2 O, MeOH) with optional cosolvent (dioxane, tetrahdyrofuran) and heating (optionally in a microwave reactor) to afford X-7, X-8, X-9, and X-12.
  • base KOH, NaOH, NaOMe
  • H 2 O, MeOH polar protic solvent
  • cosolvent dioxane, tetrahdyrofuran
  • XI-1 Exposure of commercially available XI-1 to aniline protection conditions (i.e. BOC 2 O, DIPEA), nitro reduction conditions (i.e. Pd/C, hydrogen gas), acylation conditions (i.e. ⁇ - bromo acetyl chloride, DIPEA), and then introduction of a nucleophile (i.e. dimethyl amine) affords XI-2. Deprotection of XI-2 with acid (i.e. HCI, or TFA) affords XI-3.
  • aniline protection conditions i.e. BOC 2 O, DIPEA
  • nitro reduction conditions i.e. Pd/C, hydrogen gas
  • acylation conditions i.e. ⁇ - bromo acetyl chloride, DIPEA
  • XI-2 Deprotection of XI-2 with acid (i.e. HCI, or TFA) affords XI-3.
  • APC Allophycocyanin M (molar);
  • BSA bovine serum albumin
  • DIPEA diisopropylethylamine
  • GST Glutathione S-transferase
  • OTF trifluoromethane sulfonate
  • HEPES N-(2-Hydroxyethyl)piperazine-N'- psi (pounds per square inch);
  • 2-ethanesulfonic acid 2-ethanesulfonic acid
  • RP reverse phase
  • hlGF1 R or IGF-1 R human Insulin-like rt (room temperature)
  • growth factor 1 receptor kinase SEM (2-trimethylsilyl(ethoxymethyl) hlR (human insulin receptor kinase)
  • TBAI tetrabutylammonium iodide
  • TLC Thin-layer chromatography
  • APCI Atmospheric Pressure Chemical Ionization
  • ESI ESI Ionization
  • HRMS High resolution mass spectral data
  • Step C/lntermediate A4 1 ,1-dimethylethyl 4-[2-(methylsulfonyl)ethyl]-1- piperazinecarboxylate
  • Step E/lntermediate A6 1-[2-methyl-5-(methyloxy)-4-nitrophenyl]-4-[2- (methylsulfonyl)ethyl]piperazine
  • Step F/lntermediate A1 5-methyl-2-(methyloxy)-4- ⁇ 4-[2-(methylsulfonyl)ethyl]-1- piperazinyl ⁇ aniline
  • Step A/Intermediate A8 ⁇ /, ⁇ /-dimethyl-2-[5-(methyloxy)-2,3-dihydro-1 /-/-indol-1-yl]-2- oxoethanamine
  • the brown solid (135g, 499 mmol) was dissolved in DCM (500ml), cooled to 0 0 C, then treated with K 2 CO 3 (139g, 999 mmol), and 650ml of 2 M dimethyl amine in THF. The reaction was stirred at 0 0 C for 1 hour and filtered. The resulting solid was washed with DCM. The combined filtrates were washed with water, dried over Na 2 SO 4 , filtered, and concentrated to provide ⁇ /, ⁇ /-dimethyl-2-[5-(methyloxy)-2,3-dihydro-1/-/-indol-1-yl]-2- oxoethanamine as a grey solid (111 g, 95%).
  • Step B/lntermediate A9 ⁇ /, ⁇ /-dimethyl-2-[5-(methyloxy)-6-nitro-2,3-dihydro-1/-/-indol-1- yl]-2-oxoethanamine
  • the organic layer was separated, dried over with Na 2 SO 4 , and the solvents were removed under reduced pressure to yield ⁇ /, ⁇ /-dimethyl-2-[5-(methyloxy)-6-nitro-2,3-dihydro-1/-/-indol-1-yl]-2- oxoethanamine (13Og, 99%).
  • Step C/lntermediate A7 1-[(dimethylamino)acetyl]-5-(methyloxy)-2,3-dihydro-1 /-/-indol-
  • Step A/Intermediate A1 1 1 ,1-dimethylethyl 4-(methylsulfonyl)-1-piperazinecarboxylate
  • Step G/lntermediate A17 1- ⁇ 1-[2-ethyl-5-(methyloxy)-4-nitrophenyl]-4-piperidinyl ⁇ -4-
  • Step H/lntermediate A10 5-ethyl-2-(methyloxy)-4- ⁇ 4-[4-(methylsulfonyl)-1-piperazinyl]-1- piperidinyl ⁇ aniline
  • Step B/lntermediate A18 4-[4-(1-methylethyl)-1-piperazinyl]-2-(methyloxy)aniline
  • Step A/Intermediate A21 4-[3-(methyloxy)-4-nitrophenyl]pyridine
  • n-Propyliodide 200 ml_, 2.05 mol was added to a solution of 4-(3-methoxy-4- nitrophenyl)py ⁇ dine (25.0 g, 109 mmol) in pinacolone (500 ml_).
  • the reaction was fitted with a reflux condenser, stirred, and heated at 100 0 C for 12 h. A light brown suspension was observed. An aliquot (-2.0 ml.) was taken out from the reaction mixture, concentrated, and analyzed by 1 H NMR which revealed the formation of the alkylated product and the absence of starting material.
  • Step C/lntermediate A23 4-(3-methoxy-4-nitrophenyl)-1-propyl-1 ,2,3,6- tetrahydropyridine
  • Step D/lntermediate A20 2-(methyloxy)-4-(1-propyl-4-piperidinyl)aniline
  • Step A/Intermediate A25 5-(methyloxy)-2,3-dihydro-1 /-/-indole
  • Step B/lntermediate A26 1-acetyl-5-(methyloxy)-2,3-dihydro-1 /-/-indole
  • Step C/lntermediate A27 1-acetyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole
  • Step D/lntermediate A28 5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole
  • Step E/lntermediate A24 1- ⁇ [ethyl(methyl)amino]acetyl ⁇ -5-(methyloxy)-2,3-dihydro-1 H- indol-6-amine
  • Step B/lntermediate A33 6-hydroxy-3,4-dihydro-2(1 H)-quinolinone
  • Step D/lntermediate A35 6-(methyloxy)-7-nitro-3,4-dihydro-2(1 H)-quinolinone
  • Step E/lntermediate A36 6-(methyloxy)-7-nitro-1 ,2,3,4-tetrahydroquinoline
  • Step F/lntermediate A37 ⁇ /-ethyl- ⁇ /-methyl-2-[6-(methyloxy)-7-nitro-3,4-dihydro-1 (2H)- quinolinyl]-2-oxoethanamine
  • Step G/lntermediate A31 1- ⁇ [ethyl(methyl)amino]acetyl ⁇ -6-(methyloxy)-1 ,2,3,4- tetrahydro-7-quinolinamine
  • Step A/Intermediate A39 1-(bromoacetyl)-6-(methyloxy)-7-nitro-1 , 2,3,4- tetrahydroquinoline
  • Step B/lntermediate A40 ⁇ /, ⁇ /-dimethyl-2-[6-(methyloxy)-7-nitro-3,4-dihydro-1 (2/-/)- quinolinyl]-2-oxoethanamine
  • Step C/lntermediate A38 1-[(dimethylamino)acetyl]-6-(methyloxy)-1 ,2,3,4-tetrahydro-7- quinolinamine
  • Step A Intermediate A42: 6-(methyloxy)-7-nitro-1-(1-pyrrolidinylacetyl)-1 , 2,3,4- tetrahydroquinoline
  • Step B/lntermediate A41 6-(methyloxy)-1-(1-pyrrolidinylacetyl)-1 ,2,3,4-tetrahydro-7- quinolinamine
  • Step B/lntermediate A45 4,4-dimethyl-6-(methyloxy)-3,4-dihydro-2(1/-/)-quinolinone
  • Step C/lntermediate A46 4,4-dimethyl-6-(methyloxy)-7-nitro-3,4-dihydro-2(1 /-/)- quinolinone
  • the reaction mixture was concentrated, dissolved in dichloromethane, and washed with saturated aqueous sodium bicarbonate. The organic layer was separated, the aqueous layer was filtered (due to emulsions) and back- extracted with dichloromethane. The combined organic layers were dried over magnesium sulfate, filtered, concentrated, and triturated with dichloromethane/diethyl ether/hexanes.
  • Step D/lntermediate A47 4,4-dimethyl-6-(methyloxy)-7-nitro-1 ,2,3,4-tetrahydroquinoline
  • Step E/lntermediate A48 1-(bromoacetyl)-4,4-dimethyl-6-(methyloxy)-7-nitro-1 ,2,3,4- tetrahydroquinoline
  • Step F/lntermediate A49 ⁇ 2-[4,4-dimethyl-6-(methyloxy)-7-nitro-3,4-dihydro-1 (2/-/)- quinolinyl]-2-oxoethyl ⁇ dimethylamine
  • Step G/lntermediate A43 1-[(dimethylamino)acetyl]-4,4-dimethyl-6-(methyloxy)-1 ,2,3,4- tetrahydro-7-quinolinamine
  • Step A/Intermediate A51 4,4-dimethyl-6-(methyloxy)-7-nitro-1-(1-pyrrolidinylacetyl)- 1 ,2,3,4-tetrahydroquinoline
  • Step B/lntermediate A50 4,4-dimethyl-6-(methyloxy)-1-(1-pyrrolidinylacetyl)-1 ,2,3,4- tetrahydro-7-quinolinamine
  • Step A/Intermediate A53 1- ⁇ 1-[2-methyl-5-(methyloxy)-4-nitrophenyl]-4-piperidinyl ⁇ -4- (methylsulfonyl)-piperazine
  • Step B/lntermediate A52 5-methyl-2-(methyloxy)-4- ⁇ 4-[4-(methylsulfonyl)-1-piperazinyl]- 1-piperidinyl ⁇ -aniline
  • Step A/Intermediate A55 5-(methyloxy)-6-nitro-1-(1-pyrrolidinylacetyl)-2,3-dihydro-1 /-/- indole
  • Step B/lntermediate A54 5-(methyloxy)-1-(1-pyrrolidinylacetyl)-2,3-dihydro-1 /-/-indol-6- amine
  • Step A/Intermediate A57 5-(methyloxy)-1-[(methyloxy)acetyl]-6-nitro-2,3-dihydro-1 /-/- indole
  • Step B/lntermediate A56 5-(methyloxy)-1-[(methyloxy)acetyl]-2,3-dihydro-1 /-/-indol-6-
  • Step A/Intermediate A59 1 ,1-dimethylethyl 4-(2-iodoethyl)-1-piperidinecarboxylate
  • Step B /Intermediate A60 1 ,1-dimethylethyl 4-[2-(methylthio)ethyl]-1- piperidinecarboxylate
  • Step C/lntermediate A61 1 ,1-dimethylethyl 4-[2-(methylsulfonyl)ethyl]-1- piperidinecarboxylate
  • Step F/lntermediate A58 2-(methyloxy)-4- ⁇ 4-[2-(methylsulfonyl)ethyl]-1- piperidinyl ⁇ aniline
  • Step B/lntermediate A66 1-[3-(methyloxy)-4-nitrophenyl]-4-piperidinone
  • Step D/lntermediate A68 1- ⁇ 1-[3-(methyloxy)-4-nitrophenyl]-4-piperidinyl ⁇ -4- (methylsulfonyl)piperazine
  • Step E/lntermediate A64 2-(methyloxy)-4- ⁇ 4-[4-(methylsulfonyl)-1-piperazinyl]-1- piperidinyl ⁇ aniline
  • NaBH 4 (1.18g, 31.1 mmol) was added carefully in portions (exothermic) to a suspension of 1- ⁇ 1-[3-(methyloxy)-4-nitrophenyl]-4-piperidinyl ⁇ -4-(methylsulfonyl)-piperazine (3.54g, 8.9mmol), nickel(ll)chloride hexahydrate (1.06g, 4.4mmol), MeOH (10OmL) and THF (5OmL) at 0 0 C. The ice bath was removed and the reaction mixture was warmed to rt. TLC analysis indicated the complete consumption of the starting material.
  • reaction mixture was concentrated onto silica gel and purified by flash chromatography to afford 2-(methyloxy)-4- ⁇ 4-[4-(methylsulfonyl)-1-piperazinyl]-1-piperidinyl ⁇ aniline (2.93g, 90%) as a colorless solid.
  • Step A/Intermediate A70 5-bromo-4-fluoro-2-nitrophenyl methyl ether
  • Step B/lntermediate A71 1-[2-fluoro-5-(methyloxy)-4-nitrophenyl]-4-(1- methylethyl)piperazine
  • Step Cintermediate A69 5-fluoro-4-[4-(1-methylethyl)-1-piperazinyl]-2- (methyloxy)aniline
  • Step A/Intermediate A73 1-(1-methylethyl)-4-[2-methyl-5-(methyloxy)-4- nitrophenyl]piperazine
  • Step B/lntermediate A72 5-methyl-4-[4-(1-methylethyl)-1-piperazinyl]-2- (methyloxy)aniline
  • Step B/lntermediate A76 4-[2-methyl-5-(methyloxy)-4-nitrophenyl]-1-propylpyridinium iodide
  • Step C/lntermediate A77 4-[2-methyl-5-(methyloxy)-4-nitrophenyl]-1-propyl-1 , 2,3,6- tetrahydropyridine
  • Step D/lntermediate A74 5-methyl-2-(methyloxy)-4-(1-propyl-4-piperidinyl)aniline
  • Step B/lntermediate A80 4-chloro-5-fluoro-2-nitrophenyl methyl ether
  • Step C/lntermediate A81 1-[2-chloro-5-(methyloxy)-4-nitrophenyl]-4-(1- methylethyl)piperazine
  • Step A/Intermediate A85 ⁇ /, ⁇ /-dimethyl-3-[5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indol-1- yl]-3-oxo-1 -propanamine
  • Step B/lntermediate A86 1-[3-(dimethylamino)propanoyl]-5-(methyloxy)-2,3- dihydro-1H-indol-6-amine
  • Step C/lntermediate A84 1-[3-(dimethylamino)propyl]-5-(methyloxy)-2,3-dihydro-1 H- indol-6-amine
  • Step B/ Intermediate A91 1 ,1-dimethylethyl (2S)-2- ⁇ [6-amino-5-(methyloxy)-2,3-dihydro- 1 H-indol-1-yl]carbonyl ⁇ -1-pyrrolidinecarboxylate
  • Distinct rotamers are present at room temperature by 1 H NMR.
  • 1 H NMR 400 MHz ⁇ ppm 7.54 (s, 1 H), 7.51 (s, 1 H), 6.69 (s, 2 H), 4.62 (s, 4 H), 4.36 - 4.50 (m, 1 H), 4.08 - 4.17 (m, 1 H), 3.95 - 4.07 (m, 4 H), 3.69 (s, 6 H), 3.24 - 3.48 (m, 4 H), 2.86 - 3.12 (m, 4 H), 2.08 - 2.31 (m, 2 H), 1.66 - 2.01 (m, 6 H), 1.37 (s, 9 H), 1.22 (s, 9 H).
  • Step A/Intermediate A100 1-acetyl-3,3-dimethyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/- indole
  • Step B/lntermediate A101 3,3-dimethyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indole
  • Step C/lntermediate A102 ⁇ 2-[3,3-dimethyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indol- 1-yl]-2-oxoethyl ⁇ dimethylamine
  • StepD/lntermediate A99 1-[(dimethylamino)acetyl]-3,3-dimethyl-5-(methyloxy)-2,3- dihydro-1 H-indol-6-amine
  • Step A/Intermediate A104 6-(ethyloxy)-3,4-dihydro-2(1H)-quinolinone
  • StepB/lntermediate A105 6-(ethyloxy)-7-nitro-3,4-dihydro-2(1H)-quinolinone
  • Step C/lntermediate A106 6-(ethyloxy)-7-nitro-1 ,2,3,4-tetrahydroquinoline
  • Step D/lntermediate A107 ⁇ 2-[6-(ethyloxy)-7-nitro-3,4-dihydro-1 (2/-/)-quinolinyl]-2- oxoethyl ⁇ dimethylamine
  • Step A/Intermediate A109 5-(methyloxy)-1-(1-methyl-L-prolyl)-6-nitro-2,3-dihydro-1 H- indole
  • Step B/lntermediate A108 5-(methyloxy)-1-(1-methyl-L-prolyl)-2,3-dihydro-1 /-/-indol-6-
  • Step A/Intermediate A1 11 1-methyl-D-proline
  • Step B/lntermediate A1 12 5-(methyloxy)-1-(1-methyl-D-prolyl)-6-nitro-2,3-dihydro-1 /-/- indole
  • Step C/lntermediate A110 5-(methyloxy)-1-(1-methyl-D-prolyl)-2,3-dihydro-1H-indol-6- amine
  • Step A/Intermediate A1 14 ⁇ /, ⁇ /-dimethyl-2-(6-nitro-2,3-dihydro-1 H-indol-1-yl)-2- oxoethanamine
  • Step B/lntermediate A1 13 1-[(dimethylamino)acetyl]-2,3-dihydro-1 /-/-indol-6-amine
  • a suspension of ⁇ /, ⁇ /-dimethyl-2-(6-nitro-2,3-dihydro-1 /-/-indol-1-yl)-2-oxoethanamine (13.6 g, 54.6 mmol), hydrazine hydrate (21.42 ml_, 436 mmol), iron (III) chloride (1.768 g, 10.91 mmol), activated carbon (15 g), and methanol (100 ml.) was maintained at 65 0 C for 12 hours, cooled, and filtered through celite (rinsed with additional methanol). Filtrates were concentrated, redissolved in ethyl acetate, and washed twice with saturated aqueous sodium chloride and sodium bicarbonate.
  • Step A/Intermediate A1 18 N,N-dimethyl-D-alanine
  • Step B/lntermediate A1 19 1-[(2R)- ⁇ /, ⁇ /-dimethyl-1-[5-(methyloxy)-6-nitro-2,3-dihydro-1 H- indol-1 -yl]-1 -oxo-2-propanamine
  • Step C/lntermediate A117 1-[(2R)-2-(dimethylamino)propanoyl]-5-(methyloxy)-2,3- dihydro-1 H-indol-6-amine
  • Step A/Intermediate A121 N,N-dimethyl-L-alanine
  • Step B/lntermediate A122 (2S)-N,N-dimethyl-1-[5-(methyloxy)-6-nitro-2,3-dihydro-1 H- indol-1 -yl]-1 -oxo-2-propanamine
  • Step C/lntermediate A120 1-[(2S)-2-(dimethylamino)propanoyl]-5-(methyloxy)-2,3- dihydro-1 H-indol-6-amine
  • Step A/Intermediate A123 1 ,1-dimethylethyl 4-[3-(methyloxy)-4-nitrophenyl]-1- piperazinecarboxylate
  • Step B/lntermediate A129 N,N-dimethyl-2- ⁇ 4-[3-(methyloxy)-4-nitrophenyl]-1- piperazinyl ⁇ -2-oxoethanamine
  • Step C/lntermediate A127 4- ⁇ 4-[(dimethylamino)acetyl]-1-piperazinyl ⁇ -2- (methyloxy)aniline
  • N,N-dimethyl-2- ⁇ 4-[3-(methyloxy)-4-nitrophenyl]-1-piperazinyl ⁇ -2-oxoethanamine (1.00 g, 3.10 mmol) was added to a thick-walled hydrogenation flask.
  • ethanol 15 mL
  • ethyl acetate 15 mL
  • methanol 15 mL
  • 10% palladium on carbon (0.330 g, 0.310 mmol) was added.
  • the flask was purged with vacuum and nitrogen (3x).
  • the flask was purged with vacuum and hydrogen (3x).
  • the flask was then maintained under H 2 atm (50 psi) for approximately 24 hours.
  • Step B/lntermediate A132 1-[5-(ethyloxy)-2-methyl-4-nitrophenyl]-4-[2- (methylsulfonyl)ethyl]piperazine
  • Step C/lntermediate A130 2-(ethyloxy)-5-methyl-4- ⁇ 4-[2-(methylsulfonyl)ethyl]-1- piperazinyl ⁇ aniline
  • Step A/Intermediate A134 (3R)-N,N-dimethyl-1- ⁇ 1-[3-(methyloxy)-4-nitrophenyl]-4- piperidinyl ⁇ -3-pyrrolidinamine
  • Step B/lntermediate A133 (3R)-1- ⁇ 1-[4-amino-3-(methyloxy)phenyl]-4-piperidinyl ⁇ -N,N- dimethyl-3-pyrrolidinamine
  • Step A/Intermediate A138 1 '-[2-methyl-5-(methyloxy)-4-nitrophenyl]-1 ,4'-bipiperidine
  • Step B/lntermediate A137 4-(1 ,4'-bipiperidin-1 '-yl)-5-methyl-2-(methyloxy)aniline
  • Step A/Intermediate A140 1 ,1-dimethylethyl [2-(methyloxy)-5-nitrophenyl]carbamate
  • Step B/lntermediate A141 1 ,1-dimethylethyl [5-amino-2-(methyloxy)phenyl]carbamate
  • Step C/lntermediate A142 1 ,1-dimethylethyl [5-[( ⁇ /, ⁇ /-dimethylglycyl)amino]-2-
  • Step D/lntermediate A139 ⁇ / 1 -[3-amino-4-(methyloxy)phenyl]- ⁇ / 2 , ⁇ / 2 -dimethylglycinamide hydrogen chloride
  • Step A/Intermediate A144 N,N-dimethyl-2-(6-methyl-7-nitro-3,4-dihydro-1 (2H)- quinolinyl)-2-oxoethanamine
  • Step B/lntermediate A143 1-[(dimethylamino)acetyl]-6-methyl-1 ,2,3,4-tetrahydro-7- quinolinamine
  • Step A/Intermediate A146 (2R)-1-acetyl-2-methyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/- indole
  • Step C/lntermediate A148 N,N-dimethyl-2-[(2R)-2-methyl-5-(methyloxy)-6-nitro-2,3- dihydro-1 H-indol-1 -yl]-2-oxoethanamine
  • Step A/Intermediate A150 (2S)-1-acetyl-2-methyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 H- indole
  • Step B/lntermediate A151 (2S)-2-methyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole hydrogen chloride
  • Step C/lntermediate A152 N,N-dimethyl-2-[(2S)-2-methyl-5-(methyloxy)-6-nitro-2,3- dihydro-1 H-indol-1 -yl]-2-oxoethanamine
  • the filtrate was concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH 3 )/CH 2 CI 2 .
  • the foamy oily product was chased using CH 2 CI 2 and hexanes, CH 2 CI 2 and Et 2 O, then Et 2 O to obtain N,N-dimethyl-2-[(2S)-2-methyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indol-1-yl]-2- oxoethanamine as an orange solid (2.50 g, 47%).
  • Step D/lntermediate A149 (2S)-1-[(dimethylamino)acetyl]-2-methyl-5-(methyloxy)-2,3- dihydro-1 H-indol-6-amine
  • Step A/Intermediate A154 1-acetyl-5-fluoro-6-nitro-2,3-dihydro-1 /-/-indole
  • Step B/lntermediate A155 5-fluoro-6-nitro-2,3-dihydro-1 /-/-indole

Abstract

Novel pyrrolopyrimidines as shown in formula (I): and pharmaceutically acceptable derivatives thereof. The compounds are useful in the inhibition of IGF-1R.

Description

PYRROLOPYRIMIDINE COMPOUNDS
FIELD OF THE INVENTION
The present invention relates to pyrrolopyrimidine derivatives, compositions and medicaments containing the same, as well as processes for the preparation and use of such compounds, compositions and medicaments. Such pyrrolopyrimidine derivatives are useful in the treatment of diseases associated with inappropriate IGF-1 R and IR activity.
BACKGROUND OF THE INVENTION
Receptor tyrosine kinases (RTKs) have been implicated in cellular signaling pathways that control various cellular functions, including cell division, growth, metabolism, differentiation, and survival, through reversible phosphorylation of the hydroxyl groups of tyrosine residues in proteins. Extracellular signals are transduced via activation of the cell surface receptors, with amplification and propagation using a complex choreography of cascades of protein phosphorylation, and protein dephosphorylation events to avoid uncontrolled signaling. These signaling pathways are highly regulated, often by complex and intermeshed kinase pathways where each kinase may itself be regulated by one or more other kinases and protein phosphatases. The biological importance of these finely tuned systems is such that a variety of cell proliferative disorders have been linked to defects in one or more of the various cell signaling pathways mediated by tyrosine or serine/threonine kinases.
Receptor tyrosine kinases (RTKs) catalyse phosphorylation of certain tyrosyl amino acid residues in various proteins, including themselves, which govern cell growth, proliferation and differentiation. Insulin-like growth factor- 1 receptor (IGF-1 R) is a transmembrane tyrosine kinase ubiquitous among fetal and post-natal cell types.
The IGF signaling axis is made up of multiple ligands (IGF-1 , IGF-2 and Insulin), at least six high affinity ligand binding proteins and proteases, multiple receptors (IGF- 1 R & IGF-2R, IR and IRR), and many other down stream signaling proteins (Pollak, MN et al., Nature Reviews Cancer (2004) 4(7):505-518). The structure and function of the IGF-1 R has been reviewed by Adams et al., Cell. MoI. Life Sci. (2000) 57:1050-1093 and Benito, M et al., lnt J Biochem Cell Biol (1996) 28(5):499-510. The receptor is activated by the ligands IGF-1 and IGF-2, which are mitogenic proteins that signal through the IGF-1 R and IR in an endocrine, paracrine or autocrine manner. Activation of the IGF-1 receptor elicits cellular responses which include cellular proliferation and protection of cells from apoptosis. (Id.) Overexpression of IGF-1 R leads to malignant transformation of cultured cells, while downregulation can reverse the transformed phenotype of tumor cells and potentially render them susceptible to apoptosis. (Id.)
There are two splice variants of the IR gene, the IR-B isoform which regulates glucose uptake and is expressed in liver, muscle and adipose tissue, and the exon 1 1 variant human insulin receptor isoform A (IR-A) binds IGF-2 with high affinity and promotes proliferation and protection from apoptosis (Sciacca L. Oncogene (2002) 21 (54):8240-8250). IR-A is predominantly expressed in fetal tissue and malignancies and at this receptor, IGF-2 is more potent than insulin in stimulating cancer cell migration. (Sciacca, Oncogene (2002) supra). Insulin receptor-related receptor (IRR) has 79% homology with the kinase domain of IR and is expressed only in a few limited cell types (Dandekar, AA et al., Endocrinology (1998) 139(8):3578-3584).
IGF-1 R is a hetero-tetrameric, transmembrane, cell surface receptor. (Benito, lnt J Biochem Cell Biol (1996)) An IGF-1 binding domain is part of the extracellular alpha- chain of IGF-1 R, whereas the intracellular beta-chain contains the tyrosine kinase domain. Three tyrosine residues represent autophosphorylation sites, specifically Tyr1 131 , Tyr1135, and Tyr1136, within the activation loop of the IGF-1 R beta catalytic domain (Li, W et al., J. Biol. Chem. (2006) 281 (33):23785-23791 ). Phosphorylation of all three is required for full receptor activation, and preceeds phosphorylation of juxtamembrane tyrosines and carboxy terminus serines. The insulin receptor has three similar autophosphorylation sites on the activation loop and juxtamembrane region. Activation and autophoshorylation results in the recruitment of multiple docking proteins and the generation of intracellular signaling (Benito, lnt J Biochem Cell Biol (1996)). Once activated, IGF-1 R and IR can phosphorylate or interact directly with a number of intracellular protein substrates, including IRS-1 , IRS-2, Grb2, Grb10, Grb14, She, SOC, 14.3.3, FAK, or indirectly with other proteins like PI3K and MAPK (Benito, M et al. lnt J Biochem Cell Biol (1996) 28(5):499-510) (Brown, GC et al., Biochem. J (1992) 284:1-13; Bruning, JC et al., MoI. Cell (1998) 2(5):559-569). Focal adhesion kinase (FAK) is of particular interest because of its role as a regulator of cell survival, proliferation, migration and invasion. FAK is activated by growth factor receptors such as IGF-1 R, by binding through its N-terminal domain and autophosphorylation at TyR397. Activated or over expressed FAK is common in a wide variety of cancers, and may play a role in human carcinogenesis (van Nimwegen, MJ et al., Biochem. Pharmacol. (2007) 73(5):597-609). In addition to its role in cancers, the insulin-like growth factor receptor plays important and diverse roles in growth and development (Benito, M et al. lnt J Biochem Cell Biol (1996) 28(5):499-510). It IGF-1 R has been implicated in several metabolic, and immunological diseases (Walenkamp, MJ et al., Horm. Res. (2006) 66(5):221-230; Kurmasheva, R. T et al., Biochim. Biophys. Acta -Rev on Cancer (2006) 1766(1 ):1-22; Bateman, JM et al., Cell. MoI. Life Sci. (2006) 63(15):1701-1705, LeRoith, D, et al., Can. Lett. (2003) 195:127-137 and Samani A, et al., Endocrine Reviews 28(1 ):20-47.)
The role of the IGF/IGF-1 R signaling system in cancer has been thoroughly examined over the last 15 years. In particular, the implication of IGF-1 R in human cancer stems from its roles in stimulating mitogenesis, mobility and metastasis and in protecting against apoptosis. (Kurmasheva, Biochim. Biophys. Acta (2006).) Interest has grown with the understanding that in addition to its antiapoptotic and mitogenic roles, IGF/IGF-1 R signaling seems to be necessary for the establishment and continuation of a transformed phenotype. It has been well established that constitutive activation or over expression, often results in non-adherent cell growth, even under serum depleted conditions in vitro, and is associated with the formation of tumors in nude mice. (Kaleko M et al, MoI Cell Biol. (1990) 10(2): 464-473). Perhaps even more importantly, it has been firmly established that cells, in which the gene encoding for IGF- 1 R has been deactivated, are totally resistant to transformation by agents which are normally capable of immortalizing normal cells, such as over expression of PDGFR or EGFR, the T antigen of the SV40 virus, the E5 protein of bovine papilloma virus, and activated ras. (DeAngelis T et al., Cell. Physiol. (1995) 164() :214-221 ; Coppola D et al., MoI. Cell. Biol. (1994) 14(7) :4588-4595 ; Morrione AJ, Virol. 1995 695300-5303; Sell C et al., MoI. Cell. Biol. (1994) 14(6):3604-3612; Sell C et al., Proc. Natl. Acad. Sci. USA (1993) 90(23):11217-1 1221 ). Thus, IGF-1 R has been identified as the major survival factor that protects from oncogene induced cell death (Harrington et al., EMBO J. (1994) 13():3286-3295). IGF-1 R is expressed in a large number and variety of tumors and the IGFs amplify the tumor growth through their interaction with the receptor. Evidence supporting the role of IGF-1 R in carcinogenesis can be found in studies using monoclonal antibodies directed towards the receptor which inhibit the proliferation of numerous cell lines in culture and in vivo (Arteaga C et al., Cancer Res. (1989) 49(22):6237-6241 ; Li et al., Biochem. Biophys. Res. Com. (1993) 196(1 ):92-98; Scotlandi K et al., Cancer Res. (1998) 58(18):4127-4131 ). Dominant negative IGF-1 R is capable of inhibiting tumor proliferation (Jiang et al., Oncogene (1999) 18(44):6071- 6077).
The IGF signaling axis is implicated in various tumor types including: breast cancer (Surmacz, J. Mammary Gland Bio. Neoplasia (2000) 5(1 ):95-105, LeRoith, Can. Lett. (2003) and Artega, Cancer Res. (1989)), bone and bone marrow cancers including Ewing's sarcoma, osteosarcoma, giant cell tumor of bone (Scotlandi, Cancer Res. (1998) lung cancer, including non-small cell and small cell lung carcinomas and mesotheliomas (Jiang, Y et al., Oncogene (1999) 18:6071-6077 and LeRoith, Can. Lett. (2003), prostate cancer (Djavan et al., World J Urol. (2001 ) 19(4):225-233; O'Brien et al., Urology (2001 ) 58(1 ):1-7 and LeRoith, Can. Lett. (2003)), colorectal cancer (Guo et al., Gastroenterology, 1992, 102, 1 101-1 108; Durai, R et al., Int. J Colorectal Dis. (2005) 20(3):203-220 and LeRoith, Can. Lett. (2003)), renal cancer (Kellerer M. et al., Int. J. Cancer (1995) 62(5):501-507), pancreatic cancer (Bergmann, U et al., Cancer Res. (1995) 55(10):2007-2011 ), hematopoietic cancers, including lymphoblastic T cell leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, hairy-cell leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic neutrophilic leukemia, acute lymphoblastic T cell leukemia, plasmacytoma, immunoblastic large cell leukemia, mantle cell leukemia, multiple myeloma, megakaryoblastic leukemia, acute megakaryocyte leukemia, promyelocytic leukemia, erythroleukemia, malignant lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, lymphoblastic T cell lymphoma, Burkitt's lymphoma, follicular lymphoma, myelodysplastic syndromes, (Zumkeller W et al., Leuk. Lymph (2002) 43(3):487-491 ; and Qi, Ann Hematol. (2006) 85:95-101.), neuroblastomas (Zumkeller, W et al., Horm. Metab. Res. 1999, 31 , 138-141 ), gliomas, meningiomas, medulloblastomas, astrocytomas, and glioblastoma (Zumkeller, W et al., MoI. Pathol. (2001 ) 54(4):227-229, Del VaIIe L, et al., Clin. Cancer Res. (2002) 8:1822-1830 and Trojan et al., Proc. Natl. Acad. Sci. USA (1992) 89:4874-4878.), thyroid cancer (VeIIa V et al., J. Clin. Endocrinol. Metab. (2002) 87:245-254; VeIIa V et al., MoI. Pathol. (2001 ) 54(3):121-124), and hepatocarcinoma (Alexia, C et al., Biochem. Pharmacol. (2004) 68(1 ):1003-1015). ovarian cancer, testicular cancer, vulval cancer, cervical cancer, endometrial cancer, bladder cancer, esophageal cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, GIST (gastrointestinal stromal tumor), skin cancer including melanoma, and soft-tissue sarcoma.
Thus, in virtually all types of human cancers there is a strong association between dysregulation of IGF signaling and carcinogenesis (Bohula EA et al., Anticancer Drugs (2003) 14(9):669-682). Inhibition of IGF- 1 R expression or function has been shown to block tumor growth and metastasis and also enhance sensitivity to other antineoplastic therapies, including cytotoxic drugs and radiation. (Bohula, Anticancer Drugs (2003).
SUMMARY OF THE INVENTION
We have now found a group of novel pyrrolopyrimidines that are inhibitors of
IGF-1 R.
The present invention provides a compound of formula (I):
Figure imgf000007_0001
or a pharmaceutically acceptable salt or solvate thereof wherein:
R4a and R4b are each independently selected from H, alkylCi-c6, -OH, -alkyleneci-ce-OH,
-alkyleneci-cβ-phenyl (optionally substituted with a halo), and
-alkyleneCi-c6-C(O)NH2; one or two of X1, X2, and X3 are independently selected from N, O, and S, while the remaining one or two of X1, X2, and X3 are C; m is 0 or 1 ; R1 is selected from H, halo, alkylCi-c6, -alkyleneci-cs-haloi-s, and -O-alkylCi-c6when X1 is
C, otherwise R1 is absent; R2 is selected from H, halo, alkylCi-c6, -alkyleneci-cs-haloi-s, and -O-alkylCi-c6when X2 is
C, otherwise R2 is absent; R3 is selected from H, halo, alkylCi-c6, -alkyleneci-cs-haloi-s, and -O-alkylCi-c6when X3 is
C, otherwise R3 is absent; R5 and R6 are independently selected from H and alkylCi-c3;
R7 is selected from alkylCi-c6, -O-alkylCi-c6, halo, -NR22R23, and -O-alkyleneCi-c6-halo1-3;
R8 is selected from H, halo, and alkylCi-c6; one of R9 and R10 is selected from -alk <yylleenneCi-c6-SO2-alkylci-c6, -N-alkyleneCO-c6-C(0)-alkyleneco-c6-NR22R23, -O-alkyleneco-ceCoptionally substituted with -OH)-NR22R23,
Figure imgf000008_0001
(R14)i-2 , and (R14)i-2; and the other of R9 and R10 is selected from H, alkylCi-c6, -O-alkylCi-c6, and halo;
wherein Het1 and Het2 are each independently a five or six membered heterocyclic ring having an N atom and optionally one or two additional heteroatoms selected from
N and O, and each R14 is independently selected from H, OH, halo, alkylCi-c6, -0-alkylCi-c6, -cyclopropyl, -C(O)-alkylCi-c6, SO2-alkylCi-c6, -(CH2)i-4-halo ,and -(CH2)1-4-SO2-alkylci-c6; -C(O )-alkyleneC0-c6-N R22R23, -NR22R23; or
R9 and R10, together with the atoms to which they are attached form a five, six, or seven- membered heterocyclic ring containing one or two N atom and the remainder C atoms, wherein at least one N atom is substituted with R15, and the C atoms of the heterocyclic ring are optionally substituted with (R19)i-2;
wherein R15 is selected from H, -alkylCi-c4, -alkyleneCi-c4-halo,
-C(O)-alkyleneC0-c6-N R22R23, -C(O)-alkylci-c6, -alkyleneCi-c4-N R22R23, -alkyleneCi-c4-C(O)-NR22R23, -C(O)-alkyleneCi-c4-O-alkylCi-c6, -C(O)-pyrrolidine, and -C(O)-pyrrolidine-alkylci-c6; and, each R19 is independently selected from H and alkylCi-c6; each R22 is selected from H, alkylCi-c6, -O-alkylCi-c6, -alkyleneci-cδ-O-alkylci-ce; and,
each R23 is selected from H and alkylCi-c6; or
R22 and R23 combine to form a four, five, or six membered, heterocyclic ring containing the N atom to which they are attached and optionally an additional heteroatom selected from N and O, wherein the ring is optionally substituted with -OH or -alkyld-ce-
According to another embodiment, a compound of formula I is provided as described in any one of the examples. According to another embodiment, a pharmaceutically acceptable derivative of the compound of formula I described in any one of the examples is provided.
According to another embodiment, the invention provides a pharmaceutical composition comprising compound of Formula I, or pharmaceutically acceptable derivative thereof together with one or more pharmaceutically acceptable carrier, diluent, or excipient.
According to another embodiment, the invention provides a method of treatment of a condition mediated by inappropriate activity of at least one IGF-1 R family receptor in a mammal in need thereof, with a compound of Formula I, or pharmaceutically acceptable derivative thereof.
According to another embodiment, the invention provides a method for treating a susceptible neoplasm in a mammal in need thereof, comprising: administering to the mammal, a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable derivative thereof.
According to another embodiment, the invention provides a method for treating a condition selected from breast cancer, sarcomas, lung cancer (including non-small cell lung carcinoma), prostate cancer, colorectal cancer, renal cancer, pancreatic cancer, hematologic cancers (including multiple myeloma), neuroblastomas, gliomas, head and neck cancer, thyroid cancer, hepatocarcinoma, ovarian cancer, vulval cancer, cervical cancer, endometrial cancer, testicular cancer, bladder cancer, esophageal cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, gastrointestinal stromal tumor and skin cancer (including melanoma) in a mammal in need thereof, comprising: administering to the mammal, a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable derivative thereof.
According to another embodiment, the invention provides a method for treating a condition selected from breast cancer, sarcoma, lung cancer, non-small cell lung carcinoma, prostate cancer, colorectal cancer, pancreatic cancer, hematologic cancers, multiple myeloma, head and neck cancer or ovarian cancer in a mammal in need thereof, comprising: administering to the mammal, a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable derivative thereof. According to another embodiment, the invention provides a compound of
Formula I, or a pharmaceutically acceptable derivative thereof for use in the treatment of a condition mediated by inappropriate activity of at least one IGF-1 R family receptor.
According to another embodiment, the invention provides a compound of Formula I, or a pharmaceutically acceptable derivative thereof for use in the treatment of a susceptible neoplasm in a mammal in need thereof.
According to another embodiment, the invention provides a compound of Formula I, or a pharmaceutically acceptable derivative thereof for use in the treatment of a condition selected from breast cancer, sarcomas, lung cancer (including non-small cell lung carcinoma), prostate cancer, colorectal cancer, renal cancer, pancreatic cancer, hematologic cancers (including multiple myeloma), neuroblastomas, gliomas, head and neck cancer, thyroid cancer, hepatocarcinoma, ovarian cancer, vulval cancer, cervical cancer, endometrial cancer, testicular cancer, bladder cancer, esophageal cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, gastrointestinal stromal tumor and skin cancer (including melanoma) in a mammal in need thereof.
According to another embodiment, the invention provides a compound of Formula I, or a pharmaceutically acceptable derivative thereof for use in the treatment of a condition selected from breast cancer, sarcoma, lung cancer, non-small cell lung carcinoma, prostate cancer, colorectal cancer, pancreatic cancer, hematologic cancers, multiple myeloma, head and neck cancer or ovarian cancer in a mammal in need thereof. According to another embodiment, the invention provides the use of a compound of formula I, or pharmaceutically acceptable derivative thereof in the manufacture of a medicament for use in the treatment of a condition mediated by inappropriate activity of at least one IGF-1 R family receptor. According to another embodiment, the invention provides the use of a compound of formula I, or pharmaceutically acceptable derivative thereof in the manufacture of a medicament for use in the treatment of a susceptible neoplasm in a mammal in need thereof.
According to another embodiment, the invention provides the use of a compound of formula I, or pharmaceutically acceptable derivative thereof in the manufacture of a medicament for use in the treatment of a condition selected from breast cancer, sarcomas, lung cancer (including non-small cell lung carcinoma), prostate cancer, colorectal cancer, renal cancer, pancreatic cancer, hematologic cancers (including multiple myeloma), neuroblastomas, gliomas, head and neck cancer, thyroid cancer, hepatocarcinoma, ovarian cancer, vulval cancer, cervical cancer, endometrial cancer, testicular cancer, bladder cancer, esophageal cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, gastrointestinal stromal tumor and skin cancer (including melanoma) in a mammal in need thereof. According to another embodiment, the invention provides the use of a compound of formula I, or pharmaceutically acceptable derivative thereof in the manufacture of a medicament for use in the treatment of a condition selected from breast cancer, sarcoma, lung cancer, non-small cell lung carcinoma, prostate cancer, colorectal cancer, pancreatic cancer, hematologic cancers, multiple myeloma, head and neck cancer or ovarian cancer in a mammal in need thereof. According to another embodiment, the invention provides for a process for preparing a compound of formula I, comprising the steps of reacting a compound of formula (III),
Figure imgf000011_0001
with a compound of formula (II),
Figure imgf000012_0001
in the presence of a tertiary amine to form intermediate (V)
Figure imgf000012_0002
wherein L1 and L2 are leaving groups, X is a protecting group, and R1, R2, and R3 are as described above; then reacting intermediate (V) with a compound of formula (IV)
Figure imgf000012_0003
in the presence of an acid, a catalyst, and a polar protic solvent having low nucleophilicity, under heat, wherein R7, R8, R9, and R10 are as described above; then reacting the product of the preceding step with an amine-containing heteroatom nucleophile in solvent; and then removing the protecting group X with a base in solvent. DETAILED DESCRIPTION OF THE INVENTION
As used herein, the terms "alkyl" (and "alkylene") refer to straight or branched hydrocarbon chains containing from 1 to 6 carbon atoms, unless a different number of atoms is specified. Examples of "alkyl" as used herein include, but are not limited to, methyl, ethyl, n-propyl, n-butyl, n-pentyl, isobutyl, isopropyl, and tert-butyl. Examples of "alkylene" as used herein include, but are not limited to, methylene, ethylene, propylene, butylene, and isobutylene. "Alkyl" also includes substituted alkyl. The alkyl (and alkylene) groups may be optionally substituted one or more times with a halogen or hydroxyl. Thus, the term "alkyl" may include for example, trifluoromethyl and trifluoroethyl, among other halogenated alkyls, and hydroxymethyl and other hydroxylated alkyls when specified.
As used herein, the term "alkenyl" (and "alkylene") refers to straight or branched hydrocarbon chains containing from 2 to 6 carbon atoms, unless a different number of atoms is specified, and at least one and up to three carbon-carbon double bonds. Examples of "alkenyl" as used herein include, but are not limited to ethenyl and propenyl. Examples of "alkenylene" as used herein include, but are not limited to, ethenylene, propenylene and butenylene. "Alkenyl" (and "alkenylene") also may include substituted alkenyl. The alkenyl groups may optionally be substituted one or more times with a halogen or hydroxyl, as specified. As used herein, the term "alkynyl" refers to straight or branched hydrocarbon chains containing from 2 to 6 carbon atoms, unless a different number of atoms is specified, and at least one and up to three carbon-carbon triple bonds. Examples of "alkynyl" as used herein include, but are not limited to ethynyl and propynyl. "Alkynyl" may also include substituted alkynyl. The alkynyl groups may optionally be substituted one or more times with a halogen or hydroxyl, as specified.
As used herein, the term "cycloalkyl" refers to a saturated monocyclic carbocyclic ring having from 3 to 6 carbon atoms, unless a different number of atoms is specified. "Cycloalkyl" includes by way of example cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl. "Cycloalkyl" also includes substituted cycloalkyl. The cycloalkyl may optionally be substituted on any available carbon with one or more substituents selected from the group consisting of alkoxy, halo, and haloalkyl, e.g., perfluoroalkyl.
The term "halo" or "halogen" refers to fluoro, chloro, bromo and iodo. As used herein, the term "alkoxy" refers to the group -O-alkyl, where alkyl is as defined above. Examples of "alkoxy" as used herein include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, and t-butoxy. "Alkoxy" also includes substituted alkoxy. The alkoxy groups may be optionally substituted one or more times with a halogen.
The term "aryl" refers to monocyclic carbocyclic groups and fused bicyclic carbocyclic groups having from 6 to 10 carbon atoms, unless a different number of atoms is specified, and having at least one aromatic ring. Examples of particular aryl groups include but are not limited to phenyl and naphthyl. One particular aryl group according to the invention is phenyl.
The term "heterocycle", "heterocyclic ring", or "heterocyclyl" refers to a mono- or poly-cyclic ring system containing optionally one or more degrees of unsaturation and also containing one or more heteroatoms. Heteroatoms may include N, O, and/or S. Preferred heteroatoms are N and O, particularly N. The heterocycle is three to ten- membered and is either saturated or has one or more degrees of unsaturation. Heterocycles may be optionally fused to one or more of another heterocyclic ring, heteroaryl ring, aryl ring, or cycloalkyl ring. Examples of heterocyclic groups include, e.g. indole, indoline, isoquinoline.
The term "pharmaceutically acceptable derivative" refers to salts and solvates of the selected compound.
The term "solvate" as used herein refers to a complex of variable stoichiometry formed by a solute (a compound of formula (I)) and a solvent. Such solvents for the purpose of the invention may not interfere with the biological activity of the solute. Examples of suitable solvents include, but are not limited to, water, methanol, ethanol and acetic acid. Preferably the solvent used is a pharmaceutically acceptable solvent. Examples of suitable pharmaceutically acceptable solvents include, without limitation, water, ethanol and acetic acid. Most preferably the solvent used is water. As stated above, according to the main embodiment, the present invention provides a compound of formula (I):
Figure imgf000015_0001
or a pharmaceutically acceptable salt or solvate thereof wherein:
R4a and R4b are each independently selected from H, alkylCi-c6, -OH, -alkyleneci-cβ-OH, -alkyleneCi-c6-phenyl (optionally substituted with a halo), and -alkyleneCi-c6-C(O)NH2; one or two of X1, X2, and X3 are independently selected from N, O, and S, while the remaining one or two of X1, X2, and X3 are C; m is O or 1 ; R1 is selected from H, halo, alkylCi-c6, -alkyleneci-cs-halOi-s, and -O-alkylCi-c6 when X1 is
C, otherwise R1 is absent;
R2 is selected from H, halo, alkylCi-c6, -alkyleneci-cs-halOi-s, and -O-alkylCi-c6 when X2 is C, otherwise R2 is absent;
R3 is selected from H, halo, alkylCi-c6, -alkyleneci-cs-haloi-s, and -O-alkylCi-c6 when X3 is
C, otherwise R3 is absent; R7 is selected from alkylCi-c6, -0-alkylCi-c6, halo, -NR22R23, and -O-alkyleneCi-c6-halo1-3;
R8 is selected from H, halo, and alkylCi-c6; one of R9 and R10 is selected from -alk <yylleenneci-c6-SO2-alkylCi-c6,
-N-alkyleneCO-c6-C(0)-alkyleneco-c6-NR22R23, -0-alkyleneCo-c6(optionally substituted with -OH)-NR22R23,
Figure imgf000015_0002
(R14)i-2 , and (R14)i-2; and the other of R9 and R10 is selected from H, alkylCi-c6, -O-alkylCi-c6, and halo;
wherein Het1 and Het2 are each independently a five or six membered heterocyclic ring having an N atom and optionally one or two additional heteroatoms selected from N and O, and each R14 is independently selected from H, OH, halo, alkylCi-c6, -0-alkylCi-c6, -cyclopropyl, -C(O)-alkylCi-c6, SO2-alkylCi-c6, -(CH2)i-4-halo ,and -(CH2)1-4-SO2-alkylci-c6; -C(O )-alkyleneCo-c6-N R22R23, -NR22R23; or R9 and R10, together with the atoms to which they are attached form a five, six, or seven- membered heterocyclic ring containing one or two N atom and the remainder C atoms, wherein at least one N atom is substituted with R15, and the C atoms of the heterocyclic ring are optionally substituted with (R19)i-2;
wherein R15 is selected from H, -alkylCi-c4, -alkyleneCi-c4-halo,
-C(O)-alkyleneC0-c6-N R22R23, -C(O)-alkylci-c6, -alkyleneCi-c4-N R22R23, -alkyleneCi-c4-C(O)-NR22R23, -C(O)-alkyleneCi-c4-O-alkylCi-c6, -C(O)-pyrrolidine, and -C(O)-pyrrolidine-alkylci-c6; and, each R19 is independently selected from H and alkylCi-c6; each R22 is selected from H, alkylCi-c6, -O-alkylCi-c6, -alkyleneci-cδ-O-alkylci-ce; and,
each R23 is selected from H and alkylCi-c6; or
R22 and R23 combine to form a four, five, or six membered, heterocyclic ring containing the N atom to which they are attached and optionally an additional heteroatom selected from N and O, wherein the ring is optionally substituted with -OH or -alkyb-cβ.
According to an alternative embodiment of the invention, R7 is -0-alkylCi-c6 and the remaining substituents are as described above in the main embodiment.
According to another embodiment, R7 is -O-alkylCi-c6, R10 is H, and the remaining substituents are as described above in the main embodiment. According to another embodiment, R7 is -O-alkylCi-c6, m is 0, X1 is S, X2 and X3 are C, and the remaining substituents are as described above in the main embodiment.
According to another embodiment, R7 is -O-alkylCi-c6; m is 0, X1 is O, X2 and X3 are C, and the remaining substituents are as described above in the main embodiment.
According to another embodiment, R7 is -O-alkylCi-c6; m is 0, X1 is C, X2 is S, X3 is C, and the remaining substituents are as described above in the main embodiment.
According to another embodiment, R7 is -O-alkylCi-c6; m is 1 , X1 is N, X2 and X3 are C, and the remaining substituents are as described above in the main embodiment.
According to another embodiment, the substituents are as described in the above embodiments, respectively, wherein R7 is -O-methyl.
According to another embodiment, R5 and R6 are H, and the remaining substituents are as described above in the main embodiment.
According to another embodiment, the compound is of formula (Ia):
Figure imgf000017_0001
wherein the substituents are as described above in the main embodiment. According to another embodiment, the compound is of formula (Ib):
Figure imgf000018_0001
wherein the substituents are as described above in the main embodiment. According to another embodiment, the compound is of formula (Ic):
Figure imgf000018_0002
wherein the substituents are as described above in the main embodiment. According to another embodiment, the compound is of formula (Id):
Figure imgf000018_0003
wherein the substituents are as described above in the main embodiment. According to another embodiment, R10 is H and R9 is selected from
Figure imgf000019_0001
1-2 and
Figure imgf000019_0002
\ and the remaining substituents are as described above in the main embodiment.
According to another embodiment of the invention, R9 and R10, together with the atoms to which they are attached form a five, six, or seven-membered heterocyclic ring containing one or two N atom and the remainder C atoms, wherein at least one N atom is substituted with R15, and the C atoms of the heterocyclic ring are optionally substituted with one or more groups selected from (R19)i-2, and the remaining substituents are as described above in the main embodiment.
According to another embodiment, R9 and R10, together with the atoms to which they are attached form a five, six, or seven-membered heterocyclic ring containing one or two N atom and the remainder C atoms, wherein at least one N atom is substituted with R15, and the C atoms of the heterocyclic ring are optionally substituted with one or more groups selected from (R19)i-2, R7 is -O-alkylCi-c6, and the remaining substituents are as described above in the main embodiment.
According to an alternative embodiment of the invention, R9 and R10, together with the atoms to which they are attached form a five or six-membered heterocyclic ring selected from
Figure imgf000019_0003
According to another embodiment, R9 and R10, together with the atoms to which they are attached form a five or six-membered heterocyclic ring selected from
Figure imgf000020_0001
, R7 is -O-alkyld-ce, and the remaining substituents are as described above in the main embodiment.
With regard to each of the invented compounds, substitution at the R7 position is favorable and has generally been found to boost IGF1 R potency in comparison to otherwise similar compounds that are unsubstituted at the R7 position. Further, substitution at the R7 position has been found to increase selectivity over other kinases, for instance the Aurora A and Aurora B kinase, which is often desirable.
It is to be understood that the present invention includes all combinations and subsets of the particular groups defined herein, including the substituents as defined in the Summary defined hereinabove, as illustrated in the various examples throughout the specification, and as recited in the attached claims.
Specific examples of compounds of the present invention include those recited in the Examples which follow, and pharmaceutically acceptable salts or solvates thereof.
It will be appreciated by those skilled in the art that the compounds of the present invention may be utilized in the form of a pharmaceutically acceptable salt or solvate. The pharmaceutically acceptable salts of the compounds of formula (I) include conventional salts formed from pharmaceutically acceptable (i.e., non-toxic) inorganic or organic acids or bases as well as quaternary ammonium salts. Representative salts include the following: acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, calcium edetate, camsylate, carbonate, chloride, clavulanate, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, monopotassium maleate, mucate, napsylate, nitrate, N-methylglucamine, oxalate, pamoate (embonate), palmitate, pantothenate, phosphate/diphosphate, polygalacturonate, potassium, salicylate, sodium, stearate, subacetate, succinate, tannate, tartrate, teoclate, tosylate, triethiodide, trimethylammonium and valerate. Other salts, such as oxalic, which are not themselves pharmaceutically acceptable, may be useful in the preparation of salts useful as intermediates in obtaining compounds of this invention and these form a further aspect of the invention.
Processes for preparing pharmaceutically acceptable salts and solvates of compounds such as the compounds of formula (I) are conventional in the art. See, e.g., Burger's Medicinal Chemistry And Drug Discovery 5th Edition, VoI 1 : Principles And Practice.
As will be apparent to those skilled in the art, in the processes described below for the preparation of compounds of formula (I) certain intermediates may be in the form of pharmaceutically acceptable salts or solvates of the compound. Those terms as applied to any intermediate employed in the process of preparing compounds of formula (I) have the same meanings as noted above with respect to compounds of formula (I). Processes for preparing pharmaceutically acceptable salts and solvates of intermediates are known in the art and are analogous to the process for preparing pharmaceutically acceptable salts and solvates of compounds such as the compounds of formula (I). The compounds of this invention may be in crystalline or non-crystalline form, and, if crystalline, may optionally be hydrated or solvated. This invention includes within its scope stoichiometric hydrates as well as compounds containing variable amounts of water.
Certain compounds of formula (I) may exist in stereoisomeric forms (e.g. they may contain one or more asymmetric carbon atoms or may exhibit cis-trans isomerism). The individual stereoisomers (enantiomers and diastereomers) and mixtures of these are included within the scope of the present invention. The present invention also covers the individual isomers of the compounds represented by formula (I) as mixtures with isomers thereof in which one or more chiral centres are inverted. It is understood that compounds of formula (I) may exist in tautomeric forms other than that shown in the formula and these are also included within the scope of the present invention.
Since the compounds of formula (I) are intended for use in pharmaceutical compositions it will readily be understood that they are each preferably provided in substantially pure form, for example at least 60% pure, more suitably at least 75% pure and preferably at least 85%, especially at least 98% pure (% are on a weight for weight basis). Impure preparations of the compounds may be used for preparing the more pure forms used in the pharmaceutical compositions. The compounds of the present invention are inhibitors of one or more IGFR family receptors. By "IGFR inhibitor" is meant a compound which exhibits a pICso of greater than 5.5 against at least one IGFR family receptor in the IGFR inhibition enzyme assay described below (TR-FRET) and/or an IC5O of below about 1.0 μM potency against IGFR cellular autophosphorylation and/or in cell proliferation of a cell line that is dependent upon IGF signaling (e.g. Colo205, NCI-H929) in at least one of the cellular assay described below. In a more particular embodiment "IGFR inhibitor" refers to a compound which exhibits a plC50 of greater than 7.6 against at least one IGFR family receptor in the IGFR inhibition enzyme assay described below. In an alternative particular embodiment "IGFR inhibitor" refers to a compound which exhibits an IC50 of below about 250 nM potency against IGFR cellular autophosphorylation and/or in cell proliferation of a cell line that is dependent upon IGF signaling (e.g. Colo205, NCI-H929) in at least one of the cellular assay described below.
The present invention is not limited to compounds of formula (I) which are selective for IGFR family receptor kinases; rather, the present invention expressly contemplates compounds of formula (I) which may also possess activity against receptors in addition to the IGFR family receptors. For instance, the compounds of formula (I) are selective for insulin receptor (IR) family kinases. Several compounds of the present invention also possess activity against, for instance, one or more of the Jnk1 , Jnk2, and Jnk3, anaplastic lymphoma kinase (ALK), and insulin related receptor (IRR).
The present invention further provides compounds of formula (I) for use in medical therapy in a mammal, e.g. a human. In particular, the present invention provides compounds of formula (I) for use in the treatment of a condition mediated by at least one IGFR family receptor in a mammal, and, advantageously, conditions mediated by inappropriate activity of one or more IGFR family receptor in a mammal.
The inappropriate IGFR family receptor activity referred to herein is any IGFR receptor activity that deviates from the normal IGFR family receptor activity expected in a particular mammalian subject. Inappropriate IGFR family receptor activity may take the form of, for instance, an abnormal increase in activity, or an aberration in the timing and/or control of IGFR family receptor activity. Such inappropriate activity may result then, for example, from overexpression or mutation of the protein kinase or ligand leading to inappropriate or uncontrolled activation of the receptor. Furthermore, it is also understood that unwanted IGFR family receptor activity may reside in an abnormal source, such as a malignancy. That is, the level of IGFR family activity does not have to be abnormal to be considered inappropriate, rather the activity derives from an abnormal source. The compounds of formula (I) and salts and solvates thereof, are believed to have anticancer and antitumor activity as a result of inhibition of one or more IGFR family receptor and its effect on selected cell lines whose growth is dependent on IGFR family activity.
The present invention provides compounds of formula (I) for use in the treatment of a susceptible neoplasm. "Susceptible neoplasm" as used herein refers to neoplasms which are susceptible to treatment with a IGFR inhibitor. Neoplasms which have been associated with inappropriate activity of one or more IGFR family receptors and are therefor susceptible to treatment with a IGFR inhibitor are known in the art, and include both primary and metastatic tumors and cancers. For example, susceptible neoplasms within the scope of the present invention include but are not limited to breast cancer, sarcomas, lung cancer (including non-small cell lung carcinoma), prostate cancer, colorectal cancer, renal cancer, pancreatic cancer, hematologic cancers (including multiple myeloma), neuroblastomas, gliomas, head and neck cancer, thyroid cancer, hepatocarcinoma, ovarian cancer, vulval cancer, cervical cancer, endometrial cancer, testicular cancer, bladder cancer, esophageal cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, gastrointestinal stromal tumor, and skin cancer (including melanoma). More particularly, susceptible neoplasms may be selected from breast cancer, sarcoma, lung cancer, non-small cell lung carcinoma, prostate cancer, colorectal cancer, pancreatic cancer, hematologic cancers, multiple myeloma, head and neck cancer, and ovarian cancer.
The present invention provides methods for the treatment of several conditions in a mammal in need thereof, all of which comprise the step of administering a therapeutically effective amount of a compound of formula (I). The mammal in need of treatment with a compound of the present invention is advantageously a human. As used herein, the term "treatment" refers to alleviating the specified condition, eliminating or reducing the symptoms of the condition, slowing or eliminating the progression of the condition and preventing or delaying the reoccurrance of the condition in a previously afflicted subject. As used herein, the term "therapeutically effective amount" means an amount of a compound of formula (I) which is sufficient, in the subject to which it is administered, to elicit the biological or medical response of a cell culture, tissue, system, mammal (including human) that is being sought, for instance, by a researcher or clinician. The term also includes within its scope amounts effective to enhance normal physiological function. For example, a therapeutically effective amount of a compound of formula (I) for the treatment of a condition mediated by at least one IGFR family receptor is an amount sufficient to treat the condition in the subject. Similarly, a therapeutically effective amount of a compound of formula (I) for the treatment of a susceptible neoplasm is an amount sufficient to treat the susceptible neoplasm in the subject. In one embodiment of the present invention, a therapeutically effective amount of a compound of formula (I) is an amount sufficient to regulate, modulate, bind or inhibit at least one IGFR family receptor.
The precise therapeutically effective amount of the compounds of formula (I) will depend on a number of factors including, but not limited to, the age and weight of the subject being treated, the precise condition requiring treatment and its severity, the nature of the formulation, and the route of administration, and will ultimately be at the discretion of the attendant physcian or veternarian. Typically, the compound of formula (I) will be given for treatment in the range of 0.1 to 200 mg/kg body weight of recipient (mammal) per day and more usually in the range of 1 to 100 mg/kg body weight per day. Acceptable daily dosages, may be from about 0.1 to about 2000 mg/day, and preferably from about 0.1 to about 100 mg/day. Thus, for a 70kg adult human being treated for a condition mediated by at least one IGFR family receptor, the actual amount per day would usually be from 5 to 700 mg and this amount may be given in a single dose per day or more usually in a number (such as two, three, four, five or six) of sub-doses per day or, alternatively, on an alternative dosing schedule such as weekly or monthly, such that the total daily dose is the same. A therapeutically effective amount of a salt or solvate, may be determined as a proportion of the therapeutically effective amount of the compound of formula (I) per se. It is envisaged that similar dosages would be appropriate for treatment of the other conditions referred to above.
The present invention also provides the use of a compound of formula (I) for the preparation of a medicament for the treatment of condition mediated by at least one IGFR family receptor in a mammal (e.g., a human) in need thereof. The present invention further provides the use of a compound of formula (I) for the preparation of a medicament for the treatment of a susceptible neoplasm in a mammal.
While it is possible that, for use in therapy, a therapeutically effective amount of a compound of formula (I) may be administered as the raw chemical, it is typically presented as the active ingredient of a pharmaceutical composition or formulation. Accordingly, the invention further provides a pharmaceutical composition comprising a compound of the formula (I). The pharmaceutical composition may further comprise one or more pharmaceutically acceptable carriers, diluents, and/or excipients. The carrier(s), diluent(s) and/or excipient(s) must be acceptable in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. In accordance with another aspect of the invention there is also provided a process for the preparation of a pharmaceutical formulation including admixing a compound of the formula (I) with one or more pharmaceutically acceptable carriers, diluents and/or excipients. Pharmaceutical formulations may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) route. Such formulations may be prepared by any method known in the art of pharmacy, for example by bringing into association the active ingredient with the carrier(s) or excipient(s).
Pharmaceutical formulations adapted for oral administration may be presented as discrete units such as capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in-oil liquid emulsions. For instance, for oral administration in the form of a tablet or capsule, the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like. Powders are prepared by comminuting the compound to a suitable fine size and mixing with a similarly comminuted pharmaceutical carrier such as an edible carbohydrate, as, for example, starch or mannitol. Flavoring, preservative, dispersing and coloring agent can also be present.
Capsules are made by preparing a powder mixture, as described above, and filling formed gelatin sheaths. Glidants and lubricants such as colloidal silica, talc, magnesium stearate, calcium stearate or solid polyethylene glycol can be added to the powder mixture before the filling operation. A disintegrating or solubilizing agent such as agar-agar, calcium carbonate or sodium carbonate can also be added to improve the availability of the medicament when the capsule is ingested. 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, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, 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. Tablets are formulated, for example, by preparing a powder mixture, granulating or slugging, adding a lubricant and disintegrant and pressing into tablets. A powder mixture is prepared by mixing the compound, suitably comminuted, with a diluent or base as described above, and optionally, with a binder such as carboxymethylcellulose, an aliginate, gelatin, or polyvinyl pyrrolidone, a solution retardant such as paraffin, a resorption accelerator such as a quaternary salt and/or an absorption agent such as bentonite, kaolin or dicalcium phosphate. The powder mixture can be granulated by wetting with a binder such as syrup, starch paste, acadia mucilage or solutions of cellulosic or polymeric materials and forcing through a screen. As an alternative to granulating, the powder mixture can be run through the tablet machine and the result is imperfectly formed slugs broken into granules. The granules can be lubricated to prevent sticking to the tablet forming dies by means of the addition of stearic acid, a stearate salt, talc or mineral oil. The lubricated mixture is then compressed into tablets. The compounds of the present invention can also be combined with a free flowing inert carrier and compressed into tablets directly without going through the granulating or slugging steps. A clear or opaque protective coating consisting of a sealing coat of shellac, a coating of sugar or polymeric material and a polish coating of wax can be provided. Dyestuffs can be added to these coatings to distinguish different unit dosages.
Oral fluids such as solution, syrups and elixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of the compound. Syrups can be prepared by dissolving the compound in a suitably flavored aqueous solution, while elixirs are prepared through the use of a non-toxic alcoholic vehicle. Suspensions can be formulated by dispersing the compound in a non-toxic vehicle. Solubilizers and emulsifiers such as ethoxylated isostearyl alcohols and polyoxy ethylene sorbitol ethers, preservatives, flavor additive such as peppermint oil or natural sweeteners or saccharin or other artificial sweeteners, and the like can also be added. Where appropriate, dosage unit formulations for oral administration can be microencapsulated. The formulation can also be prepared to prolong or sustain the release as for example by coating or embedding particulate material in polymers, wax or the like. The compounds of formula (I) may be administered by inhalation, such as by metered dose pressurised aerosols, metered dose inhalers, dry powder inhalers, nebulizers or insufflators.
According to one embodiment, the compound is provided in the form of a dry powder composition. As such, the composition is suitable for inhaled administration and may be incorporated into a plurality of sealed dose containers (e.g. containing the dry powder composition) mounted in a strip or ribbon inside a suitable inhalation device. The container is rupturable or peel-openable on demand and the dose of the dry powder composition may be administered by inhalation via a device such as the DISKUS™ device, marketed by GlaxoSmithKline. The DISKUS™ inhalation device is, for example, described in GB2242134A.
According to another embodiment, the compounds of formula (I) may be formulated into spray compositions for inhalation which may, for example, be formulated as aqueous solutions or suspensions or as aerosols delivered from pressurised packs, such as a metered dose inhaler (MDI), with the use of a suitable liquefied propellant. Aerosol compositions suitable for inhalation can be either a suspension or a solution and generally contain a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof and a suitable propellant such as a fluorocarbon or hydrogen-containing chlorofluorocarbon or mixtures thereof, particularly hydrofluoroalkanes, especially 1 ,1 ,1 ,2-tetrafluoroethane, 1 ,1 ,1 ,2,3,3,3-heptafluoro-n-propane or a mixture thereof. The aerosol composition may optionally contain additional formulation excipients well known in the art such as surfactants e.g. oleic acid, lecithin or an oligolactic acid derivative e.g. as described in WO94/21229 and WO98/34596 and cosolvents e.g. ethanol. The compounds of formula (I) can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines. The compounds of formula (I) may also be delivered by the use of monoclonal antibodies as individual carriers to which the compound molecules are coupled. The compounds may also be coupled with soluble polymers as targetable drug carriers. Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide -phenol, polyhydroxyethylaspartamidephenol, or polyethyleneoxidepolylysine substituted with palmitoyl residues. Furthermore, the compounds may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels. Pharmaceutical formulations adapted for transdermal administration may be presented as discrete patches intended to remain in intimate contact with the epidermis of the recipient for a prolonged period of time. For example, the active ingredient may be delivered from the patch by iontophoresis as generally described in Pharmaceutical Research, 3(6), 318 (1986). Pharmaceutical formulations adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils.
For treatments of the eye or other external tissues, for example mouth and skin, the formulations are preferably applied as a topical ointment or cream. When formulated in an ointment, the active ingredient may be employed with either a paraffinic or a water- miscible ointment base. Alternatively, the active ingredient may be formulated in a cream with an oil-in-water cream base or a water-in-oil base.
Pharmaceutical formulations adapted for topical administrations to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent.
Pharmaceutical formulations adapted for topical administration in the mouth include lozenges, pastilles and mouth washes. Pharmaceutical formulations adapted for rectal administration may be presented as suppositories or as enemas.
Pharmaceutical formulations adapted for nasal administration wherein the carrier is a solid include a coarse powder having a particle size for example in the range 20 to 500 microns which is administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close up to the nose. Suitable formulations wherein the carrier is a liquid, for administration as a nasal spray or as nasal drops, include aqueous or oil solutions of the active ingredient.
Pharmaceutical formulations adapted for administration by inhalation include fine particle dusts or mists, which may be generated by means of various types of metered, dose pressurised aerosols, nebulizers or insufflators.
Pharmaceutical formulations adapted for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations. Pharmaceutical formulations adapted for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation near isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
It should be understood that in addition to the ingredients particularly mentioned above, the formulations may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavouring agents.
In the above-described methods of treatment and uses, a compound of the invention may be employed alone, in combination with one or more other compounds of the invention, or in combination with other therapeutic methods or agents. In particular, in methods of treating a condition improved by inhibition of IGF-1 R and in methods of treating susceptible neoplasms, the compound of the invention may be used alone or in combination with one or more of a chemotherapeutic, a hormonal and/or antibody agent, surgical therapy, and radiotherapy.
The term "chemotherapeutic" as used herein refers to any chemical agent having a therapeutic effect on the subject to which it is administered. "Chemotherapeutic" agents include but are not limited to anti-neoplastic agents, analgesics and anti-emetics. As used herein, "anti-neoplastic agents" include both cytostatic and cytotoxic agents. Anti-emetics include but are not limited to 5HT3 antagonists such as ondansetron, granisetron, and the like; metaclopromide; dexamethasone and neurokinin-1 antagonists. As an additional aspect, the present invention provides the methods of treatment and uses as described above, which comprise administering a compound of the invention together with at least one chemotherapeutic agent. In one particular embodiment, the chemotherapeutic agent is an anti-neoplastic agent. In another embodiment, the present invention provides a pharmaceutical composition as described above further comprising at least one other chemotherapeutic agent, more particularly, the chemotherapeutic agent is an anti-neoplastic agent.
The compounds of the invention and at least one additional anti-neoplastic therapy may be employed in combination concomitantly or sequentially in any therapeutically appropriate combination. The administration of a compound of the invention with one or more other anti-neoplastic agents may be in combination in accordance with the invention by administration concomitantly in (1 ) a unitary pharmaceutical composition including both or all compounds or (2) separate pharmaceutical compositions each including one of the compounds. The combination may be administered separately in a sequential manner wherein one anti-neoplastic agent is administered first and the other(s) second or vice versa. Such sequential administration may be close in time or remote in time.
When a compound of the invention is used in combination with a chemotherapeutic agent, the dose of each compound may differ from that when the compound is used alone. Appropriate doses will be readily appreciated by those skilled in the art. The appropriate dose of the compound(s) of the invention and the other therapeutically active agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect, and are within the expertise and discretion of the attendent clinician. Among the many chemotherapeutic agents, which may be used in combination with a compound of the present invention, are anti-proliferative/anti-neoplastic agents. Anti-neoplastic agents may induce anti-neoplastic effects in a cell-cycle specific manner, i.e., are phase specific and act at a specific phase of the cell cycle, or bind DNA and act in a non cell-cycle specific manner, i.e., are non-cell cycle specific and operate by other mechanisms. Both types of anti-neoplastic agents may be employed in combination with the compounds of the present invention.
Typically, any chemotherapeutic agent that has activity against a susceptible neoplasm being treated may be utilized in combination with the compounds the invention, provided that the particular agent is clinically compatible with therapy employing a compound of the invention. Typical anti-neoplastic agents useful in the present invention include, but are not limited to: alkylating agents, anti-metabolites, antitumor antibiotics, antimitotic agents, topoisomerase I and Il inhibitors, hormones and hormonal analogues, matrix metalloprotease inhibitors; signal transduction pathway inhibitors including inhibitors of cell growth or growth factor function, angiogenesis inhibitors, and serine/threonine inhibitors; cyclin dependent kinase inhibitors; antisense therapies and immunotherapeutic agents.
Examples of alkylating agents include but are not limited to: nitrogen mustards such as cyclophosphamides, temozolamide, melphalan, and chlorambucil; oxazaphosphor-ines; alkyl sulfonates such as busulfan; nitrosoureas such as carmustine; triazenes such as dacarbazine; and platinum coordination complexes such as cisplatin, oxapliplatin and carboplatin.
Examples of antimetabolite anti-neoplastic agents include purine and pyrimidine analogues and anti-folate compounds, and more specifically, hydroxyurea, cytosine, arabinoside, raltitrexed, tegafur, fluorouracil (e.g., 5FU), methotrexate, cytarabine, mecaptopurine and thioguanine.
Examples of antitumor antibiotic agents include, but are not limited to, actinomycins such as dactinomycin, mitomycin-C, anthracyclins such as daunorubicin, doxorubicin, idarubicin, epirubicin; daunomycin, adriamycin and bleomycins. Examples of antimitotic agents include, but are not limited to, diterpenoids, vinca alkaloids, polo-like kinase (PLK) inhibitors and CenpE inhibitors. Examples of diterpenoids include, but are not limited to, taxol, taxotere, paclitaxel and its analog docetaxel. Examples of vinca alkaloids include, but are not limited to, vinblastine, vincristine, vindesine and vinorelbine. PLK inhibitors are discussed further below.
Examples of topoisomerase I inhibitors include camptothecins, such as amsacrine, irinotecan, topotecan, and the various optical forms of 7-(4-methylpiperazino- methylene)-10,11-ethylenedioxy-20-camptothecin. Examples of topoisomerase Il inhibitors include epipodophyllotoxins, such as etoposide and teniposide.
Examples of hormones and hormonal analogues believed to be useful in the treatment of neoplasms include, but are not limited to: antiestrogens, such as tamoxifen, toremifene, raloxifene, fulvestrant, iodoxyfene and droloxifene; anti-androgens; such as flutamide, nilutamide, bicalutamide and cyproterone acetate; adrenocorticosteroids such as prednisone and prednisolone; aminoglutethimide and other aromatase inhibitors such as anastrozole, letrazole, vorazole, and exemestane; progestrins such as megestrol acetate; 5α-reductase inhibitors such as finasteride and dutasteride; and gonadotropin-releasing hormones (GnRH) and analogues thereof, such as Leutinizing Hormone-releasing Hormone (LHRH) agonists and antagagonists such as goserelin luprolide, leuprorelin and buserelin.
An example of a matrix metalloproteinases (MMP) inhibitor is marimastat. Signal transduction pathway inhibitors useful in the present invention include, but are not limited to, inhibitors of receptor tyrosine kinases, non-receptor tyrosine kinases, SH2/SH3 domain blockers, serine/threonine kinases, phosphatidyl inositol-3-OH kinases, myo-inositol signaling, and Ras oncogenes (e.g. farnesyltransferase, geranyl- geranyl transferase, and CAAX proteases as well as anti-sense oligonucleotides, ribozymes and immunotherapy).
Several protein tyrosine kinases catalyse the phosphorylation of specific tyrosyl residues in various proteins involved in the regulation of cell growth. Such protein tyrosine kinases can be broadly classified as receptor or non-receptor kinases.
Examples of receptor tyrosine kinases, also known as "growth factor receptor inhibitors", in addition to IGF-1 R inhibitors, include but are not limited to inhibitors of: epidermal growth factor family receptors (EGFR, ErbB2, and ErbB4); platelet derived growth factor receptors (PDGFRs), vascular endothelial growth factor receptors (VEGFRs), tyrosine kinase with immunoglobulin-like and epidermal growth factor homology domains (TIE-2), macrophage colony stimulating factor (c-fms), c-kit, c-met, fibroblast growth factor receptors (FGFRs), hepatocyte growth factor receptors (HGFRs), Trk receptors (TrkA, TrkB, and TrkC), ephrin (Eph) receptors, the RET protooncogene, and Akt kinases.
Several inhibitors of growth factor receptors are under development and include ligand antagonists, antibodies, tyrosine kinase inhibitors, anti-sense oligonucleotides and aptamers. Any of these growth factor receptor inhibitors may be employed in combination with the compounds of the present invention in any of the compositions and methods/uses described herein. Trastuzumab (Herceptin®) is an example of an anti- erbB2 antibody inhibitor of growth factor function. One example of an anti-erbB1 antibody inhibitor of growth factor function is cetuximab (Erbitux™, C225). Examples of small molecule inhibitors of epidermal growth factor receptors include but are not limited to lapatinib (Tykerb™), erlotinib (TARCEVA®), gefitinib (IRESSA®), canetinib or CU 033. lmitanib is one example of a PDGFR inhibitor. Examples of VEGFR inhibitors include pazopanib, ZD6474, AZD2171 , PTK787, SU1 1248 and sunitinib.
Tyrosine kinases that are not growth factor receptor kinases are termed non- receptor tyrosine kinases. Inhibitors of non-receptor tyrosine kinases are sometimes referred to as "anti-metastatic agents" and are useful in the present invention. Targets or potential targets of anti-metastatic agents, include, but are not limited to, c-Src, Lck, Fyn, Yes, Jak, abl kinase (c-Abl and Bcr-Abl), FAK (focal adhesion kinase) and Bruton's tyrosine kinase (BTK). Non-receptor kinases and agents, which inhibit non-receptor tyrosine kinase function, are described in Sinha, S. and Corey, S. J., (1999) J.
Hematother. Stem Cell Res. 8:465-80; and Bolen, J. B. and Brugge, J.S., (1997) Annu. Rev. of Immunol. 15:371-404.
SH2/SH3 domain blockers are agents that disrupt SH2 or SH3 domain binding in a variety of enzymes or adaptor proteins including, but not limited to, PI3-K p85 subunit, Src family kinases, adaptor molecules (She, Crk, Nek, Grb2) and Ras-GAP. Examples of Src inhibitors include but are not limited to dasatinib and BMS-354825 (J.Med.Chem (2004) 47:6658-6661 ).
Examples of serine/threonine kinase inhibitors include, but are not limited to polo- like kinase inhibitors (PIk family e.g., PIkI , Plk2, and Plk3),such as 5-{6-[(4- Methylpiperazin-1-yl)methyl]-1 H-benzimidazol-1-yl}-3-{(1 R)-1-[2-
(trifluoromethyl)phenyl]ethoxy}thiophene-2-carboxamide; MAP kinase cascade blockers, which include Ras/Raf kinase inhibitors, mitogen or extracellular regulated kinases (MEKs), and extracellular regulated kinases (ERKs); Aurora kinase inhibitors (including inhibitors of Aurora A and Aurora B); protein kinase C (PKC) family member blockers; inhibitors of kappa-B (IkB) kinase family (IKK-alpha, IKK-beta); PKB/Akt kinase family inhibitors; and inhibitors of TGF-beta receptor kinases. Examples of PIk inhibitors are described in PCT Publication No. WO04/014899 to GlaxoSmithKline. Inhibitors of urokinase, also referred to as urokinase-type Plasminogen Activator
(uPA), expression may be used in combination with the compounds of the present invention in the compositions and methods described above.
Inhibitors of kinases involved in the IGF signalling axis may also be useful in combination with the compounds of the present invention. Such inhibitors include but are not limited to inhibitors of JNK1/2/3, PI3K, AKT and MEK, and 14.3.3 signalling inhibitors.
Cell cycle signaling inhibitors, including inhibitors of cyclin dependent kinases (CDKs) are also useful in combination with the compounds of the invention in the compositions and methods described above. Examples of cyclin dependent kinases, including CDK2, CDK4, and CDK6 and inhibitors for the same are described in, for instance, Rosania G. R., et al., Exp. Opin. Ther. Patents (2000) 10:215-230.
Receptor kinase angiogenesis inhibitors may also find use in the present invention. Inhibitors of angiogenesis related to VEGFR and TIE-2 are discussed above in regard to signal transduction inhibitors (both are receptor tyrosine kinases). Other inhibitors may be used in combination with the compounds of the present invention. For example, anti-VEGF antibodies, which do not recognize VEGFR (the receptor tyrosine kinase), but bind to the ligand; small molecule inhibitors of integrin (alphav beta3) that inhibit angiogenesis; endostatin and angiostatin (non-RTK) may also prove useful in combination with the compounds of the invention. One example of a VEGFR antibody is bevacizumab (Avastin™).
Inhibitors of phosphotidyl inositol-3-OH kinase family members including blockers of PI3-kinase, ATM, DNA-PK, and Ku may also be useful in combination with the present invention.
Also of potential use in combination with the compounds of the invention are myo-inositol signaling inhibitors such as phospholipase C blockers and myoinositol analogues. Examples of antisense therapies include those directed towards the targets described above such as ISIS 2503 and gene therapy approaches such as those using thymidine kinase or cytosine deaminase.
Agents used in immunotherapeutic regimens may also be useful in combination with the compounds of the invention. Immunotherapeutic regimens include ex-vivo and in-vivo approaches to increasing immunogenicity of patient tumor cells such as transfection with cytokines (IL-2, IL-4, GMCFS and MCFS), approaches to increase T- cell activity, approaches with transfected immune cells and approaches with anti- idiotypic antibodies. Agents used in proapoptotic regimens (e.g., Bcl-2 antisense oligonucleotides) may also be used in combination with the compounds of the invention. Members of the Bcl-2 family of proteins block apoptosis. Upregulation of Bcl-2 has therefore been linked to chemoresistance. Studies have shown that the epidermal growth factor (EGF) stimulates anti-apoptotic members of the Bcl-2 family (i.e., mcl-1 ). Therefore, strategies designed to downregulate the expression of Bcl-2 in tumors have demonstrated clinical benefit and are now in Phase ll/lll trials, namely Genta's G3139 bcl-2 antisense oligonucleotide. Such proapoptotic strategies using the antisense oligonucleotide strategy for Bcl-2 are discussed in Water, J.S., et al., J. Clin. Oncol. (2000) 18:1812- 1823; and Kitada, S., et al., Antisense Res. Dev. (1994) 4:71-79. Compounds of formula (I) may be prepared using the processes described below. In all of the schemes described below, it is understood that protecting groups may be employed where necessary in accordance with general principles known to those of skill in the art, for example, see T. W. Green and P. G. M. Wuts (1991 ) Protecting Groups in Organic Synthesis, John Wiley & Sons. These groups may be removed at a convenient stage of the compound synthesis using methods known to those of skill in the art. The selection of processes as well as the reaction conditions and order of their execution shall be consistent with the preparation of compounds of formula (I).
The invention also provides a process for preparing the compound of formula (I) and pharmaceutically acceptable derivatives thereof. Specifically, compounds of formula (I) are prepared by reacting the three main components of the compounds, referred to herein as the head, core, and tail of the compounds. Synthesis and Schemes
Each compound of formula (I) may be conveniently prepared by separately preparing three constituents of the compound and subsequently combining those constituents to form the compound (I). For convenience, the three constituents are referred to herein as the head (II), the core (III), and the tail (IV). For convenience, the head, core, and tail nomenclature is used throughout to refer to each constituent when referred to individually, and also to refer to the corresponding moiety when described in the context of head/core, tail/core, and/or head/core/tail combinations.
The head component of the invented compounds is an o-amino carboxamide represented by formula (II):
Figure imgf000036_0001
wherein substituents R1, R2, and R3 are as defined above. The core component of the invented compounds is pyrrolopyrimidine represented by formula (III):
Figure imgf000036_0002
wherein each of substituents L1 and L2 represent a leaving group, e.g. a halogen, preferably chlorine, or OTF, and X represents a protecting group, e.g. sulfonamide or alcholated alkyl, e.g. allyl, benzyl, or SEM.
The tail component of the invented compounds is a substituted aniline represented by formula (IV):
Figure imgf000036_0003
wherein substituents R7, R8, R9, and R10 are as defined above. The head (II), core (III), and tail (IV) of the compounds may be synthesized and combined, for instance, with the synthetic routes shown below:
Scheme 1 Preparation of Aryl Precursors
Figure imgf000037_0001
I-6 I-7
As shown in Scheme 1 , some requisite aniline precursors can be prepared from commercially available phenols such as 1-1 (R = Me & Cl from 3B Scientific Corporation) and I-6 (VWR International). Nitration of phenol 1-1 in a solvent (i.e. dichloromethane or dichloroethane), with a nitrating agent (i.e. nitric acid) and phase-transfer catalyst
(tetrabutylammonium bromide) affords the corresponding nitro-benzene I-2. Alkylation with an alkyl halide (i.e. methyl iodide, ethyl iodide), base (potassium carbonate) and solvent (typically DMF) affords precursors I-3, I-4, and I-5. Alternatively, acetophenone I-6 can be preferentially reduced at the carbonyl carbon (preferably zinc dust/HCI), and subsequently nitrated and alkylated using analogous conditions described for 1-1 , to afford I -7. Scheme 2 Tails derived via SNAR Displacements
Figure imgf000038_0001
As shown in Scheme 2, a variety of substituted anilines can be prepared via direct SNAR displacement of 4-fluoro nitrobenzenes I-3, I-4, I-5, I-7, and 11-1 (commercially available through Oakwood Products) by addition of base (for example potassium carbonate), solvent (DMF), and a primary or secondary amine (including exemplary amines 11-10 through 11-14.) Subsequent selective nitro reduction using a catalyst (iron (III) chloride or palladium on carbon), additive (activated carbon), alcoholic or ethereal solvent (MeOH, EtOH, or THF), and reducing agent (hydrogen gas or hydrazine) affords the desired anilines II-8. Alternatively SNAR displacement with a versatile multifunctional amine (including N-Boc piperazine II-3) affords Boc protected nitrobenzene II-2. Boc deprotection with acid (i.e. hydrochloric or trifluoroacetic acid), followed by alkylation at nitrogen with base and an electrophile (potassium carbonate, iso-butyl iodide) or acylation at nitrogen with a base and an electrophile followed by displacement with an amine (triethyl amine, α-CI acetyl bromide, then dimethyl amine) gives the corresponding nitro-benzenes which may be reduced as described for II-8 to afford the corresponding functionalized anilines 11-15 and 11-16. Alternatively SNAR displacement with amino-alcohol 11-5 followed by alcohol oxidation using an oxidant (for example Dess-Martin periodinane or the Swern reagent) affords ketone 11-6. Reductive amination with a buffered proton source (for example acetic acid with triethyl amine), solvent (dichloroethane), and amine (for example exemplary amine 11-17) followed by nitro reduction as described above for intermediate II-8 or with a hydride source (for example sodium borohydride) with added metal (for example NiC^) in alcoholic solvent (MeOH) affords the corresponding functionalized aniline II-9.
Scheme 3 Tails Derived from Organometallic Coupling Reactions
Buchwald nitro reduction Animation
Figure imgf000040_0002
Figure imgf000040_0001
Figure imgf000040_0003
Figure imgf000040_0004
11-2, R = H 11-3, R = Me
Figure imgf000040_0005
Alternatively similarly functionalized anilines may be generated from organometallic coupling reactions as illustrated in Scheme 3. Nitro-benenze 111-1 (see Hirst, Gavin C; Calderwood, David; Wishart, Neil; Rafferty, Paul; Ritter, Kurt; Arnold, Lee D.; Friedman, Michael M. Preparation of pyrazolopyrimidines as protein kinase inhibitors. PCT Int. Appl.(2001 ), 527 pp.) can be subjected to Buchwald-Hartwig amination conditions including typically a palladium catalyst (i.e. Pd2(dba)3), trialkyl amine or inorganic base (i.e. triethyl amine, potassium carbonate, cesium carbonate), a phosphine catalyst (i.e. xantphos), solvent (tertiary butanol, dimethyl formamide, dioxane) and an amine (for example isopropyl piperazine as above) to afford the nitrogen coupled product III-4. Nitro reduction utilizing a metal catalyst (i.e. iron (III) chloride), additive (activated carbon), alcoholic solvent (i.e. methanol) and reducing agent (hydrazine) affords functionalized aniline III-5. Alternatively Suzuki coupling protocols involving addition of a palladium catalyst (i.e. Pd(PPh3)4), a boronic acid or ester (for example piperidyl 4-boronic acid as used in Scheme 3), a solvent mixture (often dioxane or THF in water ) with an inorganic base (for example potassium carbonate) affords functional nitrobenzene 111-6. Alkylation of 111-6 with an electrophile (for example propyl iodide) in high-boiling polar solvent (for example pinacolone) affords the corresponding pyridinium salt. Reduction of this intermediate typically is carried out in two steps, first via reduction of pyridinium by addition of a hydride source (sodium borohydride or sodium cyanoborohyride) in solvent and second via reduction of the in- situ generated olefin and nitro-group using catalyst (palladium on carbon) in an alcoholic or ethereal solvent (THF, MeOH, EtOH) under pressurized hydrogen gas (typically 30-50 psi) affords piperidine analog III-7. Alternatively, selective nitro reduction of III-6 can be carried out with a poisoned hydrogenation catalyst (for example Pt-C with sulphide) under a pressure of hydrogen gas (30-50 psi) to afford the corresponding aniline. Protection of this anline with a suitable protecting group (i.e. trifluoroacetic anhydride, triethyl amine) affords acetamide III-8. One-step exhaustive pyridine reduction can be effected with a catalyst (Pt-carbon) under hydrogen gas (30-50 psi) with alcoholic or ethereal solvent (typically MeOH, EtOH, THF) to give the corresponding piperidine. Subsequent alkylation with an electrophile (i.e. methyl-vinyl sulfone) in the presence of base (triethyl amine) including in situ aniline deprotection affords the desired aniline III-9.
Scheme 4 indohne-deπved Anilines
acylating agents or
Figure imgf000042_0001
(α-amιno acid)
Primary Route
1 hydride source, acid 1 amide bond formation
2 acylating agent, solvent
3 nitrating agent, acid
4 acid, alcohol solvent
Figure imgf000042_0003
IV-1
Figure imgf000042_0002
Figure imgf000042_0004
As shown in scheme 4, bicyclic indoline tails may be prepared using 5-methoxy indole (Aldrich) as a starting material. The 5-methoxy indole is first reduced using a hydride source, e.g. Na(CN)BH3, and acid, e.g. AcOH, then protected with an acylating agent, e.g. Ac2O, in solvent, e.g. AcOH, optionally with a base. The protection provides for subsequent selective nitration with nitrating agent, e.g. HNO3/AC2O, followed by deacylation with acid, e.g. HCI in solvent, e.g. MeOH to provide intermediate IV-2
Intermediate IV-2 is subjected to subsequent acylation with an acylating agent, e.g. α-bromoacetyl chloride or an amino acid, with coupling reagents, e.g. HATU, DMAP, and base, e.g. PS-DIPEA, and solvent, e.g. THF. The α-bromoacetyl product may be reacted with an appropriate amine base, e.g. Me2NH (with solvent, e.g. THF), and the product then reduced (see reduction of II-4, Scheme 2) to provide anilino indoline tails IV-3. Alternatively, the set of reaction described above for Scheme 4 can be reordered to allow for the elimination of the acylation/deacylation sequence. For example direct acylation of indoline IV-6 (commercially available as the HCI salt from ChemPacific) with an acylating agent (α-bromo-acetyl chloride) and base (optionally a trialkyl amine base such as triethyl amine or an inorganic base such as potassium carbonate) followed by treatment with a nucleophile (for example dimethyl amine) affords IV-7. Direct nitration of IV-7 using an acid (i.e. trifluoroacetic acid) and one equivalent of a nitrating agent (for example potassium or sodium nitrate) affords the corresponding nitro-indoline which may be reduced in analogy to the reduction of II-4 (Scheme 2) to afford functionalized aniline indoline IV-8.
Scheme 5 Additional indoline-derived tails
Figure imgf000044_0001
amide reduction
Figure imgf000044_0002
Figure imgf000044_0003
As shown in scheme 5, additional indoline tails may be prepared by alternative schemes. Chloro aniline V-3 is readily prepared via acylation of 6-nitro indoline with an acylating agent, e.g. α-bromoacetyl chloride, subsequent displacement with an amine base, and nitro-reduction (hydrogenation). Subsequent reaction with an electrophilic halogen source, e.g. NCS, affords V-3.
Similarly, aniline V-5 is produced via nitration and deprotection of amide V-4 (see International Patent Publication WO 2001023374), acylation, e.g. α-bromoacetyl chloride and a suitable base (i.e. K2CO3), subsequent displacement with an amine base (for example dimethyl amine or pyrrolidine), and nitro-reduction via hydrogenation (with a palladium source, i.e. Pd/C, and hydrogen gas).
Aniline V-9 is prepared starting with nitro-indoline V-7 according to Scheme 5. Acylation with an acylating agent (for example here acroyl chloride) in the presence of a triakylamine base (i.e. Hunig's base) and solvent (dichloromethane) followed by 1 ,4- conjugate addition of a nucleophile (for example dimethyl amine) to the corresponding α,β-unsaturated amide and then nitro reduction as described for Intermediate II-4 (Scheme 2) affords functionalized aniline V-8. Further reduction of the amide can be effected via treatment of V-8 with a hydride reducing agent (lithium aluminum hydride) in ethereal solvent (for example tetrahydrofuran or diethyl ether) to afford V-9.
Scheme 6 Tetrahydroisoquinoline-denved tails
1 Borane Reductant alkylating agent, 2 acylating agent, solvent, base base, solvent 3 amine base, solvent
4 hydrogenation
Figure imgf000045_0002
Figure imgf000045_0001
Figure imgf000045_0003
VI-1 VI-2 VI-3
Figure imgf000045_0004
1 Borane Reductant
2 acylating agent, solvent, base
3 amine base, solvent
4 hydrogenation
Figure imgf000045_0005
Figure imgf000045_0006
As shown in Scheme 6, tetrahydroquinoline tails may be prepared as follows. Intermediate (VI-1 ), prepared according to Chem. Pharm. Bull. (2001 ) p.822, may be alkylated with an alkylating agent and base, e.g. MeI and K2CO3, to provide VI-2, then amide reduced, e.g. borane, treated with an acylating agent, e.g. α-chloro acetyl choride, an amine base, e.g. dimethyl amine or other exemplary amines, and nitro-reduced (as with Intermediate II-4, Scheme 2) to provide (VI-3).
Intermediate (VI-4), readily available 4-anisidine, may be acylated with 3,3- dimethylacryloyl chloride and base, e.g. K2CO3, heated with a Lewis acid, e.g. AICI3, and solvent, e.g. methylene chloride, followed by nitration with an oxidative reagent, e.g. NaNO2, in acid, e.g. TFA, to provide VI-6. Subsequent amide reduction, e.g. borane, followed by treatment with an acylating agent, e.g. α-bromoacetyl choride, displacement with an amine base, e.g. dimethyl amine, and nitro reduction provides (VI-7).
Scheme 7 Oxygen-linked Anilines iodide source nitro- reduction
Figure imgf000046_0001
Figure imgf000046_0002
1 base amine, base
Figure imgf000046_0003
Figure imgf000046_0004
Further functionalized anilines may be accessed from nitrophenols including VII- (commercially available through CiventiChem) and VII-5 (Meurs, Jan H. H.; Eilenberg, Wolf. Oxidative fluorination in amine-hydrogen fluoride mixtures. Tetrahedron (1991 ), 47(4-5), 707-14.). Alkylation of Vl 1-1 with an alkylating agent (i.e. VII-2), iodide source (i.e. tetrabutylammonium iodide or Kl) and base (i.e. potassium carbonate) affords O- akylated nitrobenenzene VII-3. Subsequent nitro reduction (see Intermediate II-4, Scheme 2) affords aniline VII-4. Similarly, akylation of VII-5 with a bifunctional electrophile (for example VII-6) and base (potassium carbonate) followed by nitro reduction in the presence of a catalyst (Pd/C), solvent (MeOH) and reducing agent (hydrogen gas, typically 30-50 psi) affords aniline VII-7. Direct chloride displacement can be effected via exposure of VII-7 to an amine nucleophile (for example dimethylamine) and base (potassium carbonate) to afford VII-8. Scheme 8 Synthesis of Carboxamide-Containing Amines
Figure imgf000047_0001
VIII-4, R = OMe VIII-5, R = OEt
1 DPPA, TEA, tBuOH
2 base, acylating agent Acid
Figure imgf000047_0004
Figure imgf000047_0002
Figure imgf000047_0003
VIII-6 VIII-7 VIII-8
Several heterocyclic carboxamide-containing amines can be prepared as illustrated in Scheme 8. Bromination of vinyl-nitrile VIII-1 can be effected via direct treatement with bromine. Subsequent thiophene formation occurs when VIII-2 is treated with a 2-mercapto ester (for example VIII-3) in alcoholic solvent (EtOH).
Furan-containing carboxamide VIII-8 can be generated from 3-furanoic acid VIII- 6. A Curtius rearrangement may be effected via activation of VIII-6 (oxalyl chloride or DPPA) and an azide source (sodium azide or DPPA) which when carried out in the presence of base (triethyl amine) in t-BuOH solvent affords the corresponding 3-N-boc furan. Subsequent exposure to 2 equivalents of a strong base (for example tertiary butyl lithium) and then an electrophile (using methylchloroformate above) affords functionalized furan VIII-7. Boc-deprotection of VIII-7 can be carried out via direct treatement with acid (TFA or HCI) to afford free aniline VIII-8. Scheme 9a Pyrrolopyπmidine Intermediates
Figure imgf000048_0001
IX-5 IX-6 IX-7
Figure imgf000048_0002
IX-11 IX-12
The head (IX-8 & IX-9, IX-10) and core (IX-1 ) can be combined as illustrated in Scheme 9a-b. 2,4-dichloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidine is combined with an appropriate amine or aniline (for example IX-8 or IX-9) and a suitable solvent (for example trifluoroethanol) with acid (TFA) to give the corresponding 4- substituted pyrrolo[2,3-d] pyrimidines (IX-2 through IX-4) or with a suitable amine (i.e. IX- 10) in solvent (iPrOH) with base (Hunig's base). The carboxylate esters present on IX-2 through IX-4 may be hydrolyzed under standard aqueous basic conditions (LiOH, water, tetrahydrofuran) and the derived acids (including IX-11 obtained directly) may be activated by conversion to the corresponding acid chlorides by treatment with a chloride source/activating agent (oxalyl chloride) and catalyst (DMF). The in situ generated acid chlorides react intramolecularly with the 5-nitrogen group to give the corresponding tetracyclic species IX-5 through IX-7 and IX-12. Tetracyclic species IX-5 through IX-7 activate the C(7) chloride toward further displacement (see Scheme 10).
Scheme 10 Elaboration of Activated Intermediates (N* = Functionalized Anline-See Schemes 1-7) (amin
base, solvent
Figure imgf000050_0001
1 N*, solvent, heat
2 amine, solvent base, solvent
Figure imgf000050_0004
Figure imgf000050_0002
X-2
Figure imgf000050_0003
Figure imgf000050_0005
Treatment of chlorides X-1 through X-3 and X-10 with a functionalized aniline (see Schemes 1-7) in solvent (tetrahydrofuran, trifluoroethanol, dioxane) with acid catalysis (hydrochloric acid) gives C(7)-CI displacement. Subsequent exposure to a nucleophile (typically a primary or secondary amine, for example ammonium, methyl amine, propyl amine) gives reaction at the carbonyl and affords X-4, X-5, X-6, and X-1 1. Removal of the tosyl protection group proceeds upon treatment with base (KOH, NaOH, NaOMe) in a polar protic solvent (H2O, MeOH) with optional cosolvent (dioxane, tetrahdyrofuran) and heating (optionally in a microwave reactor) to afford X-7, X-8, X-9, and X-12.
Scheme 11
1 protection
2 reduction
3 acylation
4 amine/nucleophile
Figure imgf000052_0002
Figure imgf000052_0001
1 nitration 1 acylation
2 deprotection 2 amine/nucleophile
3 nitro reduction
Figure imgf000052_0005
Figure imgf000052_0004
Figure imgf000052_0003
1 actylation
2 amine/nucleophile nitro reduction
Figure imgf000052_0006
Figure imgf000052_0007
Figure imgf000052_0008
Exposure of commercially available XI-1 to aniline protection conditions (i.e. BOC2O, DIPEA), nitro reduction conditions (i.e. Pd/C, hydrogen gas), acylation conditions (i.e. α- bromo acetyl chloride, DIPEA), and then introduction of a nucleophile (i.e. dimethyl amine) affords XI-2. Deprotection of XI-2 with acid (i.e. HCI, or TFA) affords XI-3.
Exposure of XI-4 (see Arp, Forrest O.; Fu, Gregory C. Kinetic Resolutions of lndolines by a Nonenzymatic Acylation Catalyst. Journal of the American Chemical Society (2006), 128(44), 14264-14265.) to nitration conditions (i.e sodium nitrate and TFA) followed by deprotection of the acyl group with acid (i.e. HCI) affords XI-5. Advancement of XI-5 or XI-7 (see: Achvlediani, R.; Natsvlishvili, M.; Baberkina, E.; Khachidze, M.; Abesadze, I.; Suvorov, N. Synthesis of 1H-pyrrolo[3,2-χl- and 1 H-pyrrolo[2,3-χjquinoline. Izvestiya Akademii Nauk Gruzii, Seriya Khimicheskaya (1996), 22(1-4), 43-47.) through a reaction sequence similar to that described above (i.e. acylation with α-bromo acetyl chloride, displacement with an amine or other nucleophile, and nitro reduction) affords anilines XI-6 and XI-9. EXAMPLES
The following specific examples are included as illustrations and are not to be construed as limiting the scope of the present invention.
As used herein, the symbols and conventions used in these processes, schemes and examples are consistent with those used in the contemporary scientific literature, for example, the Journal of the American Chemical Society or the Journal of Biological Chemistry. Standard single-letter or three-letter abbreviations are generally used to designate amino acid residues, which are assumed to be in the L-configuration unless otherwise noted. Unless otherwise noted, all starting materials were obtained from commercial suppliers and used without further purification. Specifically, the following abbreviations may be used in the examples, schemes, biological writeups, and throughout the specification:
APC (Allophycocyanin) M (molar);
ATP (adenosine triphosphate) MeOH (methanol);
Ac (acetyl) mg (milligrams) atm (atmosphere) MgCL2, magnesium chloride
BSA (bovine serum albumin) MHz (megahertz)
CHAPS (3-[3- min (minutes)
Cholamidopropyl)Dimethylammonio] -1 - mL (milliliters)
Propanesulfonate) mM (millimolar)
DIPEA(diisopropylethylamine) mmol (millimoles)
DMSO (dimethyl sulfoxide) mol (moles);
DTT (Dithiothreitol) mp (melting point);
EDTA (ethylenediaminetetraacetic acid) NaCI, Sodium chloride
Eu (Europium) NCBI, National Center for Biotechnology
G (grams) Information
GST (Glutathione S-transferase) nM, nanomolar h (hours); OTF (trifluoromethane sulfonate)
HEPES (N-(2-Hydroxyethyl)piperazine-N'- psi (pounds per square inch);
2-ethanesulfonic acid) RP (reverse phase); hlGF1 R or IGF-1 R (human Insulin-like rt (room temperature); growth factor 1 receptor kinase) SEM (2-trimethylsilyl(ethoxymethyl) hlR (human insulin receptor kinase) TBAI (tetrabutylammonium iodide);
HPLC (high performance/pressure liquid TFA (trifluoroacetic acid); chromatography) TFE (trifluoroethanol)
Hz (Hertz); THF (tetrahydrofuran);
/-PrOH (isopropanol); TLC (thin layer chromatography); i. v. (intravenous); Tr (retention time);
L (liters); μL (microliters); μM, micromolar Unless otherwise noted, reagents and solvents were obtained from commercial suppliers and were used without further purification. Unless otherwise indicated, all reactions were conducted at room temperature and all temperatures are expressed in 0C (degrees Centigrade). Throughout the specification, multi-step syntheses are described with respect to various intermediate and exemplary compounds. In general, all but the first step of each multi-step synthesis refer to a product compound of a preceding step. Unless otherwise noted, such reference is understood as reference to the compound in general, though not necessarily to the actual sample of product that was produced in carrying out the preceding step.
Thin-layer chromatography (TLC) was performed on silica gel 60 F254 precoated plates. Detection was effected by exposure to UV light (254 nm). Flash and flush column chromatography was performed using Silica Gel 60. Reverse phase preparative and analytical HPLC were performed using C18 columns and acetonitrile:water gradients with 0.05% TFA as a modifier.
Compound purity and characterization were determined by 1H-NMR, liquid chromatography-mass spectrometry (LCMS), high resolution mass spectrometry (HRMS), combustion (elemental) analysis, HPLC, and melting point. Compounds of general formula I were typically found to have purities of > 90%. 1H NMR spectra were recorded on Varian INOVA-300 and Varian INOVA-400 instruments. Chemical shifts are expressed in parts per million (ppm, δ units). Coupling constants are in units of hertz (Hz). Splitting patterns describe apparent multiplicities and are designated as s (singlet), d (doublet), dd (doublet of doublet), t (triplet), q (quartet), m (multiplet), or br (broad). Low resolution mass spectra were obtained on Micromass ZQ, Micromass ZMD,
Micromass QuattroMicro, and Micromass GCT instruments from Micromass Ltd., Altricham, UK, using either Atmospheric Pressure Chemical Ionization (APCI) or ESI Ionization (ESI).
High resolution mass spectral data (HRMS) were recorded with Micromass LCT and Micromass GCT instruments.
Combustion analyses were performed by Atlantic Microlab, Inc. (Norcross, Georgia).
Melting points were recorded in open capillary tubes and are uncorrected. Section A: Functionalized Anilines
Intermediate A1 : 5-methyl-2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1 ■ piperazinyljaniline
Figure imgf000055_0001
Step A/Intermediate A2: 5-fluoro-4-methyl-2-nitrophenol
Figure imgf000055_0002
3-fluoro-4-methylphenol (3.66 g, 29.0 mmol) was dissolved in DCE (32 ml.) and tetrabutylammonium bromide (0.935 g, 2.90 mmol) was added. 70% aqueous HNO3 (3.7 ml_, 58 mmol) was diluted with H2O (33 ml.) to make a 7% HNO3 solution. This solution was added to the reaction mixture, which was then stirred at rt for 4h at which time the reaction was judged complete by TLC. The reaction was poured into H2O and extracted with DCM (3x). The combined organic layers were dried over MgSO4 , filtered and concentrated in vacuo. The resulting residue was adsorbed onto silica gel and flash chromatographed to give 5-fluoro-4-methyl-2-nitrophenol (2.83 g, 57%). 1H NMR (400 MHz, DMSO-Cf6) δ ppm 1 1.10 (S, 1 H), 7.89 (d, J=8.1 Hz, 1 H), 6.85 (d, J=1 1.0 Hz, 1 H), 2.13 (s, 3 H).
Step B/lntermediate A3: 1-fluoro-2-methyl-5-(methyloxy)-4-nitrobenzene
Figure imgf000055_0003
5-Fluoro-4-methyl-2-nitrophenol (2.83 g, 16.5 mmol) was dissolved in N, N- dimethylformamide (25 ml_). K2CO3 (3.4 g, 25 mmol) and iodomethane (1.2 ml_, 20 mmol) were added and the mixture was stirred at rt overnight. The mixture was then poured into H2O and stirred until solids crashed out. The solids were filtered and air dried to give the 1-fluoro-2-methyl-5-(methyloxy)-4-nitrobenzene (2.76 g, 90 %). 1H NMR (400 MHz, DMSOd6) δ ppm 7.92 (d, J=8.1 Hz, 1 H), 7.25 (d, J=1 1.7 Hz, 1 H), 3.89 (s, 3 H), 2.19 (d, J=1.5 Hz, 3 H).
Step C/lntermediate A4: 1 ,1-dimethylethyl 4-[2-(methylsulfonyl)ethyl]-1- piperazinecarboxylate
Figure imgf000056_0001
To a solution of 1 ,1-dimethylethyl 1-piperazinecarboxylate (72.8 g, 390 mmol) in acetonitrile (1.4 L) was added methyl vinyl sulfone (50 g, 470 mmol) and Na2CO3 (124 g, 1170 mmol) and the mixture was refluxed overnight. Then the mixture was cooled to rt, and poured into H2O and extracted with EtOAc. The organic layer was dried over Na2SO4 and the solvent was removed under reduced pressure to give 1 ,1-dimethylethyl 4-[2-(methylsulfonyl)ethyl]-1-piperazinecarboxylate (106 g, 93% yield). 1H NMR (400 MHz, CDCI3) δ ppm 1.41-1.57 (m, 9H), 2.39-2.51 (m, 4H), 2.88 (t, J=8.4Hz, 2H), 3.02 (s, 3H), 3.14 (t, J=8.4Hz, 2H), 3.35-3.50 (m, 4H).
Step D/lntermediate A5: 1-[2-(methylsulfonyl)ethyl]piperazine hydrochloride
Figure imgf000056_0002
To the solution of 1 ,1-dimethylethyl 4-[2-(methylsulfonyl)ethyl]-1-piperazinecarboxylate (106 g, 360 mmol) in MeOH (500 ml.) was added HCI/MeOH (5M, 1000 ml_, 5000 mmol), and the mixture was heated at 50 0C for 1 h before evaporating most of the solvent. The residue was filtered and washed with MeOH to give 1-[2- (methylsulfonyl)ethyl]piperazine hydrochloride (90.3 g, 95% yield). 1H NMR (400 MHz, D2O) δ ppm 3.30-3.72 (m, 12H), 3.09 (s, 3H).
Step E/lntermediate A6: 1-[2-methyl-5-(methyloxy)-4-nitrophenyl]-4-[2- (methylsulfonyl)ethyl]piperazine
Figure imgf000057_0001
1-fluoro-2-methyl-5-(methyloxy)-4-nitrobenzene (5.0 g, 27 mmol), K2CO3 (1 1.2 g, 81 mmol), and 1-[2-(methylsulfonyl) ethyl]piperazine hydrochloride (8.0 g, 35 mmol) were weighed into a 150 ml. sealed tube, taken up in 85 ml. of DMSO and heated to 100 0C for 24 h. The mixture was poured into 500 ml. of H2O, filtered, washed with H2O, air dried for 15 min, washed with hexanes (200 ml_), washed with 20%ether/ hexanes (20OmL), and air dried for 2 h to give 1-[2-methyl-5-(methyloxy)-4-nitrophenyl]-4-[2- (methylsulfonyl)ethyl]piperazine (6.6 g, 18.5 mmol, 68%). An additional (540 mg, 5.5%) of product was obtained by extraction of the H2O layer, followed by flash chromatography. 1H NMR (400 MHz, DMSO-d6) δ ppm 7.75 (s, 1 H), 6.70 (s, 1 H), 3.90 (s, 3 H), 3.31 - 3.35 (m, 2 H), 3.04 (s, 3 H), 2.99 - 3.03 (m, 4 H), 2.77 (t, J=6.60 Hz, 2 H), 2.56 - 2.63 (m, 4 H), 2.19 (s, 3 H).
Step F/lntermediate A1 : 5-methyl-2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1- piperazinyl}aniline
Figure imgf000057_0002
1-[2-methyl-5-(methyloxy)-4-nitrophenyl]-4-[2-(methylsulfonyl)ethyl]piperazine from a separate batch (0.257 g, 0.72 mmol) was placed in a 40 ml. high vial and dissolved in 10 ml. of 1 to 1 EtOAc/MeOH. 5 wt% Platinum(sulfided)/carbon (0.165 g, 0.043 mmol) was added and the vial was quickly sealed by a screw cap septum. The vial was evacuated and filled with N2 six times to remove any oxygen. The vial was then pressurized with H2 (balloon). The solution stirred overnight. The next morning the vessel was evacuated and filled with N2 six times to remove any H2. The solution was filtered through celite and evaporated to afford 5-methyl-2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1- piperazinyl}aniline (0.194 g, 0.59 mmol, 82%). 1H NMR (400 MHz, CDCI3) δ ppm 6.54 (s, 2 H), 3.80 (s, 3 H), 3.30 - 3.37 (m, 2 H), 3.07 (s, 3 H), 2.91 - 2.98 (m, 6 H), 2.82 - 2.90 (m, 4 H), 2.15 (s, 3 H).
Intermediate A7: 1-[(dimethylamino)acetyl]-5-(methyloxy)-2,3-dihydro-1H-indol-6-
Figure imgf000058_0001
Step A/Intermediate A8: Λ/,Λ/-dimethyl-2-[5-(methyloxy)-2,3-dihydro-1 /-/-indol-1-yl]-2- oxoethanamine
Figure imgf000058_0002
A suspension of 5-(methyloxy)-2,3-dihydro-1 /-/-indole hydrogen chloride (94.5g, 509 mmol) in CH2CI2 (25OmL) was added dropwise to a mixture of α-bromoacetylchloride
(12Og, 764 mmol) and K2CO3 (78g, 560mmol) in CH2CI2 (750 mL) at 0°C. After the addition, the suspension was stirred at 0 C for 1.5 hours. The reaction was washed with water and filtered. The residue (presumed to be K2CO3) was rinsed with DCM (2x100ml). The organic layers were separated, combined, dried over Na2SO4, filtered, and concentrated to provide a brown solid (135g, 98%).
The brown solid (135g, 499 mmol) was dissolved in DCM (500ml), cooled to 00C, then treated with K2CO3 (139g, 999 mmol), and 650ml of 2 M dimethyl amine in THF. The reaction was stirred at 00C for 1 hour and filtered. The resulting solid was washed with DCM. The combined filtrates were washed with water, dried over Na2SO4, filtered, and concentrated to provide Λ/,Λ/-dimethyl-2-[5-(methyloxy)-2,3-dihydro-1/-/-indol-1-yl]-2- oxoethanamine as a grey solid (111 g, 95%).
Step B/lntermediate A9: Λ/,Λ/-dimethyl-2-[5-(methyloxy)-6-nitro-2,3-dihydro-1/-/-indol-1- yl]-2-oxoethanamine
Figure imgf000059_0001
To a solution of NaNO3 (43.9g, 516 mmol) in TFA (250 ml.) was added Λ/,Λ/-dimethyl-2- [5-(methyloxy)-2,3-dihydro-1 /-/-indol-1-yl]-2-oxoethanamine (1 10.0 g, 469 mmol) in TFA (50 ml.) at O0C and the mixture was stirred at 00C for 1.5h. The reaction mixture was poured into ice-water (100OmL), the TFA and some water was removed by reduced pressure (the volume was reduced to 400 ml_), the reaction mixture was adjusted to pH = 11 by addition of 2N NaOH (the mixture became cloudy), and the aqueous layer was extracted with CHCI3 (2x300ml_), and washed with water. The organic layer was separated, dried over with Na2SO4, and the solvents were removed under reduced pressure to yield Λ/,Λ/-dimethyl-2-[5-(methyloxy)-6-nitro-2,3-dihydro-1/-/-indol-1-yl]-2- oxoethanamine (13Og, 99%). 1H NMR (400 MHz, DMSO-d6) δ ppm 2.24 (s, 6 H), 3.15 - 3.24 (m, 4 H), 3.86 (s, 3 H), 4.18 (t, J=8.5 Hz, 2 H), 7.31 (s, 1 H), 8.45 (s, 1 H). This protocol could be repeated to generate larger quantities of Λ/,Λ/-dimethyl-2-[5- (methyloxy)-6-nitro-2,3-dihydro-1H-indol-1-yl]-2-oxoethanamine which could be carried forward in the synthetic sequence as blended batches.
Step C/lntermediate A7: 1-[(dimethylamino)acetyl]-5-(methyloxy)-2,3-dihydro-1 /-/-indol-
Figure imgf000059_0002
A suspension of N,N-dimethyl-2-[5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indol-1-yl]-2- oxoethanamine (26.6 g, 95 mmol), iron(lll)chloride (3.09 g, 19.05 mmol), activated carbon (25 g, 95 mmol), and hydrazine hydrate (37.4 ml_, 762 mmol) in methanol (500 ml.) was warmed to 650C and maintained overnight. The reaction was filtered through celite while still warm and all methanol was removed under reduced pressure. The solids were partitioned between ethyl acetate (ca 1 L) and saturated sodium chloride (ca 1 L) and the aqueous layer was washed twice with ethyl acetate (ca 500 mL). The combined organic layers were dried over sodium sulfate, taken to a residue under reduced pressure, and the derived solids triturated with hexanes/diethyl ether and filtered to afford 1-[(dimethylamino)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-amine (16.46 g, 69.3 % yield) as an off-white solid: NMR: 1H NMR (400 MHz, DMSOd6) δ ppm 2.23 (s, 6 H), 2.95 (t, J=8.25 Hz, 2 H), 3.1 1 (s, 2 H), 3.69 (s, 3 H), 4.07 (s, 2 H), 4.61 (s, 2 H), 6.68 (s, 1 H), 7.53 (s, 1 H).
Intermediate A10: 5-ethyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1 -piperazinyl]-1 - piperidinyljaniline
Figure imgf000060_0001
Step A/Intermediate A1 1 : 1 ,1-dimethylethyl 4-(methylsulfonyl)-1-piperazinecarboxylate
Figure imgf000060_0002
To 1 ,1-dimethylethyl 1-piperazinecarboxylate (568 g, 3.05 mol) in DCM (4 L) was added Et3N (617 g, 6.10 mol). After stirring for 10 min at 0 0C, MsCI (384 g, 3.35 mol) was added via addition funnel. The mixture was stirred at rt overnight. The mixture was poured into H2O (1 L) and extracted with DCM (1 L). The organic layer was separated, washed with H2O (1 L), dried (Na2SO4), and concentrated to provide 1 ,1-dimethylethyl 4- (methylsulfonyl)-i-piperazinecarboxylate (720 g, 2.72 mol, 90 %) which was used without further purification. 1H NMR (400 MHz, CDCI3) δ 1.44 (s, 9 H), 2.76 (s, 3 H), 3.1 1 - 3.17 (m, 4 H), 3.50 - 3.53 (m, 4 H). Step B/lntermediate A12: 1-(methylsulfonyl)piperazine hydrochloride
HN N-SO2CH3 -HCI
To 1 ,1-dimethylethyl 4-(methylsulfonyl)-1-piperazinecarboxylate (360 g, 1.36 mol) in MeOH (1 L) was added HCI (6 M in MeOH, 2 L) dropwise. The mixture was stirred at rt for 1 h. About 1 L of MeOH was rotovapped off. The resultant precipitate was filtered, washed with MeOH, and dried on high vacuum to provide 1-(methylsulfonyl)piperazine hydrochloride (A combination of 2 batches, 570 g) which was used without further purification. 1H NMR (400 MHz, D2O) δ 2.95 (s, 3 H), 3.27 - 3.29 (m, 4 H), 3.42 - 3.46 (m, 4 H).
Step C/lntermediate A13: 1-(methylsulfonyl)-4-(4-piperidinyl)piperazine dihydrochloride
HN )— N N-SO7CH, -2HCI
To 1-(methylsulfonyl)piperazine hydrochloride (150 g, 632 mmol) in DCE (3.5 L) was added Et3N (192 g, 1.90 mol). The mixture was stirred at rt for 1 h and then acetic acid (94.8 g, 1.58 mol) and 1 ,1-dimethylethyl 4-oxo-1-piperidinecarboxylate (251 g, 1.26 mol) was added. After stirring another hour, the reaction was cooled with an ice water bath and NaBH(OAc)3 (294 g, 1.39mol) was added in four portions. The mixture was stirred overnight at rt. The reaction mixture was neutralized with saturated Na2CO3 to pH 8-9. The organic phase was washed with brine and H2O, dried (Na2SO4), and concentrated to provide the crude Boc-protected amine (A combination of 3 batches, 720 g). This amount was split into 2 batches and used without further purification. To 1 ,1- dimethylethyl 4-[4-(methylsulfonyl)-1-piperazinyl]-1-piperidinecarboxylate (360 g, 1.04 mol) in MeOH (1 L) was added HCI (6 M in MeOH, 2 L). The mixture was stirred at rt for 30 min. About 1 L of MeOH was rotovapped off. The resultant precipitate was filtered, washed with MeOH, and dried on high vacuum to provide 1-(methylsulfonyl)-4-(4- piperidinyl)piperazine dihydrochloride (A combination of 2 batches, 600 g, 1.87 mol, 89 % over 2 steps). 1H NMR (400 MHz, D2O) δ 1.87 - 1.91 (m, 2 H), 2.33 - 2.36 (m, 2 H), 2.97 (s, 3 H), 2.99 - 3.05 (m, 2 H), 3.45 - 3.59 (m, 1 1 H).
Step D/lntermediate A14: 4-ethyl-3-fluorophenol
Figure imgf000062_0001
2-Fluoro-4-hydroxyacetophenone (15.14 g, 100 mmol) and zinc dust (19.61 g, 300 mmol) was suspended in 50 ml. of H2O with stirring. Concentrated HCI (37%, 50 ml_, 610 mmol) were added and the mixture was heated to reflux for six hours. The reaction was cooled to rt, and diethyl ether was added. The mixture was saturated with NaCI, and filtered through celite washing with diethyl ether. The filtrate was poured into brine, and the layers were separated. The aqueous layer was extracted with diethyl ether. The combined organic layers were dried over MgSO4, filtered, and concentrated in vacuo. The residue was purified by flash chromatography to afford 7.24 g (52%) of 4- ethyl-3-fluorophenol. 1H NMR (400 MHz, DMSOd6) δ 9.53 (s, 1 H), 7.00 (t, J = 8.8 Hz, 1 H), 6.52 - 6.40 (m, 2H), 2.44 (q, J = 7.5 Hz, 2H), 1.06 (t, J = 7.5 Hz, 3H).
Step E/lntermediate A15: 4-ethyl-5-fluoro-2-nitrophenol
Figure imgf000062_0002
4-Ethyl-3-fluorophenol (7.24 g, 51.6 mmol) was dissolved in 100 ml. of DCM with stirring. Tetrabutylammonium bromide (1.66 g, 5.15 mmol) was added. A seven percent aqueous solution of HNO3 (66 ml.) was prepared and added to the reaction. The biphasic mixture was stirred vigorously overnight. The reaction was poured into DCM and H2O, and the layers were separated. The aqueous layer was washed with DCM (2X). The combined organic layers were dried over MgSO4, filtered, and concentrated in vacuo. The residue was purified by flash chromatography to afford 5.72 g (60%) of 4- ethyl-5-fluoro-2-nitrophenol. 1H NMR (400 MHz, DMSO-d6) δ 11.17 (s, 1 H), 7.89 (d, J = 8.0 Hz, 1 H), 6.89 (d, J = 11.2 Hz, 1 H), 2.56 (q, J = 7.5 Hz, 2H), 1.13 (t, J = 7.5 Hz, 3H).
Step F/lntermediate A16: 1-ethyl-2-fluoro-4-(methyloxy)-5-nitrobenzene
Figure imgf000062_0003
4-Ethyl-5-fluoro-2-nitrophenol (5.72 g, 30.9 mmol) was dissolved in 60 ml. of DMSO with stirring. K2CO3 (6.41 g, 46.4 mmol) was added. Methyl iodide (2.5 ml_, 40 mmol) was added via syringe. The reaction was stirred overnight and poured into H2O and diethyl ether. The layers were separated, and the organic layer was washed with brine. The combined aqueous layers were extracted with EtOAc (2X). The combined organic layers were dried over MgSO4, filtered, and concentrated in vacuo. The residue was purified by flash chromatography to afford 5.13 g (83%) of 1-ethyl-2-fluoro-4- (methyloxy)-5-nitrobenzene. 1H NMR (400 MHz, DMSO-d6) δ 7.88 (d, J = 7.9 Hz, 1 H), 7.23 (d, J = 12.1 Hz, 1 H), 3.86 (s, 3H), 2.56 (q, J = 7.6 Hz, 2H), 1.1 1 (t, J = 7.5 Hz, 3H).
Step G/lntermediate A17: 1-{1-[2-ethyl-5-(methyloxy)-4-nitrophenyl]-4-piperidinyl}-4-
(methylsulfonyl)piperazine
Figure imgf000063_0001
A mixture of 1-ethyl-2-fluoro-4-(methyloxy)-5-nitrobenzene (0.93 g, 4.67 mmol), 1- (methylsulfonyl)-4-(4-piperidinyl)piperazine (1.155 g, 4.67 mmol) and K2CO3 (0.774 g, 5.60 mmol) in DMSO (20 ml.) was heated at 90 0C for 48 h. The reaction had not progressed sufficiently so the reaction was then heated at 120 0C for an additional 4 h. The reaction was cooled to rt, poured into H2O and extracted exhaustively with DCM. The combined organics were washed with H2O then dried over MgSO4. The resultant solution was concentrated onto silica and purified by flash chromatography to afford 1- {1-[2-ethyl-5-(methyloxy)-4-nitrophenyl]-4-piperidinyl}-4-(methylsulfonyl)piperazine (1.12 g, 56%). 1H NMR (400 MHz, DMSO-d6) δ ppm 7.73 - 7.80 (m, 1 H), 6.75 (s, 1 H), 3.91 (s, 3 H), 3.23 - 3.30 (m, 1 H), 3.05 - 3.19 (m, 3 H), 2.87 (s, 2 H), 2.70 - 2.84 (m, 2 H), 2.53 - 2.67 (m, 5 H), 1.77 - 1.94 (m, 2 H), 1.48 - 1.67 (m, 2 H), 1.19 (t, J=7.42 Hz, 3 H). (some protons obstructed by solvent).
Step H/lntermediate A10: 5-ethyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1- piperidinyl}aniline
Figure imgf000064_0001
A mixture of 1-{1-[2-ethyl-5-(methyloxy)-4-nitrophenyl]-4-piperidinyl}-4- (methylsulfonyl)piperazine (1.12 g, 2.63 mmol) and sulfided platinum on carbon (0.410 g, 0.105 mmol) in EtOAc (40 ml.) was sealed in a round bottom flask with a rubber septum. The reaction mixture was purged with N2 gas and then a balloon of H2 gas was connected and the vessel was flushed with the H2 gas. The reaction was stirred at rt for 48 h. TLC analysis showed the complete consumption of the starting nitro compound so the reaction vessel was purged with N2 and the reaction mixture was filtered through celite to remove the catalyst. The filtrate was concentrated onto silica gel and purified by flash chromatography to afford 5-ethyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1- piperazinyl]-1-piperidinyl}aniline (0.479 g, 46%). 1H NMR (400 MHz, DMSO-c/6) δ ppm 6.60 (s, 1 H), 6.46 (s, 1 H), 4.35 (br. s., 2 H), 3.71 (s, 3 H), 3.03 - 3.16 (m, 4 H), 2.81 - 2.93 (m, 5 H), 2.56 - 2.68 (m, 6 H), 2.29 - 2.42 (m, 1 H), 1.72 - 1.89 (m, 2 H), 1.44 - 1.62 (m, 2 H), 1.09 (t, J=7.51 Hz, 3 H). (some protons obstructed by solvent).
Intermediate A18: 4-[4-(1 -methylethyl)-1 -piperazinyl]-2-(methyloxy)aniline
Figure imgf000064_0002
Step A/Intermediate A19: 1-(1-methylethyl)-4-[3-(methyloxy)-4-nitrophenyl]piperazine
Figure imgf000065_0001
To 4-chloro-2-(methyloxy)-1 -nitrobenzene (3.0 g, 16.0 mmol) in dioxane (75 mL) was added 1-(1-methylethyl)piperazine (4.1 g, 32.0 mmol), XANTPHOS (1.4 g, 2.4 mmol), and Cs2CO3 (10.4 g, 32.0 mmol). The mixture was bubbled with N2 for 15 min prior to the addition of Pd2(dba)3 (1.5 g, 1.6 mmol). The reaction was stirred at 100 0C for 5 h. Following cooling to room temperature, the reaction mixture was diluted with ethyl acetate (150 mL) and water (100 mL). The organic layer was dried over sodium sulfate, filtered, taken to a residue under reduced pressure, and purified by silica gel chromatography to afford 1-(1-methylethyl)-4-[3-(methyloxy)-4-nitrophenyl]piperazine (4.0 g, 90% yield). ESIMS (M+H)+ = 280.
Step B/lntermediate A18: 4-[4-(1-methylethyl)-1-piperazinyl]-2-(methyloxy)aniline
Figure imgf000065_0002
In a pressure vessel was placed a solution of 1-(1-methylethyl)-4-[3-(methyloxy)-4- nitrophenyl]piperazine (4.0 g, 14.3 mmol) in EtOH (100 mL). The solution was purged with N2 for 15 min before the addition of 10% Pd/C (0.5 g). The reaction was stirred at RT for 5 h under 60 psi H2. After releasing H2 pressure, filtration removed the solid resin, and the filtrate was concentrated and purified with silica gel chromatography (0- 5% 2 M NH3 in MeOH/DCM) to furnish 4-[4-(1-methylethyl)-1-piperazinyl]-2- (methyloxy)aniline (3.6 g, 99% yield). 1 H NMR (400 MHz, CDCI3) δ ppm 1.09 (d, J = 6.6 Hz, 6 H), 2.66 - 2.75 (m, 5 H), 3.05 - 3.10 (m, 4 H), 3.47 (s, 3 H), 3.83 (s, 2 H), 6.42 (dd, J = 8.2, 2.4 Hz, 1 H), 6.52 (d, J = 2.2 Hz, 1 H), 6.64 (d, J = 8.4 Hz, 1 H).
Intermediate A20: 2-(methyloxy)-4-(1 -propyl-4-piperidinyl)aniline
Figure imgf000066_0001
Step A/Intermediate A21 : 4-[3-(methyloxy)-4-nitrophenyl]pyridine
Figure imgf000066_0002
Nitrogen was bubbled through dioxane (800 mL) for 1 h followed by the addition of 4- chloro-2-(methyloxy)-1-nitrobenzene (61 g, 0.33 mol), 3-pyridinylboronic acid (Boron Molecular, 40 g, 0.33 mmol), dichloro(triphenylphosphine)palladium (10 g, 14 mmol), and degassed aqueous 3 M Na2CO3 (325 mL, 975 mmol). The reaction mixture was stirred with a mechanical stirrer and heated at 90 0C for 3 h. The reaction was cooled and most of the dioxane was removed in vacuo. It was diluted with water and then extracted with ethyl acetate. Combined organic phases were dried (Mg2SO4), filtered and concentrated. The resultant solid was washed with diethyl ether to afford 4-(3-methoxy- 4-nitrophenyl)pyridine (50 g, 66% yield). 1H NMR (400 MHz, DMSO-c/6) δ ppm 4.05 (s, 3 H), 7.53 (dd, J=8.4, 1.8 Hz, 1 H), 7.69 (d, J=1.8 Hz, 1 H), 7.84 (d, J=6.2 Hz, 2 H), 8.02 (d, J=8.4 Hz, 1 H), 8.72 (d, J=6.2 Hz, 2 H). ESIMS (M+H)+ = 231. Step B/lntermediate A22: 4-[3-(methyloxy)-4-nitrophenyl]-1-propylpyridinium iodide
Figure imgf000067_0001
n-Propyliodide (200 ml_, 2.05 mol) was added to a solution of 4-(3-methoxy-4- nitrophenyl)pyιϊdine (25.0 g, 109 mmol) in pinacolone (500 ml_). The reaction was fitted with a reflux condenser, stirred, and heated at 100 0C for 12 h. A light brown suspension was observed. An aliquot (-2.0 ml.) was taken out from the reaction mixture, concentrated, and analyzed by 1 H NMR which revealed the formation of the alkylated product and the absence of starting material. The reaction was cooled; the solids were filtered off and rinsed once with cold (0 0C) acetone to afford 4-(3-methoxy-4- nitrophenyl)-1-propylpyridinium iodide as a light brown residue (29.0 g, 67% yield). 1H NMR (400 MHz, DMSOd6) δ ppm 0.92 (t, J=I.3 Hz, 3 H), 1.99 (dt, J=I.2,, 7.3 Hz, 2 H), 4.08 (s, 3 H), 4.59 (t, J=I Λ Hz, 2 H), 7.78 (dd, J=1.5, 8.4 Hz, 1 H), 7.92 (d, J=1.5 Hz, 1 H), 8.13 (d, J=8.4 Hz, 1 H), 8.65 (d, J=7.0 Hz, 2 H), 9.22 (d, J=6.6 Hz, 2 H).
Step C/lntermediate A23: 4-(3-methoxy-4-nitrophenyl)-1-propyl-1 ,2,3,6- tetrahydropyridine
Figure imgf000067_0002
4-(3-methoxy-4-nitrophenyl)-1-propylpyridinium iodide (10 g, 25 mmol) was dissolved in methanol (200 ml_). As the mixture was cooled to -10 0C the starting material started to precipitated out of the solution. Sodium borohydride (4.61 g, 1 1.3 mmol) was added in -500 mg portions. Effervescence was observed during the addition as dissolution of the solids in the reaction was observed. The reaction was stirred for 2 h at -10 0C when analysis by TLC revealed completion of the reaction. Saturated aqueous ammonium chloride was added and the reaction warmed to room temperature. The mixture was extracted with ethyl acetate, the organic layers were dried (Mg2SO4), filtered, and concentrated in vacuo to provide 4-(3-methoxy-4-nitrophenyl)-1-propyl-1 , 2,3,6- tetrahydropyridine (6.7 g, 97% yield). This protocol could be reproduced to generate additional product which could be carried forward as blended batches. 1H NMR (400 MHz, DMSOd6) δ ppm 0.88 (t, J=7.4 Hz, 3 H), 1.50 (dt, J=7.3, 7.3 Hz, 2 H), 2.37 (t, J=7.2 Hz, 2 H), 2.53 (m, 2 H), 2.63 ( m, 2 H), 3.11 (br s, 2 H), 3.96 (s, 3 H), 6.43 (t, J=3.5 Hz, 1 H), 7.16 (dd, J=QA, 1.5 Hz, 1 H), 7.29 (d, J=1.3 Hz, 1 H), 7.86 (d, J=8.6 Hz, 1 H). ESIMS (M+H)+ = 277.
Step D/lntermediate A20: 2-(methyloxy)-4-(1-propyl-4-piperidinyl)aniline
Figure imgf000068_0001
Palladium on carbon (10% by weight, 2.5 g) was added to a nitrogen-flushed Fischer- Porter vessel. A solution of the 4-(3-methoxy-4-nitrophenyl)-1-propyl-1 ,2,3,6- tetrahydropyridine (9.20 g, 33.3 mmol) in ethyl acetate (150 ml.) followed by methanol (50 ml.) were added to the vessel. The reaction was purged and then kept under a hydrogen pressure of 60 psi for 2 d. The pressure was released and the reaction was purged with nitrogen. The reaction was filtered through celite and concentrated to afford 2-methoxy-4-(1-propylpiperidin-4-yl)aniline as a white solid (7.0 g, 85% yield). 1H NMR (400 MHz, DMSOd6) δ ppm 0.86 (t, J=7.5 Hz, 3 H), 1.44 (dt, J=IA, IA Hz, 2 H), 1.52 - 1.63 (m, 2 H), 1.63 - 1.70 (m, 2 H), 1.90 (td, J= 11.6, 2.4 Hz, 2 H), 2.20 - 2.25 (m, 2 H), 2.30 (tt, J= 11.9, 4.0 Hz, 1 H), 2.89 - 2.94 (m, 2 H), 3.74 (s, 3 H), 4.45 (s, 2 H), 6.53 (s, 2 H), 6.65 (s, 1 H). ESIMS (M+H)+ = 249.
Intermediate A14: 1 -{[ethyl(methyl)amino]acetyl}-5-(methyloxy)-2,3-dihydro-1 H- indol-6-amine
Figure imgf000069_0001
Step A/Intermediate A25: 5-(methyloxy)-2,3-dihydro-1 /-/-indole
Figure imgf000069_0002
At room temperature, to a solution of 5-(methyloxy)-1 /-/-indole (12 g, 81.5 mmol, 1.0 eq) in acetic acid (150 ml.) was added sodium cyanoborohydride (10.25 g, 163 mmol, 2.0eq) in small portions, and the mixture was stirred overnight. The solvent was evaporated under reduced pressure and the residue was dissolved in ethyl acetate. The organic layer was washed with saturated aqueous sodium hydrogencarbonate solution and brine, and dried over Na2SO4 . The solvent was removed under reduced pressure to give 5-(methyloxy)-2,3-dihydro-1 H-indole (8.91 g, 45% yield) 1H NMR (CDCI3, 300 MHz) δ 6.76 (s, 1 H), 6.60 (s, 2H), 3.54 (t, 2H), 3.01 (t, 2H).
Step B/lntermediate A26: 1-acetyl-5-(methyloxy)-2,3-dihydro-1 /-/-indole
Figure imgf000069_0003
To a solution 5-(methyloxy)-2,3-dihydro-1 /-/-indole (8.9 g, 59.8 mmol, 1.0 eq) in acetic acid (120 mL) was added dropwise acetic anhydride (6.1 g, 59.8 mmol, 1.0 eq). The mixture was heated at 60 0C for 15 minutes. The reaction was quenched by pouring into water (100 mL). After cooling, a grey precipitate was formed. The precipitate was filtered by a Buchner funnel and rinsed with water to afford 1-acetyl-5-(methyloxy)-2,3- dihydro-1 H-indole (10.5 g, 91% yield). 1H-NMR (CDCI3, 300 MHz) δ 8.13 (d, J = 6.3 Hz, 1 H), 6.74-6.71 (m, 2 H), 4.05 (t, 2 H), 3.78 (s, 3 H), 3.17 (t, 2 H), 2.21 (s, 3 H).
Step C/lntermediate A27: 1-acetyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole
Figure imgf000070_0001
To a solution of 5-(methyloxy)-2,3-dihydro-1 /-/-indole (10.4 g, 54.4 mmol, 1.0 eq) in acetic anhydride (150 mL) was added dropwise fuming nitric acid (3.43 g, 54.4 mmol, 1.0 eq) at 00C, then the mixture was stirred for 1 hour at room temperature. After stirring for 1 hour the yellow precipitate which formed was filtered via Buchner funnel and washed with water and dried in vacuo to give 1-acetyl-5-(methyloxy)-6-nitro-2,3-dihydro- 1 H-indole as a yellow solid (8.0 g, 62% yield). 1H-NMR (CDCI3, 300 MHz) δ 8.43 (s, 1 H), 7.31 (s, 1 H), 4.13 (t, 2H), 3.87 (s, 3H), 3.22 (t, 2H), 2.14 (s, 3H).
Step D/lntermediate A28: 5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole
Figure imgf000070_0002
To the solution of cone. HCI (20 mL) in methanol (40 mL) was added 1-acetyl-5-
(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole (7.8 g, 33 mmol) and the resulting mixture was refluxed for 4 hours. After cooling, the solvent was removed in vacuo, the residue was neutralized with saturated aqueous sodium hydrogencarbonate solution, and a brown precipitate formed. The precipitate was filtered via Buchner funnel, washed with water, and dried under reduced pressure to give 5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole as a brown solid (6.1 g, 94% yield). 1H-NMR (DMSO, 300 MHz) δ 7.11 (s, 1 H), 6.87 (s, 1 H), 5.59 (brs, 1 H), 3.78 (s, 3H), 3.43 (t, 2H), 2.97 (t, 2H).
Step E/lntermediate A24 : 1-{[ethyl(methyl)amino]acetyl}-5-(methyloxy)-2,3-dihydro-1 H- indol-6-amine
Figure imgf000071_0001
A solution of 5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole (33.6g, 173 mmol) in CH2CI2 (15OmL) was added dropwise to a mixture of bromoacetylchloride (54.5g, 346 mmol) and K2Cθ3 (53.Og, 381 mmol) in CH2CI2 (150 ml_). The resulting solution was stirred at
O0C for 1 hour, then warmed up RT for 4 hours. Water (150 ml.) was added and the mixture was extracted with CH2CI2 (2x10OmL). The organic phase was dried over Na2SO4, the solvent was removed to yield the 1-(bromoacetyl)-5-(methyloxy)-6-nitro-2,3- dihydro-1 H-indole (54.2g, 99%). 1HNMR (400 MHz, DMSO) δ ppm 2.47(s, 2H), 3.25 (m, 2 H), 3.87 (s, 3 H), 4.21 (m, 2 H), 7.34 (s, 1 H), 8.42 (s, 1 H). The 1-(bromoacetyl)-5- (methyloxy)-6-nitro-2,3-dihydro-1 H-indole (4.Og, 12.7 mmol) was dissolved in 50 mL of dichloromethane, then K2CO3 (4.4g, 31.7 mmol) and N-methylethanamine(1.5g, 25.4 mmol) in 10 mL dichloromethane were added and the reaction was stirred at RT for 3 hours. After filtration, the organic layers were washed with water (2x10OmL) and dried over Na2SO4. The solvent was was removed under reduced pressure and the derived residue (3.4 g, 11.59mmol) was dissolved in 10 mL of EA , 25 mL of MeOH and 15 mL of THF, then 0.8 g 10% Pd/C was added, the reaction was stirred at RT overnight under H2 pressure (65 psi). The catalyst was removed via filteration, and the solvent was evaporated under reduced pressure to afford 1-
{[ethyl(methyl)amino]acetyl}-5-(methyloxy)-2,3-dihydro-1 H-indol-6-amine as a yellow solid, 3.Og (97%).1HNMR (400 MHz, DMSO-d6) δ ppm: 0.97 (t, J=7.14 Hz,3 H), 2.20 (s, 3H), 2.46 (m, 2H), 2.94 (t, 2 H), 3.16 (s, 2H), 3.68 (s, 3H), 4.06 (t, J=8.42 Hz,2 H), 4.60 (s, 2H), 6.67 (s, 1 H), 7.52 (s, 1 H). Intermediate A29: 5-(methyloxy)-1 -{[methyl(propyl)amino]acetyl}-2,3-dihydro-1 H- indol-6-amine
Figure imgf000072_0001
1-(bromoacetyl)-5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indole (4.Og, 12.7 mmol) was dissolved in 50 ml. of dichloromethane, then K2CO3 (4.4g, 31.7 mmol) and Λ/-methyl-1- propanamine (1.9g, 25.4 mmol) in 10 ml. dichloromethane were added, the reaction was stirred at RT for 3 hours. After filtration, the organic layers were washed with water (2x10OmL) and dried over Na2SO4. The solvent was removed under reduced pressure and the resulting residue was dissolved in 10 ml. of EA , 25 ml. of MeOH and 15 ml. of THF, then 0.8 g 10% Pd/C was added and the reaction was stirred at RT overnight under H2 pressure (65 psi). The catalyst was removed via filtration and the solvent was removed under reduced pressure to afford 5-(methyloxy)-1- {[methyl(propyl)amino]acetyl}-2,3-dihydro-1 H-indol-6-amine as a yellow solid, 3.Og
(96%). 1HNMR (400 MHz, DMSO-d6) δ ppm: 0.81 (t, J=7.14 Hz,3 H), 1.40 (m, 2H), 2.21 (s, 3H), 2.35(m, 2H), 2.94 (t, J=8.24 Hz, 2 H), 3.17(s, 2H), 3.70 (s, 3H), 4.07 (t, J=8.33Hz,2 H), 4.60 (s, 2H), 6.67 (s, 1 H), 7.52 (s, 1 H).
Intermediate A30: 1-({methyl[2-(methyloxy)ethyl]amino}acetyl)-5-(methyloxy)-2,3- dihydro-1 H-indol-6-amine
Figure imgf000072_0002
The 1-(bromoacetyl)-5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indole (4.Og, 12.7 mmol) was dissolved in 50 ml. of dichloromethane, then K2CO3 (4.4g, 31.7 mmol) and Λ/-methyl-2- (methyloxy)ethanamine (1.4g, 15.2 mmol) in 10 ml. dichloromethane were added, the reaction was stirred at RT overnight. The reaction mixture was diluted with 100 ml. of water, the organic solvents were washed with water (2x10OmL), and dried over Na2SO4. The solvent was removed under reduced pressure and the resulting residue was dissolved in 10 mL of EA , 25 mL of MeOH and 15 ml. of THF, then 0.8 g 10% Pd/C was added, and the reaction was stirred at RT overnight under a balloon of H2 pressure. The catalyst was removed via filtration and the solvent was evaporated under reduced pressure to yield the 1-({methyl[2-(methyloxy)ethyl]amino}acetyl)-5-(methyloxy)-2,3- dihydro-1 H-indol-6-amine as a white solid (3.1 g, 89%). 1HNMR (400 MHz, DMSO-c/6) δ ppm: 2.28(s, 3H), 2.62 (t, J=5.77 Hz,2 H), 2.94 (t, J=8.24Hz, 2H), 3.19 (s, 3H), 3.25(s, 2H), 3.41 (t, J=5.77 Hz, 2 H), 3.68 (s, 3H), 4.04 (t, J=8.33Hz,2 H), 4.61 (s, 2H), 6.67 (s, 1 H), 7.52 (s, 1 H).
Intermediate A31 : 1 -{[ethyl(methyl)amino]acetyl}-6-(methyloxy)-1 ,2,3,4-tetrahydro- 7-quinolinamine
Figure imgf000073_0001
Step A/Intermediate A32: 3-chloro-Λ/-(4-hydroxyphenyl)propanamide
Figure imgf000073_0002
A solution of 4-aminophenol (250 g, 2.29 mol, 1 equiv) in CH2CI2 (2L) and saturated NaHCO3 (2L) was stirred for 5 min at 250C, then 3-chloropropanoyl chloride (314 g, 2.52 mol 1.1 equiv) was added dropwise and the reaction was stirred for an additional 3h at 250C. The solids were filtered and dried under high vacuum to afford 3-chloro-Λ/-(4- hydroxyphenyl)propanamide suitable for direct use in the next step. (25Og, crude). 1H NMR (400 MHz, DMSO) δ, 2.96-2.99 (m, 2 H), 4.06-4.10 (m, 2 H), 6.88-6.92 (m 2 H), 7.57-7.60 (m, 2 H), 9.38-9.39 (br s, 1 H), 10.1 (br s, 1 H).
Step B/lntermediate A33: 6-hydroxy-3,4-dihydro-2(1 H)-quinolinone
Figure imgf000074_0001
A mixture of 3-chloro-Λ/-(4-hydroxyphenyl)propanamide (122 g, 0.61 mol) and AICIs (327 g, 2.45 mol) were slowly heated with vigorous stirring at 18O0C (a thick melt formed) and after 5 hours the liquid was poured over ice. The solids were collected by filtration, washed with water, and recrystallized from MeOH to afford 6-hydroxy-3,4-dihydro-2(1H)- quinolinone (8Og, 40% yield). 1H NMR (400 MHz, DMSO) δ, 2.32-2.36 (m, 2 H), 2.74 (t, 2 H, J=7.2 Hz), 6.50 (m, 1 H), 6.54 (d, 1 H, J=2.8 Hz), 6.62(d, 1 H, J=8.4 Hz), 9.00 (br, 1 H), 9.78 (br, 1 H).
Step C/lntermediate A34: 6-(methyloxy)-3,4-dihydro-2(1 H)-quinolinone
Figure imgf000074_0002
To 6-hydroxy-3,4-dihydro-2(1 H)-quinolinone ( 90.8 g, 0.56 mol, 1 equiv) in CH3CN (2L) was added potassium carbonate (230.67 g, 1.67 mol, 3 equiv). The mixture was stirred for 1 h at 250C, then MeI (75 g, 0.53 mol, 0.95 equiv) was added and the mixture was maintained at 6O0C for 12 hours. The mixture was filtered and the filtrate was taken to a residue under reduce pressure. The crude 6-(methyloxy)-3,4-dihydro-2(1 H)-quinolinone was directly used in the next step. (128g, crude) 1H NMR (400 MHz, DMSO) δ, 2.34- 2.38 (m, 2 H), 2.79 (t, 2 H, J=7.2 Hz), 3.65 (s, 3 H), 6.67 (m, 1 H), 6.72-6.75 (m, 2 H), 9.86 (br s, 1 H). This procedure was reproduced multiple times to prepare >300g quantities of 6-(methyloxy)-3,4-dihydro-2(1 H)-quinolinone.
Step D/lntermediate A35: 6-(methyloxy)-7-nitro-3,4-dihydro-2(1 H)-quinolinone
Figure imgf000075_0001
To a solution of 6-(methyloxy)-3,4-dihydro-2(1 H)-quinolinone (209 g, 1.18 mol) in TFA (1500 ml.) was added NaNO2 (96 g, 1.41 mol, 1.2 equiv) at O0C, then the temperature was raised to 250C and the mixture was stirred for 4 hours. The mixture was poured into ice and the yellow precipitate was collected via filtration and dried under high vacuum at 50 0C. The crude product was recrystallized from ethyl acetate to afford 6- (methyloxy)-7-nitro-3,4-dihydro-2(1 H)-quinolinone (75.6g, 29%). 1H NMR (400 MHz, DMSO) δ 2.39-2.43 (m, 2 H), 2.92(t, 2 H, J=7.2 Hz), 3.82 (s, 3 H), 7.22 (s, 1 H), 7.32 (s, 1 H), 10.11 (br s, 1 H).
Step E/lntermediate A36: 6-(methyloxy)-7-nitro-1 ,2,3,4-tetrahydroquinoline
Figure imgf000075_0002
To a stirred solution of 6-(methyloxy)-7-nitro-3,4-dihydro-2(1 H)-quinolinone (75 g of, 0.34 mol, 1 equiv) in THF (1 L) was added BH3-DMS (10 M, 150 mL, 1.5 mol, 4.4equiv) dropwise at 25 0C. After the addition, the mixture was stirred for 6 h at 60 0C. The reaction was cooled and quenched with excess MeOH, concentrated under reduced pressure and purified via chromatography on SiO2 to afford 6-(methyloxy)-7-nitro-
1 ,2,3,4-tetrahydroquinoline (51.5 g, 73% yield). 1H NMR (400 MHz, DMSO) δ 1.73-1.80 (m, 2 H), 2.71 (t, 2 H, J=6.4 Hz), 3.13-3.16 (m, 2 H), 3.75 (s, 3 H), 5.83(br s, 1 H), 6.88(s, 1 H), 6.95(s, 1 H).
Step F/lntermediate A37: Λ/-ethyl-Λ/-methyl-2-[6-(methyloxy)-7-nitro-3,4-dihydro-1 (2H)- quinolinyl]-2-oxoethanamine
Figure imgf000076_0001
A solution of 6-(methyloxy)-7-nitro-1 ,2,3,4-tetrahydroquinoline (3.00 g, 14.41 mmol), Hunig's Base (12.55 ml_, 72.0 mmol), and bromo-acetylchloride (1.500 ml_, 18.01 mmol) was stirred at room temperature for 2 hours, at which time quinoline was observed to still remain. Additional bromo-acetylchloride (1.500 ml_, 18.01 mmol) was added, the reaction was stirred an additional 5 hours, N-ethylmethylamine (12.38 ml_, 144 mmol) was added, and the reaction was maintained for 16 hours at room temperature. The reaction was poured into saturated aqueous sodium bicarbonate and diluted with ethyl acetate. The organic layer was dried over sodium sulfate, taken to a residue under reduced pressure, and the residue was purified by silica gel chromatography to afford Λ/- ethyl-Λ/-methyl-2-[6-(methyloxy)-7-nitro-3,4-dihydro-1 (2/-/)-quinolinyl]-2-oxoethanamine (2.02 g, 45.6 % yield) as a dark red solid. 1H NMR (400 MHz, DMSOd6) δ ppm 8.35 (s, 1 H), 7.18 (s, 1 H), 3.89 (s, 3 H), 3.75 (s, 2 H), 3.30 (s, 2 H), 2.81 (t, J=6.12 Hz, 2 H), 2.36 - 2.47 (m, 2 H), 2.19 (s, 3 H), 1.80 - 1.97 (m, 2 H), 0.86 - 1.02 (m, 3 H).
Step G/lntermediate A31 : 1-{[ethyl(methyl)amino]acetyl}-6-(methyloxy)-1 ,2,3,4- tetrahydro-7-quinolinamine
Figure imgf000076_0002
A solution of N-ethyl-N-methyl-2-[6-(methyloxy)-7-nitro-3,4-dihydro-1 (2H)-quinolinyl]-2- oxoethanamine (2.02 g, 6.57 mmol) and 10% palladium on carbon (1.05 g) in methanol (55 ml.) was maintained under 40 psi of H2 gas in a pressure flask for 24 hours. The solution was purged with nitrogen, filtered through celite, taken to a residue under reduced pressure, and purified by column chromatography to afford 1- {[ethyl(methyl)amino]acetyl}-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinamine (1.16 g, 64 % yield) as a dark orange oil. 1H NMR (400 MHz, DMSOd6) δ ppm 6.82 (s, 1 H), 6.56 (s, 1 H), 4.54 (s, 2 H), 3.72 (s, 3 H), 3.61 (t, J=5.82 Hz, 2 H), 3.25 (s, 2 H), 2.57 (t, J=6.22 Hz, 2 H), 2.43 (q, J=7.02 Hz, 2 H), 2.20 (s, 3 H), 1.70 - 1.89 (m, 2 H), 0.84 - 1.03 (m, 3 H).
Intermediate A38: 1 -[(dimethylamino)acetyl]-6-(methyloxy)-1 ,2,3,4-tetrahydro-7- quinolinamine
Figure imgf000077_0001
Step A/Intermediate A39: 1-(bromoacetyl)-6-(methyloxy)-7-nitro-1 , 2,3,4- tetrahydroquinoline
Figure imgf000077_0002
A mixture of 6-(methyloxy)-7-nitro-1 ,2,3,4-tetrahydroquinoline (0.19 g, 0.91 mmol) and triethylamine (0.5 ml_, 3.7 mmol, Aldrich) in dichloromethane was treated with dropwise addition of bromoacetyl chloride (0.33 ml_, 4.0 mmol, Fluka). A catalytic amount of 4- dimethylaminopyridine was added and the reaction stirred overnight. The mixture was diluted with saturated aqueous ammonium chloride and extracted with dichloromethane. The organic layer was dried over magnesium sulfate, filtered, and concentrated to give 1-(bromoacetyl)-6-(methyloxy)-7-nitro-1 ,2,3,4-tetrahydroquinoline which was taken directly to the next step without determination of yield. RF = 0.3 (50% EtOAc/hexanes).
Step B/lntermediate A40: Λ/,Λ/-dimethyl-2-[6-(methyloxy)-7-nitro-3,4-dihydro-1 (2/-/)- quinolinyl]-2-oxoethanamine
Figure imgf000078_0001
1-(bromoacetyl)-6-(methyloxy)-7-nitro-1 ,2,3,4-tetrahydroquinoline (crude from previous reaction, assumed 0.91 mmol) was treated with excess dimethyl amine (4.5 ml_, 9.0 mmol, 2 M in THF, Aldrich), and the reaction stirred for 16 h. The mixture was partitioned between ethyl acetate and saturated aqueous sodium bicarbonate. The organic layer was dried over magnesium sulfate, filtered, and concentrated to give Λ/,Λ/-dimethyl-2-[6- (methyloxy)-7-nitro-3,4-dihydro-1 (2H)-quinolinyl]-2-oxoethanamine which was taken directly to the next step without determination of yield. 1H NMR (400 MHz, DMSO-c/6) δ ppm 1.85 - 1.95 (m, 2 H), 2.22 (s, 6 H), 2.82 (t, J=6.6 Hz, 2 H), 3.22 (bs, 2 H), 3.72 - 3.79 (m, 2 H), 3.89 (s, 3 H), 7.19 (s, 1 H), 8.37 (bs, 1 H); ESIMS (M+H)+ = 294.
Step C/lntermediate A38: 1-[(dimethylamino)acetyl]-6-(methyloxy)-1 ,2,3,4-tetrahydro-7- quinolinamine
Figure imgf000078_0002
Λ/,Λ/-dimethyl-2-[6-(methyloxy)-7-nitro-3,4-dihydro-1 (2/-/)-quinolinyl]-2-oxoethanamine (crude from previous reaction, assumed 0.91 mmol) was dissolved in methanol (5 ml_), treated with 10% palladium on carbon (40 mg, Aldrich), and stirred under 60 psi of hydrogen pressure for 16 h in a Fischer-Porter apparatus. The pressure was released, the reaction vessel evacuated, and back-filled with nitrogen twice. The mixture was filtered through celite, the filtrate was concentrated and purified by chromatography on SiO2 (0 to 20% methanol/dichloromethane) to provide 1-[(dimethylamino)acetyl]-6- (methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinamine (0.18 g, 0.68 mmol, 75% over three steps). 1H NMR (400 MHz, CDCI3) δ ppm 1.88 - 1.97 (m, J=6.5, 6.5, 6.5, 6.5 Hz, 2 H), 2.35 (s, 6 H), 2.65 (m, 2 H), 3.24 (s, 2 H), 3.74 - 3.80 (m, 2 H), 3.84 (s, 3 H), 6.55 (s, 1 H), 6.76 (very broad singlet due to amide rotomers, 1 H), aniline NH2 not visible probably due to fast exchange.
Intermediate A41 : 6-(methyloxy)-1 -(1 -pyrrolidinylacetyl)-1 ,2,3,4-tetrahydro-7- quinolinamine
Figure imgf000079_0001
Step A : Intermediate A42: 6-(methyloxy)-7-nitro-1-(1-pyrrolidinylacetyl)-1 , 2,3,4- tetrahydroquinoline
Figure imgf000079_0002
A solution of 6-(methyloxy)-7-nitro-1 ,2,3,4-tetrahydroquinoline (4.5g, 21.6 mmol) in CH2CI2 (4OmL) was added dropwise to a mixture of bromoacetylchloride (6.8g, 43.2 mmol) and K2COs (6.6g, 47.5 mmol) in CH2CI2 (40 ml_). The resulting solution was stirred at O0C for 1 hour, then warmed up to RT for 4 hours. Water (50 ml.) was added and extracted with CH2CI2 (2x50ml_). The organic phase was dried over Na2SO4, the solvent was removed under reduced pressure, and the resulting residue was dissolved in 50 ml. of dicholomethane. Potassium carbonate (6.2g, 44.4 mmol) and pyrrolidine (4.Og, 55.4 mmol) in 10 ml. dicholomethane were added and the reaction was stirred at RT for 3 hours. After filtration, the organic layers were washed with water (2x10OmL), and dried over Na2SO4. The solvent was removed to yield the 6-(methyloxy)-7-nitro-1-(1- pyrrolidinylacetyl)-1 ,2,3,4-tetrahydroquinoline as a yellow solid (6.8g, 97%). 1H NMR (400 MHz, DMSO-dβ) δ ppm 1.66 (br. s., 4 H), 1.85 (app d, J=6.04 Hz, 2 H), 2.49 (br. s., 4 H), 2.79 (t, J=6.00 Hz, 2 H), 3.35 (br. s., 2 H), 3.71 (br. s., 2 H), 3.86 (s, 3 H), 7.16 (s, 1 H), 8.39 (s, 1 H)
Step B/lntermediate A41 : 6-(methyloxy)-1-(1-pyrrolidinylacetyl)-1 ,2,3,4-tetrahydro-7- quinolinamine
Figure imgf000080_0001
The 6-(methyloxy)-7-nitro-1-(1-pyrrolidinylacetyl)-1 ,2,3,4-tetrahydroquinoline
(6.9 g, 21.6 mmol) was dissolved in 10 mL of EA , 25 mL of MeOH and 15 mL of THF, then 1.O g 10% Pd/C was added, the reaction was stirred at RT overnight under a balloon of H2 pressure. The catalyst was removed via filtration and the solvent was evaporated under reduced pressure to yield 6-(methyloxy)-1-(1-pyrrolidinylacetyl)- 1 ,2,3,4-tetrahydro-7-quinolinamine as a white solid, 6.Og (95%).1HNMR (400 MHz, DMSOd6) δ ppm: 1.09 (m, 1 H), 1.44 (m, 1 H), 1.62 (m, 1 H), 1.74 (m, 1 H), 2.14 (br, 1 H), 2.47(s, 1 H), 2.74(br s, 2H), 2.95 (m, 4 H), 3.50 (s, 2H), 3.68 (s, 3H), 4.04 (t, J=6.13Hz,2 H), 4.71 (s, 2H), 6.68 (s, 1 H), 7.51 (s, 1 H).
Intermediate A43: 1 -[(dimethylamino)acetyl]-4,4-dimethyl-6-(methyloxy)-1 ,2,3,4- tetrahydro-7-quinolinamine
Figure imgf000080_0002
Step A/Intermediate A44: 3-methyl-Λ/-[4-(methyloxy)phenyl]-2-butenamide
Figure imgf000081_0001
A mixture of p-anisidine (10 g, 81 mmol, Aldrich) and potassium carbonate (13.5 g, 97.5 mmol, Aldrich) in acetone (125 ml.) was cooled to 0 0C and treated with dimethyl acryloyl chloride (10.2 ml_, 89.3 mmol). The mixture was allowed to warm to room temperature overnight. The reaction was diluted with water and extracted with dichloromethane. The organic layer was separated, dried over magnesium sulfate, filtered, concentrated, and purified by chromatography on SiO2 (0 to 100% ethyl acetate/dichloromethane) to give 3- methyl-Λ/-[4-(methyloxy)phenyl]-2-butenamide (12.4 g, 67%) as a light violet solid. 1H NMR (400 MHz, DMSOd6) δ ppm 1.84 (s, 3 H), 2.13 (s, 3 H), 3.71 (s, 3 H), 5.82 (s, 1 H), 6.85 (d, J=9.0 Hz, 2 H), 7.52 (d, J=9.0 Hz, 2 H), 9.67 (s, 1 H); ESIMS (M+H)+ = 206.
Step B/lntermediate A45: 4,4-dimethyl-6-(methyloxy)-3,4-dihydro-2(1/-/)-quinolinone
Figure imgf000081_0002
A solution of 3-methyl-Λ/-[4-(methyloxy)phenyl]-2-butenamide (5.8 g, 28 mmol) in dichloromethane (250 ml.) was treated with aluminum trichloride (13 g, 97 mmol, Aldrich). The mixture was refluxed for 7 h, cooled, and then poured over ice. The organic layer was separated, dried over magnesium sulfate, filtered, and concentrated to give 4,4-dimethyl-6-(methyloxy)-3,4-dihydro-2(1 H)-quinolinone (4.5 g, 77%). 1H NMR (400 MHz, DMSOd6) δ ppm 1.20 (s, 6 H), 2.29 (s, 2 H), 3.71 (s, 3 H), 6.72 - 6.81 (m, 2 H), 6.83 (d, J=2.6 Hz, 1 H), 9.97 (s, 1 H).
Step C/lntermediate A46: 4,4-dimethyl-6-(methyloxy)-7-nitro-3,4-dihydro-2(1 /-/)- quinolinone
Figure imgf000082_0001
A solution of 4,4-dimethyl-6-(methyloxy)-3,4-dihydro-2(1 /-/)-quinolinone (4.45 g, 21.7 mmol) in trifluoroacetic acid (110 ml.) was treated at 0 0C with sodium nitrite (1.9 g, 28 mmol, Aldrich). The dark brown mixture was allowed to warm to room temperature overnight. Analysis of an aliquot of the reaction mixture by 1H NMR revealed that starting material still remained. Another 1.15 g (16.7 mmol) of sodium nitrite was added in three installments of 300/500/350 mg at room temperature and the reaction stirred for 3/3/20 h respectively. The reaction mixture was concentrated, dissolved in dichloromethane, and washed with saturated aqueous sodium bicarbonate. The organic layer was separated, the aqueous layer was filtered (due to emulsions) and back- extracted with dichloromethane. The combined organic layers were dried over magnesium sulfate, filtered, concentrated, and triturated with dichloromethane/diethyl ether/hexanes. The mixture was filtered to obtain pure product (3.6 g) and the filtrate was purified by column chromatography on SiO2 (0 to 100% ethyl acetate/ dichloromethane) to obtain another 350 mg of 4,4-dimethyl-6-(methyloxy)-7-nitro-3,4-dihydro-2(1 H)-quinolinone (a total of 3.97 g, 15.8 mmol, 73%). 1H NMR (400 MHz, DMSOd6) δ ppm 1.28 (s, 6 H), 2.39 (s, 2 H), 3.92 (s, 3 H), 7.21 (s, 1 H), 7.40 (s, 1 H), 10.25 (s, 1 H).
Step D/lntermediate A47: 4,4-dimethyl-6-(methyloxy)-7-nitro-1 ,2,3,4-tetrahydroquinoline
Figure imgf000082_0002
A solution of 4,4-dimethyl-6-(methyloxy)-7-nitro-3,4-dihydro-2(1 H)-quinolinone (2.5 g, 10 mmol) in tetrahydrofuran (45 ml.) was treated with borane-dimethylsulfide complex (20.5 ml_, 41 mmol, 2 M in THF, Acros Organics). The mixture was heated at reflux for 16 h, cooled, quenched carefully via dropwise of excess methanol, concentrated, and purified by column chromatography on SiO2 (O to 100% ethyl acetate/dichloromethane) to obtain 4,4-dimethyl-6-(methyloxy)-7-nitro-1 ,2,3,4- tetrahydroquinoline (1.33 g, 5.63 mmol, 56%). 1H NMR (400 MHz, DMSOd6) δ ppm 1.26 (s, 6 H), 1.58 - 1.65 (m, 2 H), 3.13 - 3.21 (m, 2 H), 3.79 (s, 3 H), 5.95 (bs, 1 H), 6.96 (s, 1 H), 7.06 (s, 1 H).
Step E/lntermediate A48: 1-(bromoacetyl)-4,4-dimethyl-6-(methyloxy)-7-nitro-1 ,2,3,4- tetrahydroquinoline
Figure imgf000083_0001
4,4-dimethyl-6-(methyloxy)-7-nitro-1 ,2,3,4-tetrahydroquinoline (1.32 g, 5.59 mmol) was dissolved in tetrahydrofuran (50 ml_). This solution was treated with potassium carbonate (1.54 g, 1 1.2 mmol, Aldrich) and cooled to 0 0C. Bromoacetyl chloride (0.93 ml_, 11 mmol, Fluka) was added (white precipitation observed) and the reaction stirred for 15 min. The reaction mixture was then diluted with water and extracted with ethyl acetate. The organic layer was dried over magnesium sulfate, filtered, and concentrated to give 1-(bromoacetyl)-4,4-dimethyl-6-(methyloxy)-7-nitro-1 ,2,3,4-tetrahydroquinoline (assumed 5.59 mmol, 100%). This material was analyzed by 1H NMR (amide rotomers were observed) and taken to bromide displacement reactions directly without further purification. 1H NMR (400 MHz, DMSOd6) δ ppm 1.33 (s, 6 H), 1.75 - 1.86 (m, 2 H), 3.72 - 3.82 (m, 2 H), 3.95 (s, 3 H), 4.31 - 4.69 (two singlets at 4.37 and 4.63, 2 H), 7.27 (s, 1 H), 8.28 (very broad singlet, 1 H).
Step F/lntermediate A49: {2-[4,4-dimethyl-6-(methyloxy)-7-nitro-3,4-dihydro-1 (2/-/)- quinolinyl]-2-oxoethyl}dimethylamine
Figure imgf000083_0002
A solution of 1-(bromoacetyl)-4,4-dimethyl-6-(methyloxy)-7-nitro-1 , 2,3,4- tetrahydroquinoline (1.0 g, 2.8 mmol) in tetrahydrofuran (35 ml.) was treated with potassium carbonate (1.3 g, 9.4 mmol) and dimethyl amine (10 ml_, 20 mmol, 2 M in THF, Aldrich). The mixture was heated in sealed tube at 55 0C for 1 h, cooled, diluted with water, and extracted with ethyl acetate. The organic layer was separated, dried over magnesium sulfate, filtered, and concentrated to give {2-[4,4-dimethyl-6-(methyloxy)-7- nitro-3,4-dihydro-1 (2H)-quinolinyl]-2-oxoethyl}dimethylamine which was taken directly to the next step. ESIMS (M+H)+ = 322.
Step G/lntermediate A43: 1-[(dimethylamino)acetyl]-4,4-dimethyl-6-(methyloxy)-1 ,2,3,4- tetrahydro-7-quinolinamine
Figure imgf000084_0001
{2-[4,4-dimethyl-6-(methyloxy)-7-nitro-3,4-dihydro-1 (2H)-quinolinyl]-2- oxoethyl}dimethylamine (assumed 2.8 mmol) was dissolved in a mixture of ethyl acetate (50 ml.) and water (5 ml.) and transferred to a Fischer Porter apparatus. 10% Palladium on carbon (500 mg, Aldrich) was added followed by methanol (5 ml.) and the mixture stirred under 60 psi hydrogen pressure for 2 h. The pressure was released, the reaction vessel evacuated, and back-filled with nitrogen twice. The mixture was filtered through celite and the filtrate was concentrated to provide 1-[(dimethylamino)acetyl]-4,4- dimethyl-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinamine (724 mg, 2.49 mmol, 89% over three steps). 1H NMR (400 MHz, DMSOd6) δ ppm 1.20 (s, 6 H), 1.62 - 1.68 (m, 2 H), 2.20 (s, 6 H), 3.17 (s, 2 H), 3.62 - 3.69 (m, 2 H), 3.75 (s, 3 H), 4.58 (bs, 2 H), 6.56 - 6.94 (s at 6.71 on top of a very broad singlet caused by amide rotomers, 2 H); ESIMS (M+H)+ = 292.
Intermediate A50: 4,4-dimethyl-6-(methyloxy)-1 -(1 -pyrrolidinylacetyl)-1 ,2,3,4- tetrahydro-7-quinolinamine
Figure imgf000085_0001
Step A/Intermediate A51 : 4,4-dimethyl-6-(methyloxy)-7-nitro-1-(1-pyrrolidinylacetyl)- 1 ,2,3,4-tetrahydroquinoline
Figure imgf000085_0002
A solution of 1-(bromoacetyl)-4,4-dimethyl-6-(methyloxy)-7-nitro-1 , 2,3,4- tetrahydroquinoline (1.0 g, 2.8 mmol) in tetrahydrofuran (35 ml.) was treated with potassium carbonate (1.3 g, 9.4 mmol) and pyrrolidine (1.5 ml_, 18 mmol, Aldrich). The mixture was heated in sealed tube at 55 0C for 1 h, cooled, diluted with water, and extracted with ethyl acetate. The organic layer was separated, dried over magnesium sulfate, filtered, and concentrated to give 4,4-dimethyl-6-(methyloxy)-7-nitro-1-(1- pyrrolidinylacetyl)-1 ,2,3,4-tetrahydroquinoline which was taken directly to the next step. ESIMS (M+H)+ = 348.
Step B/lntermediate A50: 4,4-dimethyl-6-(methyloxy)-1-(1-pyrrolidinylacetyl)-1 ,2,3,4- tetrahydro-7-quinolinamine
Figure imgf000085_0003
4,4-dimethyl-6-(methyloxy)-7-nitro-1 -(1 -pyrrolidinylacetyl)-1 ,2,3,4-tetrahydroquinoline (assumed 2.8 mmol) was dissolved in a mixture of ethyl acetate (50 ml.) and water (5 ml.) and transferred to a Fischer Porter apparatus. 10% Palladium on carbon (500 mg, Aldrich) was added followed by methanol (5 ml.) and the mixture stirred under 60 psi hydrogen pressure for 2 h. The pressure was released, the reaction vessel evacuated, and back-filled with nitrogen twice. The mixture was filtered through celite and the filtrate was concentrated to provide 4,4-dimethyl-6-(methyloxy)-1-(1-pyrrolidinylacetyl)-1 ,2,3,4- tetrahydro-7-quinolinamine (790 mg, 89% over three steps). 1H NMR (400 MHz, DMSO- d6) δ ppm 1.19 (s, 6 H), 1.61 - 1.71 (m, 6 H), 2.50 - 2.54 (m, signal underneath DMSO, 4 H), 3.32 - 3.35 (m, signal underneath H2O, 2 H), 3.62 - 3.69 (m, 2 H), 3.75 (s, 3 H), 4.58 (bs, 2 H), 6.62 - 7.00 (s at 6.71 on top of a very broad singlet caused by amide rotomers, 2 H).
Intermediate A52: 5-methyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1 -piperazinyl]- 1 -piperidinylj-aniline
Figure imgf000086_0001
Step A/Intermediate A53: 1-{1-[2-methyl-5-(methyloxy)-4-nitrophenyl]-4-piperidinyl}-4- (methylsulfonyl)-piperazine
Figure imgf000086_0002
A mixture of 1-fluoro-2-methyl-5-(methyloxy)-4-nitrobenzene (0.90 g, 4.9 mmol), 1- (methylsulfonyl)-4-(4-piperidinyl)piperazine (1.20 g, 4.9 mmol) and K2CO3 (0.81 g, 5.8 mmol) in DMSO (15 ml.) was heated at 100 0C for 48 h. The reaction was cooled to rt, poured into H2O and extracted exhaustively with DCM. The combined organics were washed with H2O then dried over MgSO4. The resultant solution was concentrated onto silica and purified by flash chromatography to afford 1-{1-[2-methyl-5-(methyloxy)-4- nitrophenyl]-4-piperidinyl}-4-(methylsulfonyl)piperazine (1.09 g, 55%). 1H NMR (400 MHz, DMSOd6) δ ppm 7.77 (s, 1 H), 6.69 (s, 1 H), 3.90 (s, 3 H), 3.29 - 3.38 (m, 2 H), 3.06 - 3.15 (m, 4 H), 2.87 (s, 3 H), 2.68 - 2.79 (m, 2 H), 2.59 - 2.66 (m, 4 H), 2.40 - 2.48 (m, 1 H), 2.20 (s, 3 H), 1.80 - 1.92 (m, 2 H), 1.49 - 1.67 (m, 2 H).
Step B/lntermediate A52: 5-methyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]- 1-piperidinyl}-aniline
Figure imgf000087_0001
Sodium borohydride (0.30 g, 7.93 mmol) was added in portions to a stirring suspension of 1-{1-[2-methyl-5-(methyloxy)-4-nitrophenyl]-4-piperidinyl}-4-(methylsulfonyl)piperazine (1.09 g, 2.64 mmol) and NiCI2-6H2O (0.19 g, 0.8 mmol) in THF (10 ml.) and MeOH (20 ml.) at rt. The reaction mixture was allowed to stir at rt for 2 h and then it was concentrated in vacuo. The residue was taken up in DCM and passed through a bed of celite. The resultant filtrate was then concentrated onto silica gel and purified by flash chromatography to afford 5-methyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]- 1-piperidinyl}aniline (0.58 g, 57%). 1H NMR (400 MHz, DMSOd6) δ ppm 6.55 (s, 1 H), 6.43 (s, 1 H), 4.30 (s, 2 H), 3.70 (s, 3 H), 3.05 - 3.15 (m, 4 H), 2.89 - 2.96 (m, 2 H), 2.86 (s, 3 H), 2.52 - 2.65 (m, 6 H), 2.26 - 2.41 (m, 1 H), 2.06 (s, 3 H), 1.74 - 1.85 (m, 2 H), 1.43 - 1.63 (m, 2 H).
Intermediate A54: 5-(methyloxy)-1 -(1 -pyrrolidinylacetyl)-2,3-dihydro-1 H-indol-6-
Figure imgf000087_0002
Step A/Intermediate A55: 5-(methyloxy)-6-nitro-1-(1-pyrrolidinylacetyl)-2,3-dihydro-1 /-/- indole
Figure imgf000088_0001
To a 0 0C slurry of 5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole hydrogen chloride (743 mg, 3.22 mmol) and DIPEA (2.8 ml_, 16.11 mmol) was added bromoacetyl chloride (0.3 ml_, 3.54 mmol) dropwise. After stirring at rt for 40 min pyrrolidine (1 ml_, 12.89 mmol) was added and the reaction mixture was stirred overnight. The resulting mixture was concentrated and the residue was partitioned between EtOAc (200 ml.) and water (200 ml_). The organic layer was washed with a saturated NaCI solution (100 ml_). The aqueous layers were back-extracted with EtOAc (100 ml_). The organic layers were combined, dried (Na2SO4) and concentrated to obtain 5-(methyloxy)-6-nitro-1-(1- pyrrolidinylacetyl)-2,3-dihydro-1 /-/-indole as a brown solid (754 mg, 77%). 1H NMR (400 MHz, DMSO-d6) δ ppm 1.71 (br s, 4H), 2.58 (br s, 4H), 3.23 (t, J=8.42 Hz, 2H), 3.40 (s, 2H), 3.89 (s, 3H), 4.20 (t, J=8.42 Hz, 2H), 7.34 (s, 1 H), 8.38 (s, 1 H); ESIMS (M+H)+ = 305.97.
Step B/lntermediate A54: 5-(methyloxy)-1-(1-pyrrolidinylacetyl)-2,3-dihydro-1 /-/-indol-6- amine
Figure imgf000088_0002
To a solution of 5-(methyloxy)-6-nitro-1-(1-pyrrolidinylacetyl)-2,3-dihydro-1 /-/-indole (750 mg, 2.46 mmol) in THF (20 ml.) and MeOH (40 ml.) was added NiCI2.6H2O (source: Riedel De Halen) (175 mg, 0.74 mmol), followed by addition of NaBH4 (280 mg, 7.4 mmol) in small portions. After stirring overnight the reaction mixture was concentrated onto Celite and purified by silica gel chromatography using 0-10% MeOH (containing 0.2% NH3)/CH2CI2 to obtain 5-(methyloxy)-1-(1-pyrrolidinylacetyl)-2,3-dihydro-1 /-/-indol-6- amine as a brown solid (364 mg, 54%). 1H NMR (400 MHz, DMSO-d6) δ ppm 1.70 (br s, 4H), 2.56 (br s, 4H), 2.97 (t, J=8.24 Hz, 2H), 3.71 (s, 3H), 4.06 (t, J=8.33 Hz, 2H), 4.63 (s, 2H), 6.70 (s, 1 H), 7.54 (s, 1 H) a CH2 signal with 2 protons is missing, may overlap with water peak in sample; ESIMS (M+H)+ = 276.11. Intermediate A56: 5-(methyloxy)-1 -[(methyloxy)acetyl]-2,3-dihydro-1 H-indol-6- amine
Figure imgf000089_0001
Step A/Intermediate A57: 5-(methyloxy)-1-[(methyloxy)acetyl]-6-nitro-2,3-dihydro-1 /-/- indole
Figure imgf000089_0002
A slurry of 5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole (3.2 g, 13.87 mmol) in THF (100 mL) was treated with diisopropylethylamine (4.83 ml_, 27.7 mmol), followed by (methyloxy)acetyl chloride (1.903 mL, 20.81 mmol). The resulting yellow slurry was stirred at rt for 3 days, then diluted with CH2CI2 (300 mL), washed with water (150 mL), a saturated NaCI solution (150 mL), dried (Na2SO4) and concentrated to obtain 5- (methyloxy)-1-[(methyloxy)acetyl]-6-nitro-2,3-dihydro-1 /-/-indole as an orange solid (3.96 g). 1H NMR (400 MHz, DMSOd6) δ ppm 3.24 (t, J=8.42 Hz, 2H), 3.36 (s, 3H), 3.89 (s, 3H), 4.07 (t, J=8.42 Hz, 2H), 4.19 (s, 2H), 7.35 (s, 1 H), 8.47 (s, 1 H); ESIMS (M+H)+ = 266.87.
Step B/lntermediate A56: 5-(methyloxy)-1-[(methyloxy)acetyl]-2,3-dihydro-1 /-/-indol-6-
Figure imgf000089_0003
A slurry of 5-(methyloxy)-1-[(methyloxy)acetyl]-6-nitro-2,3-dihydro-1 /-/-indole (3.96 g, 14.87 mmol) and NiCI2 6H2O (1.061 g, 4.46 mmol) in THF (50 mL) and MeOH (100 mL) was treated with NaBH4 (1.69 g, 44.6 mmol) in small portions. After 5 minutes the reaction mixture was concentrated onto Celite and purified by silica gel chromatography using THF. The crude product was triturated using CH2CI2 and Et2O to obtain 5-
(methyloxy)-1-[(methyloxy)acetyl]-2,3-dihydro-1 /-/-indol-6-amine as a white solid (884 mg, 25%). 1H NMR (400 MHz, DMSO-d6) δ ppm 2.98 (t, J=8.24Hz, 2H), 3.34 (s, 3H), 3.71 (s, 3H), 3.93 (t, J=8.,33 Hz, 2H), 4.12 (s, 2H), 4.67 (s, 2H), 6.71 (s, 1 H), 7.54 (s, 1 H); ESIMS (M+H)+ = 237.4.
Intermediate A58: 2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1 - piperidinyljaniline
__/~Λ /=\
H3CO2S^ ^^ *-( O
Step A/Intermediate A59: 1 ,1-dimethylethyl 4-(2-iodoethyl)-1-piperidinecarboxylate
Figure imgf000090_0001
To Λ/-Boc-4-piperidone-ethanol (5.00 g, 21.8 mmol) in THF (100 mL) was added triphenylphosphine (6.29 g, 24.0 mmol), imidazole (1.64 g, 24.1 mmol) and iodine (6.09 g, 24.0 mmol). The reaction was stirred at rt for 2 h. The mixture was filtered through Celite® purified by flash chromatography to provide 1 ,1-dimethylethyl 4-(2-iodoethyl)-1- piperidinecarboxylate (6.31 g, 18.6 mmol, 85%). 1H NMR (400 MHz, CDCI3) δ ppm 1.07 (dq, J=12.2, 4.3 Hz, 2 H), 1.42 (s, 9 H), 1.49 - 1.58 (m, 1 H), 1.62 (d, J= 13.2 Hz, 2 H), 1.75 (q, J=IA Hz, 2 H), 2.67 (td, J=12.9, 2.5 Hz, 2 H), 3.18 (t, J=IA Hz, 2 H), 4.06 (d, J=13.0 Hz, 2 H).
Step B /Intermediate A60: 1 ,1-dimethylethyl 4-[2-(methylthio)ethyl]-1- piperidinecarboxylate
Figure imgf000090_0002
To 1 ,1-dimethylethyl 4-(2-iodoethyl)-1 -piperidinecarboxylate (3.13 g, 9.23 mmol) in DMF (50 mL) was added sodium thiomethoxide (0.661 g, 10.4 mmol) in one portion. The reaction was stirred for 3 days. Upon completion by TLC, the reaction was diluted with EtOAc (250 mL), washed with H2O (4 times), dried (MgSO4) and concentrated to provide 1 ,1-dimethylethyl 4-[2-(methylthio)ethyl]-1 -piperidinecarboxylate (2.37 g, 9.20 mmol, 99%, approximately 93% pure, used without further purification). 1H NMR (400 MHz, CDCI3) δ ppm 1.02 - 1.13 (m, 2 H), 1.42 (s, 9 H), 1.48 - 1.55 (m, 3 H), 1.63 (d, J=12.5 Hz, 2 H), 2.07 (s, 3 H), 2.45 - 2.53 (m, 2 H), 2.65 (td, J=12.8, 2.6 Hz, 2 H), 4.02 - 4.09 (m, 2 H).
Step C/lntermediate A61 : 1 ,1-dimethylethyl 4-[2-(methylsulfonyl)ethyl]-1- piperidinecarboxylate
Figure imgf000091_0001
To 1 ,1-dimethylethyl 4-[2-(methylthio)ethyl]-1-piperidinecarboxylate (2.37 g, 9.20 mmol) in DCM (20 ml.) at O0C was added mefa-chloroperoxybenzoic acid (5.29 g, 77% by weight, 23.0 mmol) in portions. The reaction was stirred overnight. Additional meta- chloroperoxybenzoic acid (2.50 g, 77% by weight, 14.5 mmol) was added and the reaction was stirred for 2 days. The mixture was washed with saturated NaHCOs, brine and H2O. The organic layer was dried (MgSO4) and concentrated. Purification by flash chromatography provided 1 ,1-dimethylethyl 4-[2-(methylsulfonyl)ethyl]-1- piperidinecarboxylate (1.44 g, 4.90 mmol, 54%). 1H NMR (400 MHz, CDCI3) δ ppm 1.09 - 1.20 (m, 2 H), 1.45 (s, 9 H), 1.51 - 1.62 (m, 1 H), 1.67 (dd, J=13.4, 2.0 Hz, 2 H), 1.77 - 1.85 (m, 2 H), 2.63 - 2.73 (m, 2 H), 2.91 (s, 3 H), 2.99 - 3.05 (m, 2 H), 4.08 - 4.15 (m, 2 H).
Step D/lntermediate A62: 4-[2-(methylsulfonyl)ethyl]piperidine
Figure imgf000091_0002
A solution of ,1-dimethylethyl 4-[2-(methylsulfonyl)ethyl]-1-piperidinecarboxylate (1.4 g, 4.9 mmol) and DCM (1 10 ml.) was cooled with an ice/H2O bath. TFA (10 ml_, 130 mmol) was added in one portion. Upon completion the reaction mixture was quenched at O0C with 6N NaOH (30 ml_), extracted with DCM, EA, dried (MgSO4) and concentrated to provide 4-[2-(methylsulfonyl)ethyl]piperidine (0.81 g, 4.3 mmol, 87%). 1 H NMR (400 MHz, CDCI3) δ ppm 1.14 - 1.26 (m, 2 H), 1.49 - 1.59 (m, 1 H), 1.70 (d, J=13.6 Hz, 2 H), 1.76 - 1.84 (m, 2 H), 2.27 (br. s., 1 H), 2.61 (td, J=12.3, 2.6 Hz, 2 H), 2.90 (s, 3 H), 2.99 - 3.05 (m, 2 H), 3.1 1 (ddd, J=12.4, 3.3, 3.0 Hz, 2 H). Step E/lntermediate A63: 1-[3-(methyloxy)-4-nitrophenyl]-4-[2- (methylsulfonyl)ethyl]piperidine
Figure imgf000092_0001
To 5-fluoro-2-nitrophenyl methyl ether (0.41 g, 2.1 mmol), 4-[2- (methylsulfonyl)ethyl]piperidine (0.74 g, 4.3 mmol) and K2CO3 (1.0 g, 7.5 mmol) was added DMSO. The mixture was stirred at room temperature overnight and allowed to stand for 25 days. The mixture was diluted with EA and washed with water (6 times). The organic layer was dried (MgSO4), filtered, and concentrated. Purification by flash column chromatography provided 1-[3-(methyloxy)-4-nitrophenyl]-4-[2- (methylsulfonyl)ethyl]piperidine (0.43 g, 1.3 mmol, 58%). 1H NMR (400 MHz, CDCI3) δ 1.38 - 1.57 (m, 3 H), 1.72 - 1.80 (m, 1 H), 1.83 - 1.89 (m, 4 H), 2.90 (s, 3 H), 2.93 - 2.98 (m, 1 H), 3.00 - 3.07 (m, 2 H), 3.87 (d, J=12.8 Hz, 2 H), 3.93 (s, 3 H), 6.48 (d, J=9.2 Hz, 1 H), 6.55 (br. s., 1 H), 7.97 (d, J=9.3 Hz, 1 H).
Step F/lntermediate A58: 2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1- piperidinyl}aniline
_ /~Λ /=\
/ \ N~(\ ff— NH9
H3CO2S^ V^ ^-< p H3C
To 1-[3-(methyloxy)-4-nitrophenyl]-4-[2-(methylsulfonyl)ethyl]piperidine (0.42 g, 1.3 mmol) in THF (60 ml.) cooled to 0 0C was added SnCb (1.0 g, 5.4 mmol) dissolved in cone. HCI dropwise. The solution was stirred overnight at room temperature (TLC confirms consumption of starting material). The mixture was cooled to 0 0C and quenched with 6 N NaOH (25 ml_). The solution was poured into EtOAc and H2O was washed with brine, back extracted with EtOAc, dried (MgSO4), filtered, and rotovaped down to give 2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1-piperidinyl}aniline (0.39 g, 1.3 mmol, 96%). 1H NMR (400 MHz, CDCI3) δ 1.18 - 1.29 (m, 2 H), 1.31 - 1.43 (m, 1 H), 1.57 - 1.65 (m, 2 H), 1.70 (d, J=12.5 Hz, 2 H), 2.37 - 2.43 (m, 2 H), 2.92 (s, 3 H), 3.07 - 3.14 (m, 2 H), 3.34 (d, J= 11.5 Hz, 2 H), 3.69 (s, 3 H), 4.16 (br. s., 2 H), 6.25 (d, J=7.7 Hz, 1 H), 6.40 - 6.48 (m, 2 H).
Intermediate A64: 2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1 -piperazinyl]-1 - piperidinyljaniline
Figure imgf000093_0001
Step A/Intermediate A65: 1-[3-(methyloxy)-4-nitrophenyl]-4-piperidinol
Figure imgf000093_0002
A mixture of 4-fluoro-2-(methyloxy)-1 -nitrobenzene (7.52 g, 44 mmol), 4- hydroxypiperidine (4.45 g, 44 mmol) and potassium carbonate (8.Og, 88 mmol) in 100 ml. DMSO was stirred for 72h. The reaction was diluted with water and extracted twice with ethyl acetate. The combined organic layers were washed with brine, dried over MgSO4, and concentrated to give 1-[3-(methyloxy)-4-nitrophenyl]-4-piperidinol (10.9 g, 99% yield). 1H NMR (400 MHz, d6-DMSO) δ 7.84 (d, J = 9.2 Hz, 1 H), 6.54 (d, J = 9.6 Hz, 1 H), 6.46 (s, 1 H), 4.73 (d, J = 4.4 Hz, 1 H), 3.86 (s, 3H), 3.80-3.69 (m, 3H), 3.18-3.12 (m, 2H), 1.79-1.76 (m, 2H), 1.42-1.34 (m, 2H).
Step B/lntermediate A66: 1-[3-(methyloxy)-4-nitrophenyl]-4-piperidinone
Figure imgf000094_0001
To a solution of (10 g, 40 mmol) in 400 ml. DCM was added sodium bicarbonate (16.8 g, 200 mmol), water (0.72 ml_, 40 mmol) and 1 ,1 ,1 -tri(acetyloxy)-1 , 1 -dihydro-1 ,2- benziodoxol-3-(1 H)-one (Dess-Martin periodinone, 20.4 g, 48 mmol, Aldrich). After 1.5 h, the reaction still contained starting material and was not progressing. The reaction was quenched with equal parts sat'd NaHCOs and sat'd Na2S2θ3. After stirring for 1 hour, the layers were separated. The aqueous phase was extracted with DCM. The combined organics were washed with water and brine, dried over MgSO4, concentrated onto silica gel and purified by flash column chromatography. Fractions containing product were concentrated to give 1.8 g of desired product. Fractions containing starting material were concentrated and resubjected to the above conditions. After workup and purification, a further 6.1 g of product was collected to give 7.9 g (79%) of 1-[3- (methyloxy)-4-nitrophenyl]-4-piperidinone. 1H NMR (400 MHz, d6-DMSO) δ 7.89 (d, J = 9.2 Hz, 1 H), 6.59 (d, J = 9.2 Hz, 1 H), 6.50 (d, J = 1.2 Hz, 1 H), 3.90 (s, 3H), 3.82-3.78 (m, 4 H), 2.49-2.46 (m, 4H).
Step C/lntermediate A67: 1-{1-[3-(methyloxy)-4-nitrophenyl]-4-piperidinyl}piperazine
Figure imgf000094_0002
To a solution of 1-[4-amino-3-(methyloxy)phenyl]-4-piperidinone (combined batches) (17.35 g, 69.3 mmol) in toluene (600 ml.) was added sequentially, TEA (25 ml_, 179.4 mmol), 1-Boc-piperazine (25.36 g, 136.2 mmol), and HOAc (6.0 ml_, 105.9 mmol). The solution was stirred at rt for 30 min. Sodium triacetoxyborohydride (12.2 g, 57.6 mmol) was added in one portion and stirred for 30 min. A second portion of sodium triacetoxyborohydride (12.2 g, 57.6 mmol) was added and stirring continued for 30 min. A third portion of sodium triacetoxyborohydride (12.2 g, 57.6 mmol) was added. The reaction was stirred (3 hours) until ketone was consumed as indicated by TLC. The reaction was quenched with a saturated solution of NaHCO3 (600 ml.) and stirred 2 days. The solution was separated and extracted with DCM (x3), dried with MgSO4, filtered and concentrated. The resultant solid was dissolved in DCM (600 ml.) and cooled to 0 0C. TFA (1 10 ml.) was added; the reaction was warmed to rt and stirred overnight. The reaction was cooled to 0 0C and quenched with 6N NaOH (320 ml.) dropwise. The solution was separated and extracted with DCM (x3), dried with MgSO4, filtered and concentrated. Purification by flash chromatography provided 1-{1-[3- (methyloxy)-4-nitrophenyl]-4-piperidinyl}piperazine (18.03 g, 56.10 mmol, 81%) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.41 (qd, J=12.0, 3.7 Hz, 2 H) 1.77 - 1.84 (m, 2 H) 2.38 - 2.47 (m, 5 H) 2.67 - 2.73 (m, 4 H) 2.88 - 2.98 (m, 2 H) 3.32 (br. s., 1 H) 3.88 (s, 3 H) 4.03 (d, J=12.8 Hz, 2 H) 6.48 (d, J=2.6 Hz, 1 H) 6.56 (dd, J=9.5, 2.6 Hz, 1 H) 7.85 (d, J=9.2 Hz, 1 H).
Step D/lntermediate A68: 1-{1-[3-(methyloxy)-4-nitrophenyl]-4-piperidinyl}-4- (methylsulfonyl)piperazine
Figure imgf000095_0001
To a suspension of 1-{1-[3-(methyloxy)-4-nitrophenyl]-4-piperidinyl}piperazine (5.Og, 11.6mmol), methane sulfonyl chloride (1.4ml_, 17.5mmol) and DCM (20OmL) was added TEA (8.1 ml_, 58.2mmol). The reaction was stirred at rt and monitored by TLC. After complete consumption of the starting material the clear yellow solution was concentrated onto silica gel and purified by chromatography to afford 1-{1-[3-(methyloxy)-4- nitrophenyl]-4-piperidinyl}-4-(methylsulfonyl)piperazine as a yellow solid (3.54g, 76%). 1H NMR (400 MHz, DMSO-d6) δ 7.85 (d, J = 9.5 Hz, 1 H), 6.57 (dd, J = 9.2, 2.6 Hz, 1 H), 6.48 (d, J = 2.6 Hz, 1 H), 4.09 - 4.00 (m, 2H), 3.89 (s, 3H), 3.10 - 3.03 (m, 4H), 2.99 - 2.90 (m, 2H), 2.84 (s, 3H), 2.61 - 2.52 (m, 5H), 1.85 - 1.77 (m, 2H), 1.50 - 1.37 (m, 2H).
Step E/lntermediate A64: 2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1- piperidinyl}aniline
Figure imgf000096_0001
NaBH4 (1.18g, 31.1 mmol) was added carefully in portions (exothermic) to a suspension of 1-{1-[3-(methyloxy)-4-nitrophenyl]-4-piperidinyl}-4-(methylsulfonyl)-piperazine (3.54g, 8.9mmol), nickel(ll)chloride hexahydrate (1.06g, 4.4mmol), MeOH (10OmL) and THF (5OmL) at 0 0C. The ice bath was removed and the reaction mixture was warmed to rt. TLC analysis indicated the complete consumption of the starting material. The reaction mixture was concentrated onto silica gel and purified by flash chromatography to afford 2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1-piperidinyl}aniline (2.93g, 90%) as a colorless solid. 1H NMR (400 MHz, DMSOd6) δ 6.49 - 6.46 (m, 2H), 6.27 (dd, J = 8.5, 2.6 Hz, 1 H), 4.18 (bs, 2H), 3.71 (s, 3H), 3.44 - 3.38 (m, 2H), 3.11 - 3.04 (m, 4H), 2.84 (s, 3H), 2.61 - 2.54 (m, 4H), 2.35 - 2.26 (m, 1 H), 1.80- 1.77 (m, 2H), 1.56 - 1.46 (m, 2H).
Intermediate A69: 5-fluoro-4-[4-(1 -methylethyl)-1 -piperazinyl]-2-(methyloxy)aniline
Figure imgf000096_0002
Step A/Intermediate A70: 5-bromo-4-fluoro-2-nitrophenyl methyl ether
Figure imgf000096_0003
1-Bromo-2,5-difluoro-4-nitrobenzene (15.0 g, 63.0 mmol) was added to a solution of sodium methoxide in methanol (164 ml_, 0.5 M, Aldrich). The reaction was heated to 60 0C for 1 h. After cooling to room temperature, the solution was concentrated, and the residue was diluted with water (100 ml.) followed by extraction with ethyl acetate (2 x 80 ml_). The organic phase was dried (Na2SO4) and concentrated to afford 5-bromo-4- fluoro-2-nitrophenyl methyl ether as an orange solid (15.2 g, 95% yield). 1H NMR (400 MHz, DMSOd6) δ ppm 3.93 (s, 3 H), 7.76 (d, J=5.5 Hz, 1 H), 8.08 (d, J=8.4 Hz, 1 H).
Step B/lntermediate A71 : 1-[2-fluoro-5-(methyloxy)-4-nitrophenyl]-4-(1- methylethyl)piperazine
Figure imgf000097_0001
To 5-bromo-4-fluoro-2-nitrophenyl methyl ether (4.0 g, 16.0 mmol) in dioxane (150 ml.) was added 1-(1-methylethyl)piperazine (4.1 g, 32.0 mmol), XANTPHOS (0.9 g, 1.6 mmol), and CS2CO3 (10.4 g, 32.0 mmol). The mixture was bubbled with N2 for 15 min prior to the addition of Pd2(dba)3 (0.7 g, 0.8 mmol). The reaction was stirred at 100 0C for 18 h. Ethyl acetate (100 ml.) was used to dilute the reaction mixture, followed by the addition of water (80 ml_). After partitioning, extraction with ethyl acetate (2 x 75 ml_), drying (Na2SO4), filtration and concentration, silica gel chromatography afforded 1-[2- fluoro-5-(methyloxy)-4-nitrophenyl]-4-(1-methylethyl)piperazine as a yellow solid (3.3 g, 70% yield). 1 H NMR (400 MHz, DMSO-d6) δ ppm 0.98 (d, J=6.6 Hz, 6 H), 2.54 - 2.61 (m, 4 H), 2.68 (sept, J = 6.6 Hz, 1 H), 3.24 - 3.31 (m, 4 H), 3.91 (s, 3 H), 6.64 (d, J=7.7 Hz, 1 H), 7.82 (d, J=13.6 Hz, 1 H).
Step Cintermediate A69: 5-fluoro-4-[4-(1-methylethyl)-1-piperazinyl]-2- (methyloxy)aniline
Figure imgf000098_0001
To 1-[2-fluoro-5-(methyloxy)-4-nitrophenyl]-4-(1-methylethyl)piperazine (2.0 g, 6.7 mmol) in MeOH (100 ml.) was added iron (III) chloride (0.3 g, 2.0 mmol) and actived carbon (2.0 g). The reaction mixture was stirred at 64 0C for 20 min before the dropwise addition of hydrazine hydrate (4.0 ml_, 80.7 mmol) over 5 min. The reaction was kept stirring at 64 0C for additional 4 h. Filtration, concentration of the residue, and purification via column chromatography on SiO2 (0-10% 2 M NH3 in MeOH/DCM) afforded 5-fluoro-4-[4-(1-methylethyl)-1-piperazinyl]-2-(methyloxy)aniline as a dark brown solid (1.7 g, 95% yield). 1 H NMR (400 MHz, DMSO-d6) δ ppm 0.96 (d, J=6.6 Hz, 6 H), 2.45 - 2.54 (m, 4 H), 2.62 (sept, J = 6.6 Hz, 1 H), 2.79 - 2.86 (m, 4 H), 3.69 (s, 3 H), 4.55 (s, 2 H), 6.38 (d, J=13.6 Hz, 1 H), 6.49 (d, J=8.2 Hz, 1 H).
Intermediate A72: 5-methyl-4-[4-(1 -methylethyl)-1 -piperazinyl]-2- (methyloxy)aniline
Step A/Intermediate A73: 1-(1-methylethyl)-4-[2-methyl-5-(methyloxy)-4- nitrophenyl]piperazine
Figure imgf000099_0001
To a solution of 1-fluoro-2-methyl-5-(methyloxy)-4-nitrobenzene (1.3g, 7.03 mmol) in dimethylsulfoxide was added potassium carbonate (1.9g, 14.0 mmol) and isopropylpiperazine (2.0 ml_, 14 mmol). The resulting suspension was warmed at 7O0C for 12 hours, poured into water, and extracted with diethyl ether. The ether layers were washed with aqueous saturated sodium chloride, dried over sodium sulfate, taken to a residue under reduced pressure, and purified by chromatography on SiO2 to afford 1-(1- methylethyl)-4-[2-methyl-5-(methyloxy)-4-nitrophenyl]piperazine (1.78g, 86% yield) as a yellow solid. 1 H NMR (400 MHz, CDCI3) δ ppm 1.1 1 (d, J=6.60 Hz, 6 H), 2.24 (s, 3 H), 2.72 (s, 4 H), 2.79 (app s, 1 H), 3.06 (s, 4 H), 3.93 (s, 3 H), 6.57 (s, 1 H), 7.81 (s, 1 H).
Step B/lntermediate A72: 5-methyl-4-[4-(1-methylethyl)-1-piperazinyl]-2- (methyloxy)aniline
Figure imgf000099_0002
To a solution of 1-(1-methylethyl)-4-[2-methyl-5-(methyloxy)-4-nitrophenyl]piperazine (1.78g, 6.08 mmol) in methanol (75 ml.) was added hydrazine (1.33 ml_, 7.0 mmol), iron (III) chloride (0.200 g, 1.22 mmol) and activated charcoal (2g). The resulting slurry was warmed to 6O0C and maintained overnight. The next morning the slurry was filtered and concentrated to a residue. The residue was partitioned between ethyl acetate and sat. NaCI(aq). The organic layer was washed twice with saturated brine, dried over sodium sulfate, and taken to a residue under reduced pressure to afford 5-methyl-4-[4-(1- methylethyl)-1-piperazinyl]-2-(methyloxy)aniline (1.5Og, 94% yield) of sufficient purity for use directly in the next step. 1H NMR (400 MHz, DMSO-c/6) δ ppm 0.98 (d, J=6.60 Hz, 6 H), 2.04 (s, 3 H), 2.52 (s, 4 H), 2.64 (sept, J = 6.6 Hz, 1 H), 2.68 - 2.72 (m, 4 H), 3.69 (s, 3 H), 4.28 (s, 2 H), 6.41 (s, 1 H), 6.55 (s, 1 H).
lntermedaite A74: 5-methyl-2-(methyloxy)-4-(1 -propyl-4-piperidinyl)aniline
Figure imgf000100_0001
Step A/Intermediate A75: 4-[2-methyl-5-(methyloxy)-4-nitrophenyl]pyridine
Figure imgf000100_0002
To a nitrogen purged solution of 1-chloro-2-methyl-5-(methyloxy)-4-nitrobenzene (8 g, 39.7 mmol, see Canadian Journal of Chemistry (1987), 65(6), 1233-40.) and 4- pyridinylboronic acid (6.34 g, 51.6 mmol) in 1 ,4 dioxane (250 mL) was added dichlorobis(triphenylphosphine)palladium(ll) (2.79 g, 3.97 mmol) and degassed 3.0M aqueous sodium carbonate (39.7 ml, 1 19 mmol). The resulting suspension was maintained at 90 0C overnight. The reaction was subsequently cooled to room temperature, filitered through celite, and partioned between saturated sodium bicarbonate and ethyl acetate. The organic layer was dried over sodium sulfate, filtered, and taken to a residue under reduced pressure. The resulting residue was purified by chromatograpy on SiO2 (0 to 30% EtOAc in CH2CI2) to afford 4-[2-methyl-5-(methyloxy)- 4-nitrophenyl]pyridine (6.2 g, 25.4 mmol, 64.0 % yield) as a pale yellow solid. 1H NMR (400 MHz, CDCIs-d) δ ppm 8.64 - 8.78 (m, 2 H), 7.79 (s, 1 H), 7.15 - 7.31 (m, 2 H), 6.90 (s, 1 H), 3.95 (s, 3 H), 2.22 (s, 3 H)
Step B/lntermediate A76: 4-[2-methyl-5-(methyloxy)-4-nitrophenyl]-1-propylpyridinium iodide
Figure imgf000101_0001
To a solution of 4-[2-methyl-5-(methyloxy)-4-nitrophenyl]pyridine (6.2 g, 25.4 mmol) in pinacolone (150 ml) at 100 0C was added 1-iodopropane (17.36 ml, 178 mmol) and the resulting solution was stirred rapdily for 24 hr., during which time a thick yellow precipitate forms. The reaction was cooled to O0C, filtered, and the solids washed with diethyl ether to give analytically pure 4-[2-methyl-5-(methyloxy)-4-nitrophenyl]-1- propylpyridinium iodide (8.74 g, 21.10 mmol, 83 % yield). 1H NMR (400 MHz, DMSO-c/6) δ ppm 9.21 (d, J=6.97 Hz, 2 H), 8.31 (d, J=6.60 Hz, 2 H), 7.95 (s, 1 H), 7.39 (s, 1 H), 4.62 (t, J=IA b Hz, 2 H), 3.94 (s, 3 H), 2.27 (s, 3 H), 1.85 - 2.09 (m, 2 H), 0.78 - 1.08 (m, 3 H)
Step C/lntermediate A77: 4-[2-methyl-5-(methyloxy)-4-nitrophenyl]-1-propyl-1 , 2,3,6- tetrahydropyridine
Figure imgf000101_0002
To a suspension of 4-[2-methyl-5-(methyloxy)-4-nitrophenyl]-1-propylpyridinium (8.74 g, 30.4 mmol) in methanol (100 ml) at 0 0C was added portionwise sodium borohydride (3.45 g, 91 mmol). The resulting solution was stirred for four hours and poured into ethyl acetate/saturated sodium bicarbonate. The organic layer was dried over sodium sulfate, filtered, taken to a residue under reduced pressure, and purified by column chromatography to afford 4-[2-methyl-5-(methyloxy)-4-nitrophenyl]-1-propyl-1 ,2,3,6- tetrahydropyridine (6.08 g, 20.94 mmol, 68.8 % yield) as a yellow oil. 1H NMR (400 MHz, DMSOd6) δ ppm 7.71 (s, 1 H), 7.00 (s, 1 H), 5.54 - 5.68 (m, 1 H), 3.87 (s, 3 H), 3.02 (q, J=2.81 Hz, 2 H), 2.59 (t, J=5.68 Hz, 2 H), 2.48 (m, 2 H), 2.27 - 2.38 (m, 2 H), 2.20 (s, 3 H), 1.40 - 1.59 (m, 2 H), 0.87 (t, J=7.52 Hz, 3 H).
Step D/lntermediate A74: 5-methyl-2-(methyloxy)-4-(1-propyl-4-piperidinyl)aniline
Figure imgf000102_0001
A suspension of 4-[2-methyl-5-(methyloxy)-4-nitrophenyl]-1-propyl-1 , 2,3,6- tetrahydropyridine (8.74 g, 30.1 mmol) and 10% palladium on carbon (3.20 g, 3.01 mmol) in ethanol (100 ml) was maintained under 60 psi of hydrogen in a Parr hydrogenator for 16 hours. Following purging with nitrogen, the mixture was filtered and solvents removed under reduced pressure to afford analytically pure [5-methyl-2- (methyloxy)-4-(1-propyl-4-piperidinyl)phenyl]amine (5.01 g, 19.09 mmol, 63.4 % yield) as a white solid. 1H NMR (400 MHz, DMSOd6) δ ppm 6.56 (s, 1 H), 6.35 (s, 1 H), 4.34 (s, 2 H), 3.67 (s, 3 H), 2.89 (d, J=11.35 Hz, 2 H), 2.39 - 2.44 (m, 1 H), 2.14 - 2.24 (m, 2 H), 2.04 (s, 3 H), 1.81 - 1.95 (m, 2 H), 1.49 - 1.64 (m, 4 H), 1.32 - 1.48 (m, 2 H), 0.82 (t, J=7.33 Hz, 3 H).
Intermediate A78: 5-chloro-4-[4-(1 -methylethyl)-1 -piperazinyl]-2-(methyloxy)aniline
Figure imgf000103_0001
Step A/Intermediate A79: 4-chloro-5-fluoro-2-nitrophenol
Figure imgf000103_0002
4-chloro-3-fluorophenol (2.38 g, 16.2 mmol) was dissolved in DCE (32 ml.) and tetrabutylammonium bromide (0.524 g, 1.62 mmol) was added. Aqueous 70% HNO3 (2.1 ml_, 32 mmol) was diluted with H2O (18.9 ml.) to make a 7% HNO3 solution. This solution was added to the reaction mixture which was then stirred at rt for 4h at which time the reaction was judged complete by TLC. The reaction was poured into H2O and extracted with DCM (3x). The combined organic layers were dried over MgSO4, filtered and concentrated in vacuo. The resulting residue was adsorbed onto silica gel and flash chromatographed to give 4-chloro-5-fluoro-2-nitrophenol (2.29 g, 74%). 1H NMR (400 MHz, DMSO-Cf6) δ ppm i 1.76 (br. s., 1 H), 8.18 (d, J=8.1 Hz, 1 H), 7.10 (d, J=10.6 Hz, 1 H).
Step B/lntermediate A80: 4-chloro-5-fluoro-2-nitrophenyl methyl ether
Figure imgf000103_0003
4-Chloro-5-fluoro-2-nitrophenol (2.29 g, 12.0 mmol) was dissolved in DMF (25 ml_). K2CO3 (2.5 g, 18 mmol) and iodomethane (0.893 ml_, 14.3 mmol) were added and the mixture was stirred at rt overnight. The mixture was then poured into H2O and extracted with EtOAc (2x) and then the combined organics were extracted with H2O (5x). The organic layer was dried with MgSO4, filtered, and concentrated in vacuo to give 4-chloro- 5-fluoro-2-nitrophenyl methyl ether without further purification (2.19 g, 89 %). 1H NMR (400 MHz, DMSOd6) δ ppm 8.22 (dd, J=7.8, 1.0 Hz, 1 H), 7.54 (d, J=11.4 Hz, 1 H), 3.90 (d, J=1.1 Hz, 3 H).
Step C/lntermediate A81 : 1-[2-chloro-5-(methyloxy)-4-nitrophenyl]-4-(1- methylethyl)piperazine
Figure imgf000104_0001
A suspension of 1-chloro-2-fluoro-4-(methyloxy)-5-nitrobenzene (0.650 g, 3.17 mmol), isopropylpiperazine (1.0 ml_, 6.34 mmol), and potassium carbonate (0.90Og, 6.50 mmol) in dimethylsulfoxide (10 ml.) was maintained at 7O0C for 24 hours. The solution was diluted with diethyl ether, washed with saturated sodium chloride (aq), and purified by silica gel chromatography to afford 1-[2-chloro-5-(methyloxy)-4-nitrophenyl]-4-(1- methylethyl)piperazine (1.0g, quant, yield). 1H NMR (400 MHz, CDCI3-C/) δ ppm 8.03 (s, 1 H), 6.57 (s, 1 H), 3.95 (s, 3 H), 3.14 - 3.32 (m, 4 H), 2.76 - 2.82 (m, 1 H), 2.66 - 2.76 (m, 4 H), 1.1 1 (d, J=6.23 Hz, 6 H)
Step D/ Intermediate A78: 5-chloro-4-[4-(1-methylethyl)-1-piperazinyl]-2- (methyloxy)aniline
Figure imgf000104_0002
A suspension of 1-[2-chloro-5-(methyloxy)-4-nitrophenyl]-4-(1-methylethyl)piperazine (1.0 g, 3.2 mmol),hydrazine hydrate (0.700 ml_, 22.4 mmol), iron (III) chloride (0.100 g, 0.64 mmol), and activated carbon (1g) in methanol (40 ml) was warmed at 6O0C for 16 hours. The solution was filtered through celite while still warm and concentrated to an oily residue. The residue was partitioned between ethyl acetate and saturated sodium chloride and the organic layer was washed three times with saturated NaCI (aq), dried over sodium sulfate, filtered, and taken to an oil under reduced pressure to afford analytically pure 5-chloro-4-[4-(1-methylethyl)-1-piperazinyl]-2-(methyloxy)aniline (0.550 g,61% yield) as a pale yellow oil. 1H NMR (400 MHz, DMSOd6) δ ppm 6.64 (s, 1 H), 6.63 (s, 1 H), 4.62 (s, 2 H), 3.74 (s, 3 H), 2.75 - 2.88 (m, 4 H), 2.59 - 2.70 (m, 1 H), 2.50 - 2.59 (m, 4 H), 0.98 (d, J=6.60 Hz, 6 H).
Intermediate A82: 2-(ethyloxy)-4-[4-(1 -methylethyl)-1 -piperazinyljaniline
Figure imgf000105_0001
Step A/ Intermediate A83: 1-[3-(ethyloxy)-4-nitrophenyl]-4-(1-methylethyl)piperazine
Figure imgf000105_0002
A solution of 2-(ethyloxy)-4-fluoro-1 -nitrobenzene (7.1 1 g, 38.4 mmol, Combi-Blocks Catalog), potassium carbonate (15.92 g, 1 15 mmol), and 1-(1-methylethyl)piperazine (8.24 ml, 57.6 mmol) in dimethyl sulfoxide (DMSO) (75 ml) was stirred at 8O0C for 16 hours. The resulting slurry was poured into diethyl ether/water and washed three times with saturated sodium chloride. The organic layer was dried over sodium sulfate, filtered, taken to a residue under reduced pressure, and purified by column chromatography to afford 1-[3-(ethyloxy)-4-nitrophenyl]-4-(1-methylethyl)piperazine (7.09 g, 24.17 mmol, 62.9 % yield) as a bright yellow solid. 1H NMR (400 MHz, DMSOd6) δ ppm 7.84 (d, J=9.53 Hz, 1 H), 6.55 (dd, J=9.53, 2.57 Hz, 1 H), 6.49 (d, J=2.20 Hz, 1 H), 4.16 (q, J=7.09 Hz, 2 H), 3.34 - 3.42 (m, 4 H), 2.58 - 2.74 (m, 1 H), 2.48 - 2.55 (m, 4 H), 1.33 (t, J=6.97 Hz, 3 H), 0.97 (d, J=6.60 Hz, 6 H).
Step B/ Intermediate A82: 2-(ethyloxy)-4-[4-(1-methylethyl)-1-piperazinyl]aniline
Figure imgf000106_0001
A suspension of 1-[3-(ethyloxy)-4-nitrophenyl]-4-(1-methylethyl)piperazine (7.09 g, 24.17 mmol), hydrazine hydrate (9.49 ml, 193 mmol), iron (III) chloride (0.587 g, 3.63 mmol), and activated carbon (8 g, 24.17 mmol) in methanol (500 ml) was warmed at 6O0C for 16 hours. The solution was filtered through celite while still warm and concentrated to an oily residue. The residue was partitioned between ethyl acetate and saturated sodium chloride and the organic layer was washed three times with saturated NaCI (aq), dried over sodium sulfate, filtered, and taken to an oil under reduced pressure to afford analytically pure {2-(ethyloxy)-4-[4-(1-methylethyl)-1-piperazinyl]phenyl}amine (5.007 g, 19.01 mmol, 79 % yield) as a pale yellow oil. 1H NMR (400 MHz, DMSO-c/6) ppm 6.49 (d, J=8.43 Hz, 1 H), 6.43 (d, J=2.57 Hz, 1 H), 6.25 (dd, J=8.43, 2.57 Hz, 1 H), 4.15 (s, 2 H), 3.94 (q, J=6.97 Hz, 2 H), 2.82 - 2.91 (m, 4 H), 2.57 - 2.68 (m, 1 H), 2.49 - 2.55 (m, 4 H), 1.29 (t, J=6.97 Hz, 3 H), 0.97 (d, J=6.60 Hz, 6 H).
Intermediate A84: 1 -[3-(dimethylamino)propyl]-5-(methyloxy)-2,3-dihydro-1 H- indol-6-amine
Figure imgf000106_0002
Step A/Intermediate A85: Λ/,Λ/-dimethyl-3-[5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indol-1- yl]-3-oxo-1 -propanamine
Figure imgf000107_0001
A suspension of 5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole (1.5Og, 7.73 mmol) and polymer supported diisopropylethylamine (4.1 g, 16 mmol) in dichloromethane (100 ml.) was cooled to O0C and 2-propenoyl chloride (0.750 ml_, 9.3 mmol) was added dropwise. The solution was warmed slowly to room temperature and all solids were observed to dissolve. The mixture was filtered through celite, the celite washed twice with dichloromethane, and the combined filtrates were concentrated under reduced pressure to give a crude residue which was dissolved directly in a 1.0M dimethyl amine solution in tetrahydrofuran (40 ml_). The solution was maintained at 6O0C for 12 hours, cooled, taken to a residue under reduced pressure, and purified by chromatography on SiO2 to afford Λ/,Λ/-dimethyl-3-[5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indol-1-yl]-3-oxo-1- propanamine (1.52g, 67% Yield). This protocol could be reproduced on similar scale to generate larger quantities of product which would be carried forward as blended batches. 1H NMR (400 MHz, CDCI3) δ ppm 8.68 (s, 1 H), 6.91 (s, 1 H), 4.15 (t, J=8.43 Hz, 2 H), 3.91 (s, 3 H), 3.26 (t, J=8.43 Hz, 2 H), 2.81 (t, J=7.33 Hz, 2 H), 2.66 (t, J=I A 5 Hz, 2 H), 2.27 - 2.41 (m, 6 H).
Step B/lntermediate A86: 1-[3-(dimethylamino)propanoyl]-5-(methyloxy)-2,3- dihydro-1H-indol-6-amine
Figure imgf000107_0002
To Λ/,Λ/-dimethyl-3-[5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indol-1-yl]-3-oxo-1- propanamine (4.8g, 51.6 mmol) in degassed tetrahydrofuran (50 ml.) was added activated palladium on carbone (10%, 3.Og) . The reaction mixture was maintained under 40 psi hydrogen gas for 16 hours, purged with nitrogen and filtered through celite. The filtrates were concentrated to afford 1-[3-(dimethylamino)propanoyl]-5-(methyloxy)- 2,3-dihydro-1 H-indol-6-amine (4.15g, 96% yield). 1H NMR (400 MHz, CDCI3) δ ppm 7.72 (s, 1 H), 6.63 (s, 1 H), 3.95 - 4.15 (m, 2 H), 3.81 (s, 3 H), 3.76 (s, 2 H), 3.10 (t, J=8.43 Hz, 2 H), 2.83 - 2.96 (m, 2 H), 2.70 (t, J=IAb Hz, 2 H), 2.41 (s, 6 H).
Step C/lntermediate A84: 1-[3-(dimethylamino)propyl]-5-(methyloxy)-2,3-dihydro-1 H- indol-6-amine
Figure imgf000108_0001
A solution of 1-[3-(dimethylamino)propanoyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6- amine (1.35 g, 5.13 mmol) in tetrahydrofuran (50 ml) was treated with lithium aluminum hydride (1.0M soln. in THF, 25.6 ml, 25.6 mmol) at O0C and allowed to warm to room temperature overnight. The solution was poured cautiously into a saturated ammonium chloride (aq)/ice mixture and extracted three times with ethyl acetate. The organic layer was dried over sodium sulfate, filtered, taken to a residue under reduced pressure, and purified by column chromatography to afford 1-[3-(dimethylamino)propyl]-5-(methyloxy)- 2,3-dihydro-1 H-indol-6-amine (0.410 g, 1.644 mmol, 32.1 % yield) as a dark brown oil. 1H NMR (400 MHz, CDCI3) δ ppm 6.65 (s, 1 H), 6.01 (s, 1 H), 3.76 (s, 3 H), 3.65 (s, 2 H), 3.22 (t, J=8.13 Hz, 2 H), 2.90 - 3.00 (m, 2 H), 2.77 - 2.88 (m, 2 H), 2.31 - 2.39 (m, 2 H), 2.24 (s, 6 H), 1.67 - 1.83 (m, 2 H).
Intermediate A87: 5-{[3-(dimethylamino)propyl]oxy}-2-fluoroaniline
Figure imgf000108_0002
Step A/ Intermediate A88: 4-fluoro-3-nitrophenol
Figure imgf000108_0003
To a solution of 1-(4-fluoro-3-nitrophenyl)ethanone (12 g, 65.6 mmol, 1 eq) in H2SO4 (160 mL) and AcOH (96 mL) was added MeCO3H (50 ml_, 285.7 mmol, 4.3 eq) dropwise at O0C. The mixture was stirred at room temperature overnight. The mixture was added to ice-water and extracted with EtOAc, the organic layer was washed with sat NaHCOs, dried over Na2SO4, and concentrated and purifed by column chromatography to afford 4- fluoro-3-nitrophenol. (4.0 g). 1H NMR (400 MHz, DMSO) δ ppm 7.43-7.51 (m, 1 H), 8.17- 8.27 (m, 1 H), 8.47-8.55 (m, 1 H). Exchangeable protons are absent likely due to rapid exchange.
Step B/ Intermediate A89: 3-chloropropyl 4-fluoro-3-nitrophenyl ether
Figure imgf000109_0001
To a solution of 4-fluoro-3-nitrophenol (4 g, 25.4 mmol, 1 eq) in DMF (50 mL) was added NaH (60%, 1.5 g, 37 mmol, 1.4 eq) at O0C, the mixture was stirred for 30 min. 1-Bromo- 3-chloropropane (4.5 g, 29 mmol, 1.1 eq) was added to the mixture at 0 0C dropwise.
The mixture was stirred for 4h, poured into water, and extracted with EtOAc. The organic layer was washed with sat NaHCOs, dried over Na2SO4, concentrated and purified by column chromatography to afford 3-chloropropyl 4-fluoro-3-nitrophenyl ether. (3.6 g, 62% yield)(2 batches). 1H NMR (400 MHz, CDCI3) δ ppm 2.17-2.25 (m, 2H), 3.65-3.70 (m, 2H), 4.07-4.13 (m, 2H), 7.05-7.20 (m, 2H), 7.47-7.51 (m, 1 H)
Step C/ Intermediate A90: {5-[(3-chloropropyl)oxy]-2-fluorophenyl}amine
Figure imgf000109_0002
To a solution of 3-chloropropyl 4-fluoro-3-nitrophenyl ether (2 g, 8.5 mmol) in ethanol (40 mL) was added Pd/C (0.6 g) under N2 and the mixture was stirred under H2 for 2h at room temperature. The mixture was filtered and filtrates concentrated under reduced pressure to afford {5-[(3-chloropropyl)oxy]-2-fluorophenyl}amine (1.8 g, 100% yield — performed twice and batches blended). 1H NMR (400 MHz, MeOD) δ ppm 2.10-2.21 (m, 2H), 3.68-3.78 (m, 2H), 3.97-4.07 (m, 2H), 6.12-6.20 (m, 1 H), 6.37-6.47 (m, 1 H), 6.75- 6.85 (m, 1 H). Exchangeable protons are absent likely due to rapid exchange.
Step D/ Intermediate A87: 5-{[3-(dimethylamino)propyl]oxy}-2-fluoroaniline
Figure imgf000110_0001
To a solution of {5-[(3-chloropropyl)oxy]-2-fluorophenyl}amine (2.8 g, 13.8 mmol, 1 eq) in MeCN (25 ml.) was added K2CO3 (7.6 g, 55 mmol, 4 eq) and dimethylamine HCI salt (1.34 g, 13.8 mmol, 1 eq) and the mixture was heated to 8O0C overnight. The solvent was removed under reduced pressure. The crude product was poured into water and extracted with EtOAc. The organic layer was dried over Na2SO4, and concentrated (2 batches). To the solution of crude product (3.6 g) in MeOH (10 ml.) was added sat HCI/MeOH (20 ml.) at O0C, the mixture was stirred for 1 h. The solvent was removed under reduced pressure. The solid was washed with Et2O and filtered to give 5-{[3- (dimethylamino)propyl]oxy}-2-fluoroaniline. (3.85 g, 95%). 1H NMR (400 MHz, MeOD): δ ppm 2.05-2.15 (m, 2 H), 2.78 (s, 6 H), 3.19-3.23 (m, 2 H), 3.98-4.01 (m, 2 H), 6.82-6.85 (m, 2 H), 7.09-7.15 (m, 1 H)
Intermediate A91 : 1 ,1 -dimethylethyl (2S)-2-{[6-amino-5-(methyloxy)-2,3-dihydro- 1 H-indol-1 -yl]carbonyl}-1 -pyrrolidinecarboxylate
Figure imgf000110_0002
Step A/ Intermediate A92: 1 ,1 -dimethylethyl (2S)-2-{[5-(methyloxy)-6-nitro-2,3-dihydro- 1 H-indol-1 -yl]carbonyl}-1 -pyrrolidinecarboxylate
Figure imgf000110_0003
A solution of 5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole (3 g, 13.01 mmol), 1-{[(1 ,1- dimethylethyl)oxy]carbonyl}-L-proline (3.22 g, 14.96 mmol), HATU (6.43 g, 16.91 mmol), and DIPEA (6.82 ml, 39.0 mmol) in N,N-Dimethylformamide (80 ml) was stirred at room temperature overnight. The solution was poured into ethyl acetate and washed successively with 0.5% lithium chloride (aq) and saturated sodium chloride (aq). The organic layer was dried over sodium sulfate, filtered, taken to a residue under reduced pressure, and purified by chromatography on SiO2 to afford 1 ,1-dimethylethyl (2S)-2-{[5- (methyloxy^β-nitro^^-dihydro-I H-indol-i-yllcarbonylJ-i-pyrrolidinecarboxylate (4.75 g, 12.14 mmol, 93 % yield) as a pale yellow solid. Distinct Rotameric Forms Present at Room Temperature. 1H NMR (400 MHz, CDCI3) δ ppm 8.73 (s, 1 H), 8.72 (s, 1 H), 6.94 (s, 1 H), 6.90 (s, 1 H), 4.53 - 4.61 (m, 1 H), 4.46 - 4.53 (m, 1 H), 4.38 - 4.47 (m, 1 H), 4.20 - 4.32 (m, 1 H), 4.03 - 4.19 (m, 2 H), 3.92 (s, 3 H), 3.91 (s, 3 H), 3.59 - 3.74 (m, 2 H), 3.48 - 3.57 (m, 1 H), 3.38 - 3.49 (m, 1 H), 3.15 - 3.36 (m, 4 H), 2.06 - 2.33 (m, 4 H), 1.78 - 2.07 (m, 4 H), 1.45 (s, 9 H), 1.34 (s, 9 H).
Step B/ Intermediate A91 : 1 ,1-dimethylethyl (2S)-2-{[6-amino-5-(methyloxy)-2,3-dihydro- 1 H-indol-1-yl]carbonyl}-1-pyrrolidinecarboxylate
Figure imgf000111_0001
A suspension of 1 ,1-dimethylethyl (2S)-2-{[5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indol-1- yl]carbonyl}-1-pyrrolidinecarboxylate (4.75 g, 12.14 mmol) in methanol (100 miytetrahydrofuran (100 ml) was warmed to 550C until all solids had dissolved and then cooled to room temperature. Nitrogen was bubbled through the solution for 25 minutes, and then the 10% palladium on carbon (1.1 g, 12.14 mmol) was added (as a slurry in degassed methanol) and the suspension was maintained under 50 psi of H2 gas for 16 hours with rapid stirring. The solution was again purged with nitrogen, filtered through celite, and all volatiles removed to afford 1 ,1-dimethylethyl (2S)-2-{[6-amino-5- (methyloxy)-2,3-dihydro-1 /-/-indol-1-yl]carbonyl}-1-pyrrolidinecarboxylate (3.9 g, 10.79 mmol, 89 % yield) as a white foam. Distinct rotamers are present at room temperature by 1H NMR. 1H NMR (400 MHz) δ ppm 7.54 (s, 1 H), 7.51 (s, 1 H), 6.69 (s, 2 H), 4.62 (s, 4 H), 4.36 - 4.50 (m, 1 H), 4.08 - 4.17 (m, 1 H), 3.95 - 4.07 (m, 4 H), 3.69 (s, 6 H), 3.24 - 3.48 (m, 4 H), 2.86 - 3.12 (m, 4 H), 2.08 - 2.31 (m, 2 H), 1.66 - 2.01 (m, 6 H), 1.37 (s, 9 H), 1.22 (s, 9 H).
Intermediate A93: 5-methyl-2-(methyloxy)-4-{1 -[2-(methylsulfonyl)ethyl]-4- piperidinyljaniline
Figure imgf000112_0001
Step A/ Intermediate A94: 4-[2-methyl-5-(methyloxy)-4-nitrophenyl]pyridine
Figure imgf000112_0002
A solution of 5-bromo-4-methyl-2-nitrophenyl methyl ether (6.Og, 24.38 mmol),
PdCI2(dppf)*DCM adduct (1.43 g, 1.95 mmol) and 4-pyridylboronic acid (20.4 g, 165.8 mmol) in dioxane was deoxygenated by bubbling with N2(g) for ca 15 min. To this solution was added degassed 2.0 N Na2CO3(aq) (170 ml_, 14.0 equiv.) and the resulting slurry was stirred at 8O0C for 24 h. The dioxane was removed under reduced pressure and the solids were dissolved in EtOAc and washed with H2O, dried (Na2SO4), and purified by flash chromatography to give 4-[2-methyl-5-(methyloxy)-4- nitrophenyl]pyridine (2.81 g, 11.5 mmol, 57%). 1H NMR (400 MHz, CDCI3) δ ppm 8.72 (d, J=4.77 Hz, 2 H), 7.79 (s, 1 H), 7.22 - 7.31 (m, 2 H), 6.90 (s, 1 H), 3.95 (s, 3 H), 2.22 (s, 3 H).
Step B/ Intermediate A95: 5-methyl-2-(methyloxy)-4-(4-pyridinyl)aniline
Figure imgf000112_0003
4-[2-methyl-5-(methyloxy)-4-nitrophenyl]pyridine (2.44 g, 9.99 mmol) was placed in a 250 ml. high pressure vessel and dissolved in 60 ml. of 1 to 1 EtOAc/MeOH. 5 wt% Platinum(sulfided)/carbon (1.17 g, 0.3 mmol) was added and the vessel quickly sealed by a rubber septum. The flask was evacuated and filled with N2 six times to remove any oxygen. The vessel was then pressurized with H2 (60 psi). The solution was stirred for 72 h. The vessel was evacuated and filled with N2 six times to remove any H2. The solution was filtered through celite and evaporated to give 5-methyl-2-(methyloxy)-4-(4- pyridinyl)aniline (2.28 g, 10.6 mmol, quantitative yield). 1H NMR (400 MHz, DMSO-c/6) ppm 8.50 - 8.53 (m, 2 H), 7.30 - 7.33 (m, 2 H), 6.68 (s, 1 H), 6.54 (s, 1 H), 4.88 (bs, 2 H), 3.75 (s, 3 H), 2.12 (s, 3 H).
Step C/ Intermediate A96: 2,2,2-trifluoro-N-[5-methyl-2-(methyloxy)-4-(4-pyridinyl) phenyl]acetamide
Figure imgf000113_0001
To 5-methyl-2-(methyloxy)-4-(4-pyridinyl)aniline (2.28 g, 10.6 mmol) in THF was added triethylamine (3.23 g, 32 mmol), and trifluoroacetic anhydride (4.5 g, 21.3 mmol). The mixture was heated to 40 0C overnight. The solvent was concentrated and the residue purified by chromatography on SiO2 to give 2,2,2-trifluoro-N-[5-methyl-2-(methyloxy)-4- (4-pyridinyl) phenyl]acetamide (2.37 g, 7.64 mmol, 71%). 1H NMR (400 MHz, DMSOd6) δ ppm 10.76 (s, 1 H), 8.62 - 8.65 (m, 2 H), 7.42 - 7.44 (m, 2 H), 7.35 (s, 1 H), 6.98 (s, 1 H), 3.81 (s, 3 H), 2.16 (s, 3 H).
Step D/ Intermediate A97: 2,2,2-trifluoro-Λ/-[5-methyl-2-(methyloxy)-4-(4- piperidinyl)phenyl]acetamide acetate
Figure imgf000113_0002
2,2,2-Trifluoro-N-[5-methyl-2-(methyloxy)-4-(4-pyridinyl) phenyl]acetamide (2.37 g, 7.64 mmol) was placed in a 250 ml. high pressure vessel and dissolved in 70 ml. of HOAc. Platinum oxide (0.173 g, 0.76 mmol) was added and the vessel sealed quickly by a rubber septum. The flask was evacuated and filled with N2 six times to remove any oxygen. The vessel was then pressurized with H2 (60 psi). The solution was stirred for 24 h. The vessel was evacuated and filled with N2 six times to remove any H2. The solution was filtered through celite and evaporated to give 2,2,2-trifluoro-Λ/-[5-methyl-2- (methyloxy)-4-(4-piperidinyl)phenyl]acetamide acetate (2.83 g, 7.46 mmol, 98%). 1H NMR (400 MHz, DMSOd6) δ ppm 7.1 1 (s, 1 H), 6.86 (s, 1 H), 3.76 (s, 3 H), 3.14 (d, J=12.27 Hz, 2 H), 2.82 - 2.91 (m, 1 H), 2.72 - 2.81 (m, 2 H), 2.20 (s, 3 H), 1.81 (s, 3 H), 1.64 - 1.75 (m, 4 H).
Step E Intermediate A93: 5-methyl-2-(methyloxy)-4-{1-[2-(methylsulfonyl)ethyl]-4- piperidinyl}aniline
Figure imgf000114_0001
To 2,2,2-trifluoro-Λ/-[5-methyl-2-(methyloxy)-4-(4-piperidinyl)phenyl]acetamide acetate (1.5 g, 4.0 mmol) in 30 ml. of dioxane was added TEA (2 g, 20 mmol) and methyl vinyl sulfone (1.06 g, 10 mmol). The mixture was heated to 90 0C for 72 h. The solvent was concentrated and the crude product was purified by flash chromatography to give 480 mg of 2,2,2-trifluoro-Λ/-(5-methyl-2-(methyloxy)-4-{1 -[2-(methylsulfonyl)ethyl]-4- piperidinyl}phenyl)acetamide and 5-methyl-2-(methyloxy)-4-{1 -[2-(methylsulfonyl)ethyl]- 4-piperidinyl}aniline (0.40 g, 1.2 mmol, 31%). 1H NMR (400 MHz, CDCI3) δ ppm 6.62 (s, 1 H), 6.51 (s, 1 H), 5.22 (s, 2 H), 3.80 (bs, 3 H), 3.29 (t, J=6.87 Hz, 2 H), 3.13 (d, J=11.17 Hz, 2 H), 3.04 (s, 3 H), 2.98 (t, J=6.96 Hz, 2 H), 2.61 - 2.70 (m, 1 H), 2.24 - 2.31 (m, 2 H), 2.17 (s, 3 H), 1.73 - 1.81 (m, 4 H).
Intermediate A98: 5-{[3-(dimethylamino)propyl]oxy}-2-(methyloxy)aniline
Figure imgf000114_0002
In 2-butanone (200 ml.) were added 4-(methyloxy)-3-nitrophenol (6.0 g, 26.7 mmol), 3- dimethylaminopropylchlordiehydrochloride (12.6 g, 80.0 mmol), potassium carbonate (16.6 g, 120.2 mmol), and TBAI (0.5 g). The reaction was kept stirring at 80 0C for 48 h. After concentration the residue was partitioned between ethyl acetate and water and the aqueous layer was washed three times with ethyl acetate. The combined organic layers were dried (Na2SO4), filitered, and concentrated. Silica gel column chromatography afforded the purified dimethyl(3-{[4-(methyloxy)-3-nitrophenyl]oxy}propyl)amine, and the product was dissolved in ethanol (100 ml.) with the addition of 10% palladium on carbon (0.5 g). The reaction was kept stirring under H2 at 60 psi over the weekend. After releasing the H2 pressure, filtration removed the catalyst and the filtrate was concentrated. The crude product was purified by silica gel column chromatography to afford 5-{[3-(dimethylamino)propyl]oxy}-2-(methyloxy)aniline (5.3 g, 90% yield). 1H NMR (400 MHz, DMSOd6) δ ppm 1.71 - 1.81 (m, 2 H), 2.11 (s, 6 H), 2.29 (t, J=IA Hz, 2 H), 3.66 (s, 3 H), 3.81 (t, J=6.4 Hz, 2 H), 4.63 - 4.73 (m, 2 H), 6.02 (dd, J=QA, 2.9 Hz, 1 H), 6.22 (d, J=2.9 Hz, 1 H), 6.62 (d, J=8.8 Hz, 1 H).
Intermediate A99: 1 -[(dimethylamino)acetyl]-3,3-dimethyl-5-(methyloxy)-2,3- dihydro-1H-indol-6-amine
Figure imgf000115_0001
Step A/Intermediate A100: 1-acetyl-3,3-dimethyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/- indole
Figure imgf000115_0002
A solution of 1-acetyl-3,3-dimethyl-5-(methyloxy)-2,3-dihydro-1 H-indole (2.44 g, 11.13 mmol, see PCT Int. Appl. (2001 ), WO 2001023374 A1 20010405) in trifluoroacetic acid (50 ml.) was stirred at O0C and potassium nitrate (1.181 g, 1 1.68 mmol) was added in one portion. The reaction was maintained at O0C for 3 hours and poured into water. The solids were collected via filtration to afford 1-acetyl-3,3-dimethyl-5-(methyloxy)-6- nitro-2,3-dihydro-1 H-indole (2.545 g, 87 % yield) as a yellow solid. 1H NMR (400 MHz, DMSOd6) δ ppm 8.41 (s, 1 H), 7.34 (s, 1 H), 3.91 (s, 3 H), 2.15 (s, 2 H), 1.36 (s, 6 H). Note: proton resonance corresponding to acetyl was extremely broad and/or not evident for a variety of lots.
Step B/lntermediate A101 : 3,3-dimethyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indole
Figure imgf000116_0001
A solution of 1-acetyl-3,3-dimethyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indole (2.55 g, 9.65 mmol) in methanol (75 ml.) was treated with 4.0N HCI in dioxane (19.30 ml_, 77 mmol) and maintained at 7O0C for 16 hours. The solution was concentrated under reduced pressure, solids were triturated with diethyl ether and the slurry was filtered to afford 3,3-dimethyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indole (2.340 g, 94 % yield) as an orange solid. 1H NMR (400 MHz, DMSOd6) δ ppm 7.68 (s, 1 H), 7.36 (s, 1 H), 3.89 (s, 3 H), 3.41 (s, 2 H), 1.33 (s, 6 H).
Step C/lntermediate A102: {2-[3,3-dimethyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indol- 1-yl]-2-oxoethyl}dimethylamine
Figure imgf000116_0002
A solution of 3,3-dimethyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indole (2.38 g, 10.71 mmol), Hunig's base (9.33 ml_, 53.5 mmol) and bromo-acetylchloride (1.159 ml_, 13.92 mmol) in tetrahydrofuran (100 ml.) was maintained at O0C for 2 hours. Dimethylamine (2.0M in THF, 42.8 ml_, 86 mmol) was added to the solution and the reaction was warmed to room temperature and maintained for 5 hours. The reaction was poured into saturated sodium bicarbonate, the organic layer was dried over sodium sulfate, filtered, and purified by column chromatography to afford {2-[3,3-dimethyl-5-(methyloxy)-6-nitro- 2,3-dihydro-1 H-indol-1-yl]-2-oxoethyl}dimethylamine (2.14 g, 65.0 % yield) as an orange solid. 1H NMR (400 MHz, DMSOd6) δ ppm 8.44 (s, 1 H), 7.35 (s, 1 H), 3.97 (s, 2 H), 3.92 (s, 3 H), 3.21 (s, 2 H), 2.26 (s, 6 H), 1.34 (s, 6 H).
StepD/lntermediate A99: 1-[(dimethylamino)acetyl]-3,3-dimethyl-5-(methyloxy)-2,3- dihydro-1 H-indol-6-amine
Figure imgf000117_0001
A suspension of {2-[3,3-dimethyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indol-1 -yl]-2- oxoethyl}dimethylamine (2.20 g, 7.16 mmol), hydrazine hydrate (2.81 ml_, 57.3 mmol), iron(lll)chloride (0.232 g, 1.432 mmol), and activated carbon (2 g, 7.16 mmol) was warmed at 650C for 16 hours. The solution was filtered while still warm through celite, taken to a residue under reduced pressure, and partitioned between chloroform and saturated sodium bicarbonate. The organic layer was washed with saturated sodium chloride (aq), dried over sodium sulfate, filtered, taken to a residue under reduced pressure, and purified by column chromatography to afford 1-[(dimethylamino)acetyl]- 3,3-dimethyl-5-(methyloxy)-2,3-dihydro-1 H-indol-6-amine (1.41 g, 71 % yield) as a light yellow solid. 1H NMR (400 MHz, DMSOd6) δ ppm 7.49 (s, 1 H), 6.69 (s, 1 H), 4.62 (s, 2 H), 3.82 (s, 2 H), 3.73 (s, 3 H), 3.12 (s, 2 H), 2.23 (s, 6 H), 1.22 (s, 6 H).
Intermediate A103: 1 -[(dimethylamino)acetyl]-6-(ethyloxy)-1 ,2,3,4-tetrahydro-7- quinolinamine
Figure imgf000117_0002
Step A/Intermediate A104: 6-(ethyloxy)-3,4-dihydro-2(1H)-quinolinone
Figure imgf000118_0001
To 6-hydroxy-3,4-dihydro-2(1 /-/)-quinolinone ( 90.2 g, 0.55 mol, 1 equiv) in DMF (1.8L) was added potassium carbonate (228 g, 1.65 mol, 3 equiv). The slurry was stirred for 45 min. at 250C, then iodoethane (114 g, 0.73 mol, 1.33 equiv) was added and stirring was continued for 12 hours. The mixture was filtered, the filtrate was poured into water, and the white precipitate was collected via filtration, washed with water (50OmL), and dried under high vacuum to provide 6-(ethyloxy)-3,4-dihydro-2(1 H)-quinolinone (72.3g, 69%). 1H NMR (400 MHz, DMSO) δ, 1.25 (t, J=7.2 Hz, 3H), 2.35 (m, 2 H), 2.78 (m, 2H), 3.90 (m, 2H), 6.70-6.73 (m, 3H), 9.83 (br, 1 H).
StepB/lntermediate A105: 6-(ethyloxy)-7-nitro-3,4-dihydro-2(1H)-quinolinone
Figure imgf000118_0002
To the solution of 6-ethoxy-3,4-dihydroquinolin-2(1 H)-one (72.3 g, 0.38 mol, 1 equiv) in TFA (800 mL) was added NaNO2 (33.7 g, 0.496 mol,1.3 equiv) at O0C. The solution was allowed to warm to room temperature and stirring was continued for 4 hours. The mixture was poured into ice and the yellow precipitate was isolated via filtration and dried under high vacuum at 50 0C. The crude product was recrystallized from ethyl acetate to afford 6-(ethyloxy)-7-nitro-3,4-dihydro-2(1 H)-quinolinone (69.2 g, 77% yield). 1H NMR (400 MHz, DMSO) δ 1.31 (t, J=6.8 Hz, 3H), 2.45-2.49 (m, 2 H), 2.95 (t, 2 H, J=6.4 Hz), 4.13-4.15 (m, 2 H), 7.25 (s, 1 H), 7.34 (s, 1 H), 10.16 (bs, 1 H).
Step C/lntermediate A106: 6-(ethyloxy)-7-nitro-1 ,2,3,4-tetrahydroquinoline
Figure imgf000119_0001
To a stirred solution of 6-ethoxy-3,4-dihydroquinolin-2(1 /-/)-one (69 g of, 0.29 mol, 1 equiv) in THF (1 L) was added BH3-DMS (10 M, 120 ml_, 1.2 mol, 4equiv) dropwise at 25 0C. The mixture was stirred for 4 h at 60 0C. The reaction was cooled and quenched carefully with excess MeOH, then the mixture was concentrated under reduced pressure and the resultant solid suspended in a mixture of Et2O and EtOAc and filtered to afford 6-(ethyloxy)-7-nitro-1 ,2,3,4-tetrahydroquinoline (26.7 g, 42% yield) as an orange solid. 1H NMR (400 MHz, DMSO) δ 1.24 (t, J = 6.8 Hz, 3 H), 1.73-1.77 (m, 2 H), 2.68 (m, 2 H), 3.95-4.01 (m, 2 H), 5.81 (s, 1 H), 6.85 (s, 1 H), 6.89 (s, 1 H). Several protons obstructed by solvent.
Step D/lntermediate A107: {2-[6-(ethyloxy)-7-nitro-3,4-dihydro-1 (2/-/)-quinolinyl]-2- oxoethyl}dimethylamine
Figure imgf000119_0002
A suspension of 6-(ethyloxy)-7-nitro-1 ,2,3,4-tetrahydroquinoline (5.0 g, 22.50 mmol) and potassium carbonate (6.22 g, 45.0 mmol) in dichloromethane (100 ml.) was treated with bromoacetylchloride (2.336 ml_, 28.1 mmol) and stirred for 25 minutes. Water was added and the organic layer was dried over sodium sulfate, filtered, taken to a residue under reduced pressure, redissolved in THF, and 2.0M dimethyl amine in THF (67.5 ml_, 135 mmol) was added. The solution was stirred overnight, diluted with saturated sodium bicarbonate, and the organic layer was dried over sodium sulfate, filtered, taken to a residue under reduced pressure, and purified by column chromatography to afford {2-[6- (ethyloxy)-7-nitro-3,4-dihydro-1 (2H)-quinolinyl]-2-oxoethyl}dimethylamine (5.8 g, 84 % yield) as a pale yellow oil. 1H NMR (400 MHz, DMSO-d6) δ ppm 8.34 (s, 1 H), 7.16 (s, 1 H), 4.17 (q, J=6.96 Hz, 2 H), 3.74 (t, J=5.62 Hz, 2 H), 3.21 (s, 2 H), 2.80 (t, J=6.32 Hz, 2 H), 2.21 (s, 6 H), 1.76 - 2.01 (m, 2 H), 1.33 (t, J=6.92 Hz, 3 H). Step E/lntermediate A103: 1-[(dimethylamino)acetyl]-6-(ethyloxy)-1 ,2,3,4-tetrahydro-7- quinolinamine
Figure imgf000120_0001
A solution of {2-[6-(ethyloxy)-7-nitro-3,4-dihydro-1 (2H)-quinolinyl]-2- oxoethyl}dimethylamine (5.8 g, 18.87 mmol) in degassed methanol (50.0 ml.) was added to a suspension of 10% Pd/C (3.0 g, 16.36 mmol) in degassed methanol (5 ml.) and maintained under a 50 psi hydrogen gas with rapid stirring for 16 hours. The suspension was purged with nitrogen, filtered through celite, all volatiles removed, and the resulting residue was purified by flash column chromatography to afford 1-
[(dimethylamino)acetyl]-6-(ethyloxy)-1 ,2,3,4-tetrahydro-7-quinolinamine (2.85 g, 63 %) as a viscous yellow liquid which solidified upon standing for 10 hours. 1H NMR (400 MHz, DMSOd6) δ ppm 6.80 (s, 1 H), 6.55 (s, 1 H), 4.52 (s, 2 H), 3.95 (q, J=6.89 Hz, 2 H), 3.61 (t, J=6.02 Hz, 2 H), 3.15 (s, 2 H), 2.55 (t, J=6.52 Hz, 2 H), 2.20 (s, 6 H), 1.73 - 1.86 (m, 2 H), 1.32 (t, J=7.02 Hz, 3 H).
Intermediate A108: 5-(methyloxy)-1 -(1 -methyl-L-prolyl)-2,3-dihydro-1 H-indol-6-
Figure imgf000120_0002
Step A/Intermediate A109: 5-(methyloxy)-1-(1-methyl-L-prolyl)-6-nitro-2,3-dihydro-1 H- indole
Figure imgf000120_0003
A solution of 5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indole (2.7 g, 11.71 mmol), 1-methyl- L-proline (Acros, 1.739 g, 13.46 mmol), HATU (5.79 g, 15.22 mmol), and DIPEA (6.13 ml_, 35.1 mmol) in N,N-dimethylformamide (25 ml.) was stirred at room temperature overnight. The next morning the solution had completely solidified. The solids were diluted with ethyl acetate/THF and washed with saturated sodium bicarbonate until all solids had dissolved. The organic layer was dried over sodium sulfate, filtered, concentrated onto celite, and purified by column chromatography to afford 5-
(methyloxy)-1-(1-methyl-L-prolyl)-6-nitro-2,3-dihydro-1 /-/-indole (3.4 g, 95 % yield) as a yellow solid. 1H NMR (400 MHz, DMSOd6) δ ppm 8.51 (s, 1 H), 7.35 (s, 1 H), 4.20 - 4.31 (m, 1 H), 4.10 - 4.20 (m, 1 H), 3.88 (s, 3 H), 3.56 - 3.70 (m, 1 H), 3.25 (t, J=8.43 Hz, 2 H), 3.15 - 3.21 (m, 1 H), 2.52 - 2.64 (m, 1 H), 2.45 (s, 3 H), 2.21 - 2.35 (m, 1 H), 1.75 - 1.93 (m, 3 H).
Step B/lntermediate A108: 5-(methyloxy)-1-(1-methyl-L-prolyl)-2,3-dihydro-1 /-/-indol-6-
Figure imgf000121_0001
A suspension of 5-(methyloxy)-1-(1-methyl-L-prolyl)-6-nitro-2,3-dihydro-1 H-indole (2.50 g, 8.19 mmol) in methanol (100 ml_)/tetrahydrofuran (100 mL) was warmed to 650C and maintained until all solids had dissolved. The solution was cooled, purged with nitrogen for 40 minutes, and 10% palladium on carbon (1.1 g, 8.19 mmol) was added and the suspension was maintained under 40 psi of H2 gas with rapid stirring for 24 hours. The solution was carefully purged with nitrogen, filtered through a pad of celite, and all solvents removed under reduced pressure to afford 5-(methyloxy)-1-(1-methyl-L-prolyl)- 2,3-dihydro-1 H-indol-6-amine (2.23 g, 99% yield). 1H NMR (400 MHz, DMSOd6) δ ppm 7.57 (s, 1 H), 6.70 (s, 1 H), 4.76 (s, 2 H), 4.06 - 4.17 (m, 1 H), 3.93 - 4.06 (m, 1 H), 3.70 (s, 3 H), 3.46 (s, 1 H), 3.06 - 3.18 (m, 1 H), 2.98 (t, J=8.25 Hz, 2 H), 2.41 - 2.47 (m, 1 H), 2.37 (s, 3 H), 2.12 - 2.29 (m, 1 H), 1.66 - 1.92 (m, 3 H).
Intermediate A110: 5-(methyloxy)-1 -(1 -methyl-D-prolyl)-2,3-dihydro-1H-indol-6- amine
Figure imgf000122_0001
Step A/Intermediate A1 11 : 1-methyl-D-proline
Figure imgf000122_0002
To a solution of D-proline (30 g, 260.7mmol, 1eq) and 37% aqueous formaldehyde (80.5ml_, 782mmol, 3eq) in H2O (900 ml.) was added 10% Pd/C (30 g) under N2. The mixture was stirred at 5O0C under H2 (30psi) overnight. The mixture was heated to boiling and filtered to remove the catalyst. The solution was concentrated under reduced pressure, and then water (150 ml.) was added to the mixture and concentrated, the procedure was repeated three times so as to remove unreacted formaldehyde. The crude product was resuspended in MeCN/H2O (1 :1 , v/v) and lyophilized. The solid product was recrystallized from EtOH/acetone, filtered, liberally washed with acetone, and dried under reduced pressure to give 1-methyl-D-proline (12.3g, 40%) as white solid. 1H NMR (400 MHz, MeOD) δ ppm 1.95-1.99 (s, 1 H), 2.1-2.2 (s, 2H), 2.4-2.5 (m , 1 H), 3.05-3.15 (m, 1 H), 3.3-3.35 (s, 1 H),3.65-3.75 (m, 1 H), 3.75-3.85 (m, 1 H). Several protons obstructed by residual solvent.
Step B/lntermediate A1 12: 5-(methyloxy)-1-(1-methyl-D-prolyl)-6-nitro-2,3-dihydro-1 /-/- indole
Figure imgf000122_0003
A solution of 5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indole (4.35 g, 18.86 mmol), 1-methyl- D-proline (2.436 g, 18.86 mmol), HATU (9.32 g, 24.52 mmol), and DIPEA (9.88 ml_, 56.6 mmol) in N,N-dimethylformamide (25 ml.) was stirred at room temperature overnight. The solution was poured into ethyl acetate/saturated sodium bicarbonate and the organic layer was washed three times with saturated sodium chloride (aq). The organic layer was dried over sodium sulfate, filtered, taken to a residue under reduced pressure, and purified by column chromatography to afford 5-(methyloxy)-1-(1-methyl-D-prolyl)-6- nitro-2,3-dihydro-1 H-indole (3.62 g, 11.86 mmol, 62.9 % yield) as a pale brown oil. 1H NMR (400 MHz, DMSOd6) δ ppm 8.52 (s, 1 H), 7.37 (s, 1 H), 4.21 - 4.34 (m, 1 H), 4.05 - 4.21 (m, 1 H), 3.90 (s, 3 H), 3.78 - 3.88 (m, 1 H), 3.22 - 3.31 (m, 2 H), 3.16 (d, J=3.01 Hz, 1 H), 2.64 - 2.81 (m, 1 H), 2.54 (s, 3 H), 2.30 - 2.44 (m, 1 H), 1.67 - 2.09 (m, 3 H).
Step C/lntermediate A110: 5-(methyloxy)-1-(1-methyl-D-prolyl)-2,3-dihydro-1H-indol-6- amine
Figure imgf000123_0001
A suspension of 5-(methyloxy)-1-(1-methyl-D-prolyl)-6-nitro-2,3-dihydro-1 H-indole (3.62 g, 11.86 mmol) in tetrahydrofuran (50.0 ml_)/methanol (50 ml.) was warmed to 6O0C and maintained until all solids dissolved. The solution was cooled, degassed by bubbling with N2 gas for 20 minutes, added to a pressure flask containing 10% palladium on carbon (1.1 g, 8.19 mmol), and maintained under 60 psi H2 gas for 24 hours. The solution was purged with nitrogen, filtered through celite, the celite washed with methanol, and all volatiles removed under reduced pressure to afford 5-(methyloxy)-1- (1-methyl-D-prolyl)-2,3-dihydro-1 H-indol-6-amine (2.35 g, quantitative yield) as a pale yellow solid. 1H NMR (400 MHz, DMSOd6) δ ppm 7.57 (s, 1 H), 6.72 (s, 1 H), 4.69 (s, 2 H), 4.06 - 4.17 (m, 1 H), 3.93 - 4.06 (m, 1 H), 3.71 (s, 3 H), 3.64 (s, 1 H), 3.32 (s, 1 H), 3.21 (d, J=10.83 Hz, 1 H), 3.00 (t, J=8.03 Hz, 2 H), 2.49 (t, 3 H), 2.30 (d, J=8.23 Hz, 1 H), 1.65 - 1.96 (m, 3 H).
Intermediate A113: 1-[(dimethylamino)acetyl]-2,3-dihydro-1H-indol-6-amine
Figure imgf000124_0001
Step A/Intermediate A1 14: Λ/,Λ/-dimethyl-2-(6-nitro-2,3-dihydro-1 H-indol-1-yl)-2- oxoethanamine
Figure imgf000124_0002
A susupension of 6-nitro-2,3-dihydro-1 H-indole (10 g, 60.9 mmol) and potassium carbonate (16.84 g, 122 mmol) in dichloromethane (250 ml.) was treated with bromoacetylchloride (6.33 ml_, 76 mmol) and stirred for 25 minutes. Water was added and the organic layer was dried over sodium sulfate, filtered, taken to a residue under reduced pressure. The residue was redissolved in THF and 2.0M dimethyl amine in THF (Aldrich, 152 ml_, 305 mmol) was added. The solution was stirred overnight, diluted with saturated sodium bicarbonate, and the organic layer was dried over sodium sulfate, filtered, taken to a residue under reduced pressure, and purified by column chromatography to afford Λ/,Λ/-dimethyl-2-(6-nitro-2,3-dihydro-1 /-/-indol-1-yl)-2- oxoethanamine (13.6 g, 54.6 mmol, 90 % yield) as a pale yellow solid. 1H NMR (400 MHz, DMSOd6) δ ppm 8.80 (d, J=2.01 Hz, 1 H), 7.90 (dd, J=8.23, 2.21 Hz, 1 H), 7.48 (d, J=8.23 Hz, 1 H), 4.26 (t, J=8.53 Hz, 2 H), 3.18 - 3.31 (m, 4 H), 2.28 (s, 6 H).
Step B/lntermediate A1 13: 1-[(dimethylamino)acetyl]-2,3-dihydro-1 /-/-indol-6-amine
Figure imgf000124_0003
A suspension of Λ/,Λ/-dimethyl-2-(6-nitro-2,3-dihydro-1 /-/-indol-1-yl)-2-oxoethanamine (13.6 g, 54.6 mmol), hydrazine hydrate (21.42 ml_, 436 mmol), iron (III) chloride (1.768 g, 10.91 mmol), activated carbon (15 g), and methanol (100 ml.) was maintained at 650C for 12 hours, cooled, and filtered through celite (rinsed with additional methanol). Filtrates were concentrated, redissolved in ethyl acetate, and washed twice with saturated aqueous sodium chloride and sodium bicarbonate. The organic layer was dried over sodium sulfate, filtered, and concentrated to afford 1-[(dimethylamino)acetyl]- 2,3-dihydro-1 /-/-indol-6-amine (9.6 g, 80 % yield) as a white solid. 1H NMR (400 MHz, DMSOd6) δ ppm 7.44 (d, J=1.40 Hz, 1 H), 6.83 (d, J=8.03 Hz, 1 H), 6.20 (dd, J=7.93, 2.1 1 Hz, 1 H), 4.94 (s, 2 H), 4.06 (t, J=8.43 Hz, 2 H), 3.14 (s, 2 H), 2.91 (t, J=8.33 Hz, 2 H), 2.25 (s, 6 H).
Intermediate A115: 5<:hloro-1-[(dimethylamino)acetyl]-2,3-dihydro-1H-indol-6-
Figure imgf000125_0001
A solution of 1-[(dimethylamino)acetyl]-2,3-dihydro-1 H-indol-6-amine (3.0 g, 13.68 mmol) in acetonitrile (150 ml.) was treated with N-chlorosuccinimide (2.010 g, 15.05 mmol) and stirred at room temperature for 15 minutes. The solution was poured into chloroform/saturated sodium bicarbonate (aq.) and the organic layer was dried over sodium sulfate, filtered, taken to a residue under reduced pressure, and purified by column chromatography to afford 5-chloro-1-[(dimethylamino)acetyl]-2,3-dihydro-1 H- indol-6-amine (0.510 g, 15 % yield) as a pale yellow solid. 1H NMR (400 MHz, DMSO- d6) δ ppm 7.66 (s, 1 H), 7.01 (s, 1 H), 5.21 (s, 2 H), 4.08 (t, J=8.33 Hz, 2 H), 3.16 (s, 2 H), 2.95 (t, J=8.33 Hz, 2 H), 2.25 (s, 6 H).
Intermediate A116: 6-(methyloxy)-1-(1-methyl-L-prolyl)-1,2,3,4-tetrahydro-7- quinolinamine
Figure imgf000126_0001
A solution of 6-(methyloxy)-7-nitro-1 , 2,3, 4-tetrahydroquinoline (3.0 g, 14.41 mmol), 1- methyl-L-proline (2.140 g, 16.57 mmol), HATU (7.12 g, 18.73 mmol), and DIPEA (6.29 ml_, 36.0 mmol) in Λ/,Λ/-dimethylformamide (25 ml.) was stirred for 2.5 days at room temperature. Reaction was poured into ethyl acetate and washed successively with 5% lithium chloride (aq) and saturated sodium chloride (aq). The organic layer was dried over sodium sulfate, filtered, and purified by column chromatography (10% MeOH/CH2CI2) to afford 6-(methyloxy)-1-(1-methyl-L-prolyl)-7-nitro-1 , 2,3,4- tetrahydroquinoline (2.09 g, 6.54 mmol, 45.4 % yield). The residue was suspended in ethanol (300 ml.) and Λ/,Λ/-dimethylacetamide (50 ml.) was added followed by tin(ll)chloride dihydrate (6.78 g, 30.1 mmol) and 1.0M HCI (2.505 ml_, 2.505 mmol). After overnight stirring the reaction was quenched with excess saturated NaHCOs (300 ml.) and allowed to stir at rt for 30 min. The solids were removed through a celite pad, rinsed with MeOH, and evaporated. The aqueous layer was extracted with DCM (2x100 ml_). The combined organic layers were filtered through a cotton plug, and concentrated by rotary evaporation to provide 6-(methyloxy)-1-(1-methyl-L-prolyl)-1 ,2,3,4-tetrahydro-7- quinolinamine (1.1 g, 26% over two steps). ESIMS (M+H)+ = 290.
Intermediate A117: 1-[(2R)-2-(dimethylamino)propanoyl]-5-(methyloxy)-2,3- dihydro-1 H-indol-6-amine
Figure imgf000126_0002
Step A/Intermediate A1 18: N,N-dimethyl-D-alanine
H ..O.Λ> To a solution of D-alanine (30 g, 336mmol) and 37% aqueous formaldehyde (90.5ml_, 930mmol) in H2O (900 ml.) was added 10% Pd/C (30 g) under N2, the mixture was stirred at 5O0C under H2 (30psi) overnight. The mixture was heated to boiling and filtered to remove the catalyst. The solution was concentrated under reduced pressure, and then water (150 ml.) was added to the mixture and concentrated, the procedure was repeated 3 times in order to remove any unreacted formaldehyde. The crude product was resuspended in ACN/H2O (1 :1 , v/v) and lyophilized. The solid product was recrystallized from EtOH/acetone, filtered, liberally washed with acetone, and dried under reduced pressure to give N,N-dimethyl-D-alanine (30.4g, 77.07%) as white, very hygroscopic solid. 1H NMR (400 MHz, MeOD) δ ppm 1.41-1.49 (m, 3H), 2.8-2.85 (s, 6H), 3.6-3.65 (m, 1 H), 4.9-4.95 (s,1 H)
Step B/lntermediate A1 19: 1-[(2R)-Λ/,Λ/-dimethyl-1-[5-(methyloxy)-6-nitro-2,3-dihydro-1 H- indol-1 -yl]-1 -oxo-2-propanamine
Figure imgf000127_0001
To a solution of 5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indole hydrogen chloride (2 g, 8.67 mmol) in N,N-dimethylformamide (100 ml.) was added N,N-dimethyl-D-alanine (1.524 g, 13.01 mmol), PyBOP (13.54 g, 26.0 mmol), and DIPEA (15.14 ml_, 87 mmol). After stirring overnight at rt, the solvent was removed and the residue was suspended in dichloromethane (150 ml_), washed with water (100 ml_), concentrated on the rotovap, and purified by column chromatography (DCM to 5% MeOH/DCM) to provide 1-[(2R)- Λ/,Λ/-dimethyl-1-[5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indol-1-yl]-1 -oxo-2-propanamine (2.2 g, 86%). ESIMS (M+H)+ = 294.
Step C/lntermediate A117: 1-[(2R)-2-(dimethylamino)propanoyl]-5-(methyloxy)-2,3- dihydro-1 H-indol-6-amine
Figure imgf000128_0001
To a solution of (2R)-Λ/,Λ/-dimethyl-1-[5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indol-1-yl]-1- oxo-2-propanamine (2.2 g, 7.50 mmol) in ethyl acetate (250 ml.) was added Pd/C (0.798 g, 0.750 mmol) and the reaction maintained under 50 psi of H2(g) overnight in a Fisher- Porter apparatus. The crude material was filtered through a celite pad, washed with ethyl acetate, concentrated, and placed under high vaccuum overnight to afford 1-[(2R)- 2-(dimethylamino)propanoyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-amine (1.4 g, 71%). ESIMS (M+H)+ = 264.
Intermediate A120: 1-[(2S)-2-(dimethylamino)propanoyl]-5-(methyloxy)-2,3- dihydro-1 H-indol-6-amine
Figure imgf000128_0002
Step A/Intermediate A121 : N,N-dimethyl-L-alanine
Figure imgf000128_0003
To a solution of L-alanine (30 g, 360mmol) and 37% aqueous formaldehyde (90 ml_, 930 mmol) in H2O (900 ml.) was added 10% Pd/C (30 g) under N2, the mixture was stirred at 5O0C under H2 (30psi) overnight. The mixture was heated to boiling and filtered to remove the catalyst. The solution was concentrated under reduced pressure, and then water (150 ml.) was added to the mixture and concentrated, the procedure was repeated for 3 times in order to remove any unreacted formaldehyde. The crude product was resuspended in ACN/H2O (1 :1 , v/v) and lyophilized. The solid product was recrystallized from EtOH/acetone, filtered, liberally washed with acetone, and dried under reduced pressure to give Λ/,Λ/-dimethyl-/.-alanine (30.4g, 77.07%) as white, very hygroscopic solid. 1H NMR (400 MHz, MeOD) δ ppm 1.41-1.49 (d, J=7.2Hz,3H), 2.8-2.85 (s, 6H), 3.6-3.65 (q, J=7.2Hz ,1 H), 4.7-4.95 (br,1 H).
Step B/lntermediate A122: (2S)-N,N-dimethyl-1-[5-(methyloxy)-6-nitro-2,3-dihydro-1 H- indol-1 -yl]-1 -oxo-2-propanamine
Figure imgf000129_0001
To a solution of 5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indole hydrogen chloride (2 g, 8.67 mmol) in N,N-dimethylformamide (100 ml.) was added N,N-dimethyl-L-alanine (1.524 g, 13.01 mmol), PyBOP (13.54 g, 26.0 mmol), and DIPEA (15.14 ml_, 87 mmol). After stirring overnight at rt, the solvent was removed, the residue was suspended in dichloromethane (150 ml_), washed with water (100 ml_), concentrated, and purified by column chromatography (DCM to 5% MeOH/DCM) to give (2S)-Λ/,Λ/-dimethyl-1-[5- (methyloxy)-6-nitro-2,3-dihydro-1 H-indol-1-yl]-1-oxo-2-propanamine (2.2 g, 86%). ESIMS (M+H)+ = 294.
Step C/lntermediate A120: 1-[(2S)-2-(dimethylamino)propanoyl]-5-(methyloxy)-2,3- dihydro-1 H-indol-6-amine
Figure imgf000129_0002
To a solution of (2S)-N,N-dimethyl-1-[5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indol-1-yl]-1- oxo-2-propanamine (2.2 g, 7.50 mmol) in ethyl acetate (250 ml.) was added 10% Pd/C (0.798 g, 0.750 mmol) and the reaction was maintained under 50 psi of H2(g) overnight on the Fisher-Porter apparatus. The reaction was filtered through a celite pad which was washed with ethyl acetate, concentrated by rotary evaporation, and maintained under high vacuum for 12 hours to afford 1-[(2S)-2-(dimethylamino)propanoyl]-5- (methyloxy)-2,3-dihydro-1 H-indol-6-amine (1.5 g, 76%). ESIMS (M+H)+ = 264. Intermediate A123: 2-(methyloxy)-4-[4-(2-methylpropyl)-1 -piperazinyl]aniline
Figure imgf000130_0001
Step A/Intermediate A123: 1 ,1-dimethylethyl 4-[3-(methyloxy)-4-nitrophenyl]-1- piperazinecarboxylate
Figure imgf000130_0002
4-chloro-2-(methyloxy)-1 -nitrobenzene (1Og, 53.2 mmol, Aldrich), 1 ,1-dimethylethyl 1- piperazinecarboxylate (2Og, 107.5 mmol), cesium carbonate (35.Og, 107.5 mmol), Pd2dba3 (5g, 5.5 mmol), and XANTPHOS (4.62g, 8.0 mmol) were added to degassed dioxane (100 ml.) and heated to 1000C under a water cooled reflux condenser for 12 hours. The dioxane was removed under reduced pressure and the solids were partitioned between methylene chloride (500 ml.) and water (500 ml_). The organic layer was dried over sodium sulfate, taken to a residue under reduced pressure, and triturated with a methylene choride/hexanes mixture (15:85) to precipitate out analytically pure 1- dimethylethyl 4-[3-(methyloxy)-4-nitrophenyl]-1 -piperazinecarboxylate as a yellow solid (13.2g, 39.2 mmol, 73% yield). 1H NMR (400 MHz, CDCI3) δ ppm 1.49 (s, 9 H), 3.33 - 3.42 (m, 4 H), 3.62-3.64 (m, 4 H), 3.96 (s, 3 H), 6.39 (s, 1 H), 6.44 (dd, J=9.16, 2.56 Hz, 1 H), 8.01 (d, J=9.16 Hz, 1 H).
Step B/lntermediate A125: 1-[3-(methyloxy)-4-nitrophenyl]piperazine
Figure imgf000131_0001
1-dimethylethyl 4-[3-(methyloxy)-4-nitrophenyl]-1-piperazinecarboxylate (2.25g, 6.67 mmol) was dissolved in methylene chloride (50 mL) and trifluoroacetic acid (10 ml_). The resulting solution turned dark immediately and was stirred overnight. The solution was concentrated and partitioned between methylene chloride and 2.0N sodium hydroxide. The organic layer was collected and the aqueous layer was saturated by addition of sodium chloride and subsequently backextracted with additional methylene chloride. The combined organic layers were dried over sodium sulfate, filtered, and taken to a residue under reduced pressure to afford analytically pure 1-[3-(methyloxy)-4- nitrophenyl]piperazine as a yellow solid (1.7g, 7.2 mmol, quant, yield). 1H NMR (400 MHz, DMSOd6) δ ppm 2.28 - 2.37 (m, 1 H), 2.76 (q, J=4.64 Hz, 4 H), 3.29 - 3.34 (m, 4 H), 3.86 (s, 3 H), 6.46 (d, J=2.38 Hz, 1 H), 6.54 (dd, J=9.43, 2.47 Hz, 1 H), 7.84 (d, J=9.34 Hz, 1 H).
Step C/lntermediate A126: 1-[3-(methyloxy)-4-nitrophenyl]-4-(2-methylpropyl)piperazine
Figure imgf000131_0002
A solution of 1-[3-(methyloxy)-4-nitrophenyl]piperazine (2.09g, 8.44 mmol), 2-methyl- iodopropane (2.1g, 1 1.39 mmol), and potassium carbonate (4.7g, 34 mmol) in acetonitrile (80 mL) was heated in a pressure vessel at 8O0C for 24 hours. The reaction was cooled and the acetonitrile was removed under reduced pressure. The solids were dissolved in methylene chloride/water. The organic layer was dried over sodium sulfate, filtered, and the solvent removed under reduced pressure. The resultant residue was purified by chromatography on SiO2 (10% MeOH/CH2CL2 with 0.2% NH3) to give 1-[3- (methyloxy)-4-nitrophenyl]-4-(2-methylpropyl)piperazine as a pale yellow solid (1.7Og, 5.8 mmol, 69% yield). 1H NMR (400 MHz, CDCI3) ppm 0.91 (d, J=6.59 Hz, 6 H), 1.80 (sept t, J = 7.05, 6.91 , 1 H), 2.13 (d, J=6.04 Hz, 2 H), 2.52 (s, 4 H), 3.38 (s, 4 H), 3.93 (s, 3 H), 6.29 (d, J=2.38 Hz, 1 H), 6.40 (dd, J=9.43, 2.47 Hz, 1 H), 7.98 (d, J=9.52 Hz, 1 H).
Step D/lntermediate A123: 2-(methyloxy)-4-[4-(2-methylpropyl)-1-piperazinyl]aniline
Figure imgf000132_0001
A solution of 1-[3-(methyloxy)-4-nitrophenyl]-4-(2-methylpropyl)piperazine (1.7g, 5.82 mmol), FeCI3 (0.280 g, 1.75 mmol), activated carbon (2.Og), and hydrazine hydrate (2.23 ml_, 70 mmol) was heated in methanol (50 ml.) for 3 hours. The mixture was filtered over celite and concentrated to give 2-(methyloxy)-4-[4-(2-methylpropyl)-1- piperazinyl]aniline as a white solid (1.24g, 4.80 mmol, 83% yield). 1H NMR (400 MHz, CDCI3) δ ppm 0.94 (d, J=6.22 Hz, 6 H), 1.85 (s, 1 H), 2.20 (s, 2 H), 2.61 (s, 4 H), 3.10 (s, 4 H), 3.54 (s, 2 H), 3.83 (s, 3 H), 6.43 (m, 1 H), 6.52 (d, J=1.83 Hz, 1 H), 6.64 (d, J=8.42 Hz, 1 H).
Intermediate A127: 4-{4-[(dimethylamino)acetyl]-1 -piperazinyl}-2- (methyloxy)aniline
Figure imgf000132_0002
Step A/Intermediate A128: 1-(bromoacetyl)-4-[3-(methyloxy)-4-nitrophenyl]piperazine
Figure imgf000133_0001
A solution of bromoacetyl chloride (0.404 ml_, 4.85 mmol) was stirred in dichloromethane (8 ml.) at 0 0C. Potassium carbonate (0.436 g, 3.16 mmol) was added. In a separate vial, the 1-[3-(methyloxy)-4-nitrophenyl]piperazine (1.0 g, 4.21 mmol) was mixed with dichloromethane (8 ml_), then added dropwise to the reaction. Upon complete addition the reaction became a slurry, so additional dichloromethane was added to facilitate stirring (approximately 8 ml_). This mixture was stirred at 0 0C for about 1 hr. TLC indicated incomplete reaction so additional bromoacetyl chloride (0.100 ml.) was added. The reaction mixture was stirred for 1 h at rt. The reaction was then diluted with water and extracted with dichloromethane. The water layer's pH was checked and it was acidic, so aqueous (saturated) Na2CO3 was added until the pH was basic. The aqueous layer was extracted with dichloromethane and EtOAc. The combined organic layers were dried over MgSO4 and concentrated under vacuum to give 1-(bromoacetyl)-4-[3- (methyloxy)-4-nitrophenyl]piperazine as a bright yellow solid (1.49 g, 99%). 1H NMR (400 MHz, CDCI3) δ ppm 3.42-3.48 (m, 2 H), 3.49-3.58 (m, 2H), 3.71-3.79 (m, 2H), 3.79- 3.87 (m, 2H), 3.92 (s, 2H), 3.98 (s, 3H), 6.35 (d, J=1.8 Hz, 1 H), 6.45 (dd, J=9.2 Hz, 2.2 Hz, 1 H), 8.03 (d, J=9.3 Hz, 1 H).
Step B/lntermediate A129: N,N-dimethyl-2-{4-[3-(methyloxy)-4-nitrophenyl]-1- piperazinyl}-2-oxoethanamine
Figure imgf000133_0002
To a 0 0C slurry of 1-(bromoacetyl)-4-[3-(methyloxy)-4-nitrophenyl]piperazine (1.49 g, 4.16 mmol) in dichloromethane (8 mL) under nitrogen was added dimethylamine (2M in THF, 8.32 mL, 16.64 mmol). The resulting slurry was stirred for 30 minutes at 0 0C, then was allowed to warm to rt and stirred for about 1 hour. The resulting mixture was concentrated under vacuum. The residue was dissolved in EtOAc and water. The aqueous layer was extracted with EtOAc (with 5% IPA) and dichloromethane (with 5% IPA). The combined organic layers were dried over MgSO4 and concentrated under vacuum to give Λ/,Λ/-dimethyl-2-{4-[3-(methyloxy)-4-nitrophenyl]-1-piperazinyl}-2- oxoethanamine as a yellow solid (1.01 g, 75%). 1H NMR (400 MHz, CDCI3) δ ppm 2.46 (s, 6H), 3.38 (s, 2H), 3.40-3.52 (m, 4H), 3.75-3.87 (m, 4H), 3.97 (s, 3 H), 6.34 (d, J=1.6 Hz, 1 H), 6.43 (dd, J=9.3, 2.2 Hz, 1 H), 8.02 (d, J=9.3 Hz, 1 H); ESIMS (M+H)+ = 323.
Step C/lntermediate A127: 4-{4-[(dimethylamino)acetyl]-1-piperazinyl}-2- (methyloxy)aniline
Figure imgf000134_0001
N,N-dimethyl-2-{4-[3-(methyloxy)-4-nitrophenyl]-1-piperazinyl}-2-oxoethanamine (1.00 g, 3.10 mmol) was added to a thick-walled hydrogenation flask. To the flask were added ethanol (15 mL), ethyl acetate (15 mL), and methanol (15 mL). Under nitrogen, 10% palladium on carbon (0.330 g, 0.310 mmol) was added. The flask was purged with vacuum and nitrogen (3x). The flask was purged with vacuum and hydrogen (3x). The flask was then maintained under H2 atm (50 psi) for approximately 24 hours. The resulting mixture was diluted with methanol and dichloromethane, and filtered through a Celite pad using methanol and dichloromethane. The filtrate was concentrated under vacuum and the residue was purified by silica gel chromatography to give 4-{4-
[(dimethylamino)acetyl]-1-piperazinyl}-2-(methyloxy)aniline as a light grey solid (617 mg, 68%). 1H NMR (400 MHz, CDCI3) δ ppm 2.30 (s, 6H), 2.97-3.07 (m, 4H), 3.16 (s, 2H), 3.73-3.81 (m, 4H), 3.85 (s, 3H), 6.37-6.45 (m, 1 H), 6.52 (s, 1 H), 6.66 (d, J=8.2 Hz, 1 H). Intermediate A130: 2-(ethyloxy)-5-methyl-4-{4-[2-(methylsulfonyl)ethyl]-1- piperazinyljaniline
Figure imgf000135_0001
Step A/Intermediate A131 : 1-(ethyloxy)-5-fluoro-4-methyl-2-nitrobenzene
Figure imgf000135_0002
5-fluoro-4-methyl-2-nitrophenol (1 1.1 g, 65.0 mmol) and iodoethane (12.2 g, 78.0 mmol) were dissolved in 100 mL of DMSO with stirring. K2CO3 (13.5 g, 97.5 mmol) was added. The reaction was stirred for 3 h and then diluted with DCM and filtered. The filtrate was poured into H2O and extracted with DCM. The combined organic layers were dried over MgSO4, filtered, and concentrated in vacuo to provide 1-(ethyloxy)-5-fluoro-4-methyl-2- nitrobenzene (8.00 g, 39.4 mmol) as a pale yellow crystalline solid. 1H NMR (400 MHz, CDCI3) δ 7.76 (d, J = 8.1 Hz, 1 H), 6.70 (d, J = 11.0 Hz, 1 H), 4.10 (q, J = 7.0 Hz, 2H), 2.22 (s, 3H), 1.45 (t, J = 7.0 Hz, 3H).
Step B/lntermediate A132: 1-[5-(ethyloxy)-2-methyl-4-nitrophenyl]-4-[2- (methylsulfonyl)ethyl]piperazine
Figure imgf000135_0003
1-(ethyloxy)-5-fluoro-4-methyl-2-nitrobenzene (5.74 g, 28.8 mmol) was dissolved in DMSO (60 ml_). K2CO3 (1 1.9 g, 86.4 mmol) and 1-[2-(methylsulfonyl)ethyl]piperazine hydrochloride (9.85 g, 43.2 mmol) were added and the reaction mixture was heated to 100 0C and allowed to stir overnight. The mixture was cooled to rt then diluted with DCM and filtered. The filtrate was poured into H2O and extracted with DCM. The combined organics were dried with MgSO4, filtered, and concentrated in vacuo to give 1-[5- (ethyloxy)-2-methyl-4-nitrophenyl]-4-[2-(methylsulfonyl)ethyl]piperazine (9.70 g, 26.1 mmol, 90%) as an orange solid. 1H NMR (400 MHz, CDCI3) δ 7.75 (s, 1 H), 6.51 (s, 1 H), 4.1 1 (q, J = 7.0 Hz, 2H), 3.16 (t, J = 6.5 Hz, 2H), 3.03 (s, 3H), 3.00-2.91 (m, 6H), 2.72- 2.62 (m, 4H), 2.21 (s, 3H), 1.44 (t, J = 7.0 Hz, 3H).
Step C/lntermediate A130: 2-(ethyloxy)-5-methyl-4-{4-[2-(methylsulfonyl)ethyl]-1- piperazinyl}aniline
Figure imgf000136_0001
1 -[5-(ethyloxy)-2-methyl-4-nitrophenyl]-4-[2-(methylsulfonyl)ethyl]piperazine
(9.7 g, 26 mmol) was taken up in EtOH (100 ml_). The catalyst, 5% sulfided platinum on carbon (1.0 g) was added. The reaction was placed under 1 atm of H2 gas and was allowed to stir at rt overnight. The catalyst was filtered off and the filtrate was concentrated onto silica in vacuo. The product was purified on a 120 g ISCO column (DCM to 2% (2N NH3 in MeOH) to give 2-(ethyloxy)-5-methyl-4-{4-[2-
(methylsulfonyl)ethyl]-1-piperazinyl}aniline (1.9 g, 5.7 mmol, 22 %) as a pale orange oil. The mixed fractions (product and starting material) were resubmitted to the above conditions to provide additional product (5.2 g, 15 mmol, 58%). 1H NMR (400 MHz, CDCI3) δ 6.53 (s, 1 H), 6.52 (s, 1 H), 3.99 (q, J = 7.0 Hz, 2H), 3.56 (s, 2H), 3.15 (t, J = 6.3 Hz, 2H), 3.05 (s, 3H), 2.91 (t, J = 6.4 Hz, 2H), 2.84-2.78 (m, 4H), 2.67-2.54 (m, 4H), 2.13 (s, 3H), 1.38 (t, J = 7.0 Hz, 3H).
Intermediate A133: (3/?)-1-{1 -[4-amino-3-(methyloxy)phenyl]-4-piperidinyl}-Λ/,Λ/- dimethyl-3-pyrrolidinamine
Figure imgf000137_0001
Step A/Intermediate A134: (3R)-N,N-dimethyl-1-{1-[3-(methyloxy)-4-nitrophenyl]-4- piperidinyl}-3-pyrrolidinamine
Figure imgf000137_0002
A mixture of 1-[3-(methyloxy)-4-nitrophenyl]-4-piperidinone (1.0 g, 4.0 mmol), commercially available (3R)-(+)-3-dimethylaminopyrrolidine (1.0 g, 8.0 mmol), acetic acid (0.34 ml, 6 mmol) and triethylamine (0.56 ml, 4.0 mmol) in 1 ,2-dichloroethane (40 ml.) was stirred for 30 minutes. Sodium triacetoxyborohydride (1.3 g, 6.0 mmol) was added. When tic indicated the absence of starting material, the reaction was quenched by addition of sat'd NaHCO3. The reaction was diluted with DCM and the layers were separated. The aqueous phase was extracted with DCM. The combined organic layers were washed with water, dried over MgSO4 and concentrated to give a quantitative yield of (3R)-N,N-dimethyl-1-{1-[3-(methyloxy)-4-nitrophenyl]-4-piperidinyl}-3-pyrrolidinamine as a yellow oil. 1 H NMR (400 MHz, d~ 6DMSO) δ 7.84 (d, J = 9.2 Hz, 1 H), 6.55 (dd, J = 9.6 and 2.4 Hz, 1 H), 6.46 (d, J = 2.4 Hz, 1 H), 3.87-3.84 (m, 5H), 3.05-2.99 (m, 2H), 2.73 (t, J = 7.8 Hz, 1 H), 2.62-2.55 (m, 2H), 2.47-2.43 (m under DMSO peak, 1 H), 2.28-2.20 (m, 2H), 2.05 (s, 6H), 1.86-1.74 (m, 3H), 1.57-1.50 (m, 1 H), 1.44-1.34 (m, 2H).
Step B/lntermediate A133: (3R)-1-{1-[4-amino-3-(methyloxy)phenyl]-4-piperidinyl}-N,N- dimethyl-3-pyrrolidinamine
Figure imgf000138_0001
(3R)-Λ/,Λ/-dimethyl-1-{1-[3-(methyloxy)-4-nitrophenyl]-4-piperidinyl}-3-pyrrolidinamine (1.4g, 4.0 mmol) (crude from previous step) and 5% sulfided platinum on carbon (50 mg, catalytic amount) in EtOAc was stirred under 60 psi hydrogren pressure in a Fisher- Porter vessel. After 16 h, the reaction was filtered through a pad of celite, rinsing well with EtOAc. The filtrate was concentrated to give (3R)-1-{1-[4-amino-3- (methyloxy)phenyl]-4-piperidinyl}-N,N-dimethyl-3-pyrrolidinamine as a grey solid (1.09g, 3.4 mmol). 1H NMR (400 MHz, CDCI3 δ 6.61 (d, J = 8.4 Hz, 1 H), 6.50 (d, J = 2.4 Hz, 1 H), 6.40 (dd, J = 8.4 and 2.4 Hz, 1 H), 3.82 (s, 3H), 3.49-3.42 (m, 4H), 3.00 (t, J = 8.0 Hz, 1 H), 2.89-2.84 (m, 1 H), 2.75-2.61 (m, 3H), 2.53-2.47 (m, 1 H), 2.28 (t, J = 8.2 Hz, 1 H), 2.20 (s, 6H), 2.17-2.08 (m, 1 H), 2.04-1.92 (m, 3H), 1.73-1.60 (m, 3H).
Intermediate A135: 1-{2-[6-amino-5-(methyloxy)-2,3-dihydro-1H-indol-1-yl]-2- oxoethyl}-3-piperidinol
Figure imgf000138_0002
The 1-(bromoacetyl)-5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole (4.Og, 12.7 mmol) was dissolved in 50 ml. of dichloromethane, then K2CO3 (4.4g, 31.7 mmol) and 3-piperidinol (1.54g, 15.2 mmol) in 10 ml. dichloromethane were added, the reaction was stirred at RT overnight. The reaction mixture was diluted with 100 ml. of water, the organic solvents were washed with water (2x10OmL), dried over Na2SO4 and the solvent was removed under reduced pressure. The resulting residue was dissolved in 10 mL of EA, 25 mL of MeOH and 15 mL of THF, then 0.8 g 10% Pd/C was added, the reaction was stirred at RT overnight under balloon H2 pressure. The catalyst was removed via filtration and the solvent was removed under reduced pressure to yield 1-{2-[6-amino-5- (methyloxy)-2,3-dihydro-1 /-/-indol-1-yl]-2-oxoethyl}-3-piperidinol as a white solid (3.Og, 92%). 1HNMR (400 MHz, DMSOd6) δ ppm: 1.09 (m, 1 H), 1.44 (m, 1 H), 1.62 (m, 1 H), 1.74 (m, 1 H), 2.14(br s, 1 H), 2.47(s, 1 H), 2.74(br, 2H), 2.95 (m, 4 H), 3.50 (s, 2H), 3.68 (s, 3H), 4.04 (t, J=6.13Hz,2 H), 4.71 (s, 2H), 6.68 (s, 1 H), 7.51 (s, 1 H).
Intermediate A136 : 1 -{2-[6-amino-5-(methyloxy)-2,3-dihydro-1H-indol-1 -yl]-2- oxoethyl}-3-pyrrolidinol
Figure imgf000139_0001
The 1 -(bromoacetyl)-5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole (4.Og, 12.7 mmol) was dissolved in 50 ml. of dichloromethane, then K2CO3 (4.4g, 31.7 mmol) and 3-pyrrolidinol (1.33g, 15.2 mmol) in 10 ml. dichloromethane were added, and the reaction was stirred at RT overnight. The reaction mixture was diluted with 100 ml. of water, the organic solvents were washed with water (2x10OmL), dried over Na2SO4, and the solvent was removed under reduced pressure. The resulting residue was dissolved in 10 ml. of EA , 25 mL of MeOH and 15 ml. of THF, then 0.8 g 10% Pd/C was added, and the reaction was stirred at RT overnight under a balloon of H2 pressure. The catalyst was removed via filtration and the solvent was evaporated under reduced pressure to yield 1-{2-[6- amino-5-(methyloxy)-2,3-dihydro-1 /-/-indol-1-yl]-2-oxoethyl}-3-pyrrolidinol as a white solid (3.Og, 92%).1H NMR (400 MHz, DMSO-d6) δ ppm: 1.60 (m, 1 H), 1.97 (m, 1 H),, 2.47 (s, 1 H), 2.59 (m, 1 H), 2.72 (m, 1 H), 2.85 (m, 1 H), 2.96 (m, 4H), 3.50(s, 2H), 3.68 (s, 3H), 4.00 (t, J=8.33Hz,2 H), 4.86 (s, 2H), 6.68 (s, 1 H), 7.51 (s, 1 H).
Intermediate A137: 4-(1,4'-bipiperidin-1'-yl)-5-methyl-2-(methyloxy)aniline
Figure imgf000140_0001
Step A/Intermediate A138: 1 '-[2-methyl-5-(methyloxy)-4-nitrophenyl]-1 ,4'-bipiperidine
Figure imgf000140_0002
A yellow slurry of 1-fluoro-2-methyl-5-(methyloxy)-4-nitrobenzene (10 g, 54.0 mmol), 1 ,4'-bipiperidine (10.91 g, 64.8 mmol) and K2CO3 (18.66 g, 135 mmol) in dimethyl sulfoxide (200 ml) was heated at 130 0C for 2h, then was allowed to cool to 60 0C. The resulting slurry was filtered, the solids were washed with DMSO (50 ml_). The filtate was concentrated and the solids were washed with CH2CI2 (200 ml_). The filtrate was washed with water (200 ml_). The aqueous layer was back extracted with EtOAc (100 ml_). The organic layers were combined, concentrated onto Celite and purified by silica gel chromatography using 0-20% MeOH (containing 0.2% NH3)/CH2CI2 to obtain 1 '-[2- methyl-5-(methyloxy)-4-nitrophenyl]-1 ,4'-bipiperidine (1 1.0 g, 61 %) as an orange solid. 1H NMR (400 MHz, DMSO-d8) δ ppm 1.25-1.70 (m, 8H), 1.82 (d, J=11.72 Hz, 2H), 2.19 (s, 3H), 2.28-2.43 (m, 1 H), 2.70 (t, J=11.81 Hz, 2H), 3.32 (d, J=11.35 Hz, 3H), 3.90 (s, 3H), 6.68 (s, 1 H), 7.76 (s, 1 H), 3 protons are missing, they presumably overlap with DMSO peak; ESIMS (M+H)+ = 334.
Step B/lntermediate A137: 4-(1 ,4'-bipiperidin-1 '-yl)-5-methyl-2-(methyloxy)aniline
Figure imgf000141_0001
The 1'-[2-methyl-5-(methyloxy)-4-nitrophenyl]-1 ,4'-bipiperidine (1.5g, 4.5 mmol) was dissolved in 30 ml of THF, then 0.3 g 10% Pd/C was added, the reaction was stirred at RT overnight under H2 pressure (60 psi). The catalyst was filtered out, the solvent was removed to yield the 4-(3'-methyl-1 ,4'-bipiperidin-1 '-yl)-2-(methyloxy)aniline as a yellow solid, 1.3g (95%). 1H NMR (400 MHz, DMSOd6) δ ppm 1.33 (br. s., 2 H), 1.47 (br. s., 6 H), 1.73 (dd, J=6.97, 3.21 Hz, 2 H), 2.03 (s, 3 H), 2.48 (dt, J=3.57, 1.79 Hz, 6H), 2.90 (d, J=11.55 Hz, 2H), 3.31 (s., 1 H), 3.68 (s, 3H), 4.27 (s., 2H), 6.40 (s, 1 H), 6.52 (s, 1 H).
Intermediate A139 Λ/1-[3-amino-4-(methyloxy)phenyl]-Λ/2,Λ/2-dimethylglycinamide hydrogen chloride
Figure imgf000141_0002
Step A/Intermediate A140: 1 ,1-dimethylethyl [2-(methyloxy)-5-nitrophenyl]carbamate
Figure imgf000141_0003
To a solution of 2-(methyloxy)-5-nitroaniline (10 g, 59.5 mmol) in anhydrous THF (100 ml.) was added triethylamine (9.95 ml_, 71.4 mmol), followed by DMAP (0.727 g, 5.95 mmol) and BOC-anhydride (14.50 ml_, 62.4 mmol). The reaction mixture was stirred at rt for 19 h. The resulting slurry was diluted with EtOAc (300 ml_), washed with a 10% aqueous citric acid solution and a saturated NaCI solution. The organic layer was filtered through a pad of Celite. The filtrate was concentrated to afford a yellow foamy solid, which was then triturated using CH2Cb. The reaction was repeated on the same scale. The two crops of crude material were combined, dissolved in THF, concentrated onto Celite and purified by silica gel chromatography using 20-100% CH2Cl2/hexanes to afford 1 ,1-dimethylethyl [2-(methyloxy)-5-nitrophenyl]carbamate (4.66 g 14% for 2 batches). 1H NMR (400 MHz, DMSOd6) δ ppm 1.48 (s, 9H), 3.94 (s, 3H), 7.21 (d, J=9.16 Hz, 1 H), 7.97 (dd, J=9.06, 2.84 Hz, 1 H), 8.48 (s, 1 H), 8.68 (d, J=2.75 Hz, 1 H).
Step B/lntermediate A141 : 1 ,1-dimethylethyl [5-amino-2-(methyloxy)phenyl]carbamate
Figure imgf000142_0001
10% Pd on carbon (0.5 g, 17.37 mmol) was placed under N2 atm. EtOH (20 ml.) was added, followed by a slurry of 1 ,1-dimethylethyl [2-(methyloxy)-5-nitrophenyl]carbamate (4.66 g, 17.37 mmol) in EtOH (230 ml_). The reaction mixture was placed under H2 atm via a balloon and stirred at rt. After 3 days the resulting mixture was filtered through a pad of Celite using EtOH. The filtrate was concentrated. The residue was taken up into Et2O and concentrated again to afford 1 ,1-dimethylethyl [5-amino-2- (methyloxy)phenyl]carbamate (4.15 g, 100%). 1H NMR (400 MHz, DMSO-c/6) δ ppm 1.45 (s, 9H), 3.66 (s, 3H), 4.65 (br s, 2H), 6.20 (dd, J=8.61 , 2.75 Hz, 1 H), 6.68 (d, J=8.61 Hz, 1 H), 7.10 (d, J=2.01 Hz, 1 H), 7.56 (s, 1 H).
Step C/lntermediate A142: 1 ,1-dimethylethyl [5-[(Λ/,Λ/-dimethylglycyl)amino]-2-
(methyloxy)phenyl]carbamate
Figure imgf000142_0002
A solution of 1 ,1-dimethylethyl [5-amino-2-(methyloxy)phenyl]carbamate (1.5 g, 6.30 mmol) in THF (50 ml.) was cooled to 0 0C. K2CO3 (2.61 g, 18.89 mmol) was added, followed by dropwise addition of bromoacetyl chloride (0.685 ml_, 8.18 mmol). After 10 min the reaction mixture was allowed to warm to rt and treated with a Me2NH solution (2M in THF, 9.45 ml_, 18.90 mmol). After 4 h more Me2NH solution (2M in THF, 9.45 ml_, 18.90 mmol) was added and the mixture was stirred at rt. After 3 days the resulting mixture was concentrated onto Celite and purified by silica gel chromatography using 1- 10% MeOH (containing 0.2% NH3)/CH2CI2 to obtain 1 ,1-dimethylethyl [5-[{N,N- dimethylglycyl)amino]-2-(methyloxy)phenyl]carbamate as a white foamy semi-solid (1.7 g, 83%). 1H NMR (400 MHz, DMSOd6) δ ppm 1.46 (s, 9H), 2.26 (s, 6H), 3.21 (s, 2H), 3.76 (s, 3H), 6.91 (d, J=8.79 Hz, 1 H), 7.37 (dd, J=8.79, 2.38 Hz, 1 H), 7.86 (s, 1 H), 7.96 (d, J=2.38 Hz, 1 H), 9.56 (s, 1 H); ESIMS (M+H)+ = 323.
Step D/lntermediate A139: Λ/1-[3-amino-4-(methyloxy)phenyl]-Λ/2,Λ/2-dimethylglycinamide hydrogen chloride
Figure imgf000143_0001
A solution of 1 ,1-dimethylethyl [5-[(N,N-dimethylglycyl)amino]-2-
(methyloxy)phenyl]carbamate (1.7 g, 5.26 mmol) in dioxane (10 ml.) was treated with a 4N HCI solution in dioxane (19.71 ml_, 79 mmol). The resulting white slurry was maintained at rt. After 3 h the slurry was filtered, the solids were washed with Et2O to obtain Λ/1-[3-amino-4-(methyloxy)phenyl]-Λ/2,Λ/2-dimethylglycinamide hydrogen chloride as a white solid (1.28 g, 94%). 1H NMR (400 MHz, DMSO-d6) δ ppm 2.87 (s, 6H), 3.56 (s, 3H), 4.17 (s, 2H), 7.18 (d, J=9.16 Hz, 1 H), 7.50-7.67 (m, 1 H), 7.73-7.89 (m, 1 H), 10.14 (br s, 3H), 11.07-11.35 (m, 1 H).
Intermediate A143 1 -[(dimethylamino)acetyl]-6-methyl-1 ,2,3,4-tetrahydro-7-quinolinamine
Figure imgf000143_0002
Step A/Intermediate A144: N,N-dimethyl-2-(6-methyl-7-nitro-3,4-dihydro-1 (2H)- quinolinyl)-2-oxoethanamine
Figure imgf000144_0001
To a bright orange solution of 6-methyl-7-nitro-1 ,2,3,4-tetrahydroquinoline (2 g, 10.41 mmol) — (see Achvlediani, R.; Natsvlishvili, M.; Baberkina, E.; Khachidze, M.; Abesadze, I.; Suvorov, N. Synthesis of 1H-pyrrolo[3,2^y]- and IH-pyrrolo^S^ylquinoline. Izvestiya Akademii Nauk Gruzii, Seriya Khimicheskaya (1996), 22(1-4), 43-47.) — in THF (50 ml.) was added K2CO3 (4.31 g, 31.2 mmol). The resulting mixture was cooled to 0 0C and was treated with bromoacetyl chloride (1.040 ml_, 12.49 mmol) dropwise. The reaction mixture was srirred for 20 min. EtOAc (100 ml.) and water (100 ml.) were then added. The organic layer was dried (Na2SO4) and concentrated. The crude 1- (bromoacetyl)-6-methyl-7-nitro-1 ,2,3,4-tetrahydroquinoline (3.26 g, 10.41 mmol) was dissolved in THF (5OmL). A 2M Me2NH solution in THF (50 ml.) was added. The resulting slurry was filtered, the solids were rinsed with THF. The filtrate was concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to afford as a yellow solid (2.09 g, 72%). 1H NMR (400 MHz, DMSO-de) δ ppm 1.81-1.97 (m, 2H), 2.24 (s, 6H), 2.47 (s, 3H), 2.80 (t, J=6.89 Hz, 2H), 3.25 (s, 2H), 3.68-6.85 (m, 2H), 7.29 (s, 1 H), 8.54 (br s, 1 H); ESIMS (M+H)+ = 278.40.
Step B/lntermediate A143: 1-[(dimethylamino)acetyl]-6-methyl-1 ,2,3,4-tetrahydro-7- quinolinamine
Figure imgf000144_0002
10% Pd on carbon (0.5 g, 17.37 mmol) was placed under N2 atm. MeOH (20 ml.) was added, followed by a solution of N,N-dimethyl-2-(6-methyl-7-nitro-3,4-dihydro-1 (2H)- quinolinyl)-2-oxoethanamine (2.07 g, 7.46 mmol) in MeOH (80 ml_). The mixture was placed under H2 atm via balloon and maintained for 1 day. The resulting mixture was filtered through a pad of Celite using MeOH. The filtrate was concentrated, the residue was taken up into Et2O and concentrated again to afford 1-[(dimethylamino)acetyl]-6- methyl-1 ,2,3,4-tetrahydro-7-quinolinamine as a grey solid (1.55 g, 84%). 1H NMR (400 MHz, DMSOd6) δ ppm 1.71-1.87 (m, 2H), 1.99 (s, 3H), 2.20 (s, 6H), 2.45-2.58 (m, 2H), 3.18 (s, 2H), 3.61 (t, J=6.13 Hz, 2H), 4.66 (br s, 2H), 6.69 (s, 1 H), 6.78 (br s, 1 H); ESIMS (M+H)+ = 248.17.
Intermediate A145
(2/?)-1 -[(dimethylamino)acetyl]-2-methyl-5-(methyloxy)-2,3-dihydro-1H-indol-6- amine
Figure imgf000145_0001
Step A/Intermediate A146: (2R)-1-acetyl-2-methyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/- indole
Figure imgf000145_0002
A solution of (2R)-1-acetyl-2-methyl-5-(methyloxy)-2,3-dihydro-1 H-indole (100 mg, 0.487 mmol) — see (Arp, Forrest O.; Fu, Gregory C. Kinetic Resolutions of lndolines by a Nonenzymatic Acylation Catalyst. Journal of the American Chemical Society (2006), 128(44), 14264-14265.)— in TFA (2 ml.) at O0C was treated with sodium nitrite (33.6 mg, 0.487 mmol). The resulting red solution was stirred at 0 0C for 1 h. H2O (20 ml.) was added and the resulting slurry was filtered and the solids were washed with water to afford (2R)-1-acetyl-2-methyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole as a yellow solid (85 mg, 70%). 1H NMR (400 MHz, DMSO-d8) δ ppm 1.22 (d, J=6.41 Hz, 3H), 2.22 (s, 3H), 2.76 (d, J=17.21 Hz, 1 H), 3.45 (dd, J=17.12, 8.88 Hz, 1 H), 3.89 (s, 3H), 4.59-4.74 (m, 1 H), 7.35 (s, 1 H), 8.40 (s, 1 H); ESIMS (M+H)+ = 251.00. This procedure was repeated on 5 g of (2R)-1-acetyl-2-methyl-5-(methyloxy)-2,3- dihydro-1 H-indole to obtain 5.8 g of (2R)-1-acetyl-2-methyl-5-(methyloxy)-6-nitro-2,3- dihydro-1 /-/-indole. Step B/lntermediate A147: (2R)-2-methyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole hydrogen chloride
Figure imgf000146_0001
A slurry of (2R)-1-acetyl-2-methyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indole (5.78 g, 23.10 mmol) in MeOH (50 ml.) and a 4N HCI solution in dioxane (57.7 ml_, 231 mmol) was heated at 70 0C for 8 h. The resulting mixture was allowed to cool to rt and concentrated to afford (2R)-2-methyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole hydrogen chloride as a brown sticky solid (5.47 g, 97%). 1H NMR (400 MHz, DMSO-c/6) δ ppm 1.38 (d, J=6.59 Hz, 3H), 2.82 (dd, J=16.85, 7.69 Hz, 1 H), 3.32 (dd, J=16.85, 8.06 Hz, 1 H), 3.90 (s, 3H), 4.09-4.26 (m, 1 H), 7.39 (s, 1 H), 7.60 (s, 1 H); ESIMS (M+H)+ = 209.00.
Step C/lntermediate A148: N,N-dimethyl-2-[(2R)-2-methyl-5-(methyloxy)-6-nitro-2,3- dihydro-1 H-indol-1 -yl]-2-oxoethanamine
Figure imgf000146_0002
A 0 0C slurry of (2R)-2-methyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indole hydrogen chloride (4.55 g, 18.60 mmol) and K2CO3 (10.28 g, 74.4 mmol) in THF (200 ml.) was treated with bromoacetyl chloride (3.10 ml_, 37.2 mmol). After 2 h the reaction mixture was allowed to warm to rt. A 2M Me2NH solution in THF (55.8 ml_, 1 12 mmol) was added and the reaction mixture was stirred at rt for 18 h. The resulting thick slurry was filtered and the solids were rinsed with EtOAc. The filtrate was concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to obtain N,N-dimethyl-2-[(2R)-2-methyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indol-1-yl]- 2-oxoethanamine as an orange solid (2.92 g, 54%). 1H NMR (400 MHz, DMSO-c/6) δ ppm 1.23 (d, J=6.04 Hz, 3H), 2.28 ( s, 6H), 2.75 (d, J=17.03 Hz, 1 H), 3.14 (d, J=14.83 Hz, 1 H), 3.44 (d, J=14.65 Hz, 2H), 3.90 (s, 3H), 4.77-4.92 (m, 1 H), 7.36 (s, 1 H), 8.43 (s, 1 H); ESIMS (M+H)+ = 294.01. Step D/lntermediate A145: (2R)-1-[(dimethylamino)acetyl]-2-methyl-5-(methyloxy)-2,3- dihydro-1 H-indol-6-amine
Figure imgf000147_0001
10% palladium on carbon (0.25 g, 8.52 mmol) was placed under N2 atm. MeOH (10 ml.) was added, followed by a solution of N,N-dimethyl-2-[(2R)-2-methyl-5-(methyloxy)-6- nitro-2,3-dihydro-1 H-indol-1-yl]-2-oxoethanamine (2.92 g, 9.96 mmol) in MeOH (190 ml_). The slurry was purged with N2, then was placed under H2 atm via a rubber balloon and maintained at rt for 2 days. The resulting mixture was filtered through a pad of Celite and the filtrate was concentrated. The oily residue was taken up into Et2O and concentrated again to obtain (2R)-1-[(dimethylamino)acetyl]-2-methyl-5-(methyloxy)-2,3- dihydro-1 H-indol-6-amine as a beige solid (2.43 g, 93%). 1H NMR (400 MHz, DMSO-d8) δ ppm 1.17 (d, J=5.86 Hz, 3H), 2.26 (s, 6H), 2.44 (d, J=14.83 Hz, 1 H), 3.02 (d, J=14.28 Hz, 1 H), 3.10-3.27 (m, 1 H), 3.36 (d, J=14.28 Hz, 1 H), 3.72 (s, 3H), 4.52-4.77 (m, 3H), 6.72 (s, 1 H), 7.48 (s, 1 H); ESIMS (M+H)+ = 264.23.
Intermediate A149
(2S)-1-[(dimethylamino)acetyl]-2-methyl-5-(methyloxy)-2,3-dihydro-1H-indol-6- amine
Figure imgf000147_0002
Step A/Intermediate A150: (2S)-1-acetyl-2-methyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 H- indole
Figure imgf000147_0003
A 0 0C orange solution of (2S)-1-acetyl-2-methyl-5-(methyloxy)-2,3-dihydro-1 H-indole (5.19 g, 25.3 mmol) in TFA (30 ml.) was treated with NaNO2 (1.745 g, 25.3 mmol) portion wise. After 2 h more NaNO2 (800 mg, 11.6 mmol) was added. After 1 h the resulting mixture was poured into water (450 ml_). The resulting slurry was filtered and the solids were washed with water. The solids were dissolved in CH2CI2. The layers were separated, the organic layer was dried (Na2SO4) and concentrated to afford (2S)-1- acetyl-2-methyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole as a green solid (5.73 g, 91 %). 1H NMR (400 MHz, DMSO-d6) δ ppm 1.23 (d, J=6.23 Hz, 3H), 2.22 (s, 3H), 2.76 (d, J=17.21 Hz, 1 H), 3.45 (dd, J=17.03, 8.97 Hz, 1 H), 3.89 (s, 3H), 4.58-4.78 (m, 1 H), 7.35 (s, 1 H), 8.41 (s, 1 H); ESIMS (M+H)+ = 251.02.
Step B/lntermediate A151 : (2S)-2-methyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole hydrogen chloride
Figure imgf000148_0001
A slurry of (2S)-1-acetyl-2-methyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indole (5.7 g, 22.78 mmol) in MeOH (50 ml.) and a 4N HCI solution in dioxane (56.9 ml_, 228 mmol) was heated at 70 0C for 1O h. The resulting mixture was allowed to cool to rt and concentrated to afford (2S)-2-methyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 /-/-indole hydrogen chloride as a beige solid (5.55 g, 100%). 1H NMR (400 MHz, DMSO-c/6) δ ppm 1.39 (d, J=6.41 Hz, 3H), 2.84 (dd, J=16.94, 7.60 Hz, 1 H), 3.34 (dd, J=16.85, 8.06 Hz, 1 H), 3.90 (s, 3H), 4.12-4.27 (m, 1 H), 7.41 (s, 1 H), 7.66 (s, 1 H); ESIMS (M+H)+ = 209.00.
Step C/lntermediate A152: N,N-dimethyl-2-[(2S)-2-methyl-5-(methyloxy)-6-nitro-2,3- dihydro-1 H-indol-1 -yl]-2-oxoethanamine
Figure imgf000148_0002
A slurry of (2S)-2-methyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indole hydrogen chloride (4.43 g, 18.11 mmol) and K2CO3 (10.01 g, 72.4 mmol) in THF (200 mL) at O0C was treated with bromoacetyl chloride (3.02 mL, 36.2 mmol). After 2 h the resulting mixture was allowed to warm to rt. A 2M Me2NH solution in THF (54.3 mL, 109 mmol) was added. After 18 h the resulting thick slurry was filtered and the solids were washed with EtOAc. The filtrate was concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2. The foamy oily product was chased using CH2CI2 and hexanes, CH2CI2 and Et2O, then Et2O to obtain N,N-dimethyl-2-[(2S)-2-methyl-5-(methyloxy)-6-nitro-2,3-dihydro-1 H-indol-1-yl]-2- oxoethanamine as an orange solid (2.50 g, 47%). 1H NMR (400 MHz, DMSO-c/6) δ ppm 1.23 (d, J=6.04 Hz, 3H), 2.28 (s, 6H), 2.75 (d, J=17.21 Hz, 1 H), 3.14 (d, J=14.65 Hz, 1 H), 3.44 (d, J=14.65 Hz, 2H), 3.90 (s, 3H), 4.75-4.92 (m, 1 H), 7.37 (s, 1 H), 8.43 (s, 1 H); ESIMS (M+H)+ = 293.88.
Step D/lntermediate A149: (2S)-1-[(dimethylamino)acetyl]-2-methyl-5-(methyloxy)-2,3- dihydro-1 H-indol-6-amine
Figure imgf000149_0001
10% palladium on carbon (0.25 g, 8.52 mmol) was placed under N2 atm. MeOH (10 mL) was added, followed by a solution of N,N-dimethyl-2-[(2S)-2-methyl-5-(methyloxy)-6- nitro-2,3-dihydro-1 H-indol-1-yl]-2-oxoethanamine (2.50 g, 8.52 mmol) in MeOH (190 mL). The slurry was purged with N2, then was placed under H2 atm via a rubber balloon and maintained at rt for 2 days. The resulting mixture was filtered through a pad of Celite and the filtrate was concentrated. The oily residue was taken up into Et2O and concentrated again to obtain (2S)-1-[(dimethylamino)acetyl]-2-methyl-5-(methyloxy)-2,3- dihydro-1 H-indol-6-amine as a beige solid (2.10 g, 94%). 1H NMR (400 MHz, DMSO-c/8) δ ppm 1.17 (d, J=5.68 Hz, 3H), 2.26 (s, 6H), 2.44 (d, J=15.02 Hz, 1 H), 3.03 (d, J=14.28 Hz, 1 H), 3.10-3.25 (m, 1 H), 3.36 (d, J=14.28 Hz, 1 H), 3.72 (s, 3H), 4.54-4.76 (m, 3H), 6.72 (s, 1 H), 7.48 (s, 1 H); ESIMS (M+H)+ = 264.20.
Intermediate A153 i-^dimethylaminoJacetyll-Λ^jΛ^-dimethyl^jS-dihydro-IH-indole-Sjθ-diamine
Figure imgf000150_0001
Step A/Intermediate A154: 1-acetyl-5-fluoro-6-nitro-2,3-dihydro-1 /-/-indole
Figure imgf000150_0002
To a solution of 5-fluoro-1 /-/-indole (5.0 g, 37 mmols) in acetic acid (300 ml) was added sodium cyanoborohydride (2.79 g, 44 mmols). After stirring overnight at rt, the reaction was concentrated under reduced pressure, redissolved in ethyl acetate (300 ml) and the pH was adjusted to 8 with saturated sodium bicarbonate. The organic layer was separated, filtered through a cotton plug, evaporated, and placed on the high vacuum for one hour prior to the next synthetic step. The crude material was then dissolved in acetic acid (100 ml), followed by the addition of acetic anhydride (3.5 ml, 37 mmols). After heating at 6OC for one hour, the reaction was poured into ice, stirred for one hour, solid was removed by vacuum filtration, rinsed with water and allowed to dry overnight under house vacuum. Next, to a solution of the 1-acetyl-5-fluoro-2,3-dihydro-1 H-indole (500 mg, 2.79 mmol) in sulfuric acid (10 ml) at OC was added nitric acid (0.196 ml, 3.07 mmols) by a slow dropwise addition. After stirring 30 min at OC, the reaction was quenched by pouring into ice water. The solids were removed by vacuum filtration, redissolved in dichloromethane (50 ml), adsorbed to silica gel, and purified by LC( 25% to 75% ethyl acetate/hexanes ). The lower Rf spot was isolated and the structure was determined to be the desired regioisomer by IR profiling. 1 H NMR (400 MHz, DMSO- d6) δ ppm 2.18 (s, 3 H) 3.25 (t, J=8.63 Hz, 2 H) 4.18 (t, J=8.53 Hz, 2 H) 7.50 (d, J= 11.24 Hz, 1 H) 8.64 (d, J=7.22 Hz, 1 H).
Step B/lntermediate A155: 5-fluoro-6-nitro-2,3-dihydro-1 /-/-indole
Figure imgf000151_0001
A solution of 1-acetyl-5-fluoro-6-nitro-2,3-dihydro-1 H-indole (1.22 g, 5.44 mmol) and 4.0 M HCI/dioxane (6.80 ml, 27.2 mmol) in tetrahydrofuran (50 ml) and methanol (100 ml) was heated overnight at 5OC. The reaction was quenched with saturated NaHCOβ (200 ml), solvent removed, aqueous layer extracted with dichloromethane (250 ml), organic layer adsorbed to silica gel and purified by LC (20-75% ethyl acetate/hexanes) to afford the indoline (0.9 g, 91 %). ESIMS (M+H)+ = 183.
Step C/lntermediate A156: 1-[(dimethylamino)acetyl]-Λ/,Λ/-dimethyl-6-nitro-2,3-dihydro- 1 /-/-indol-5-amine
Figure imgf000151_0002
To a solution of 5-fluoro-6-nitro-2,3-dihydro-1 H-indole (0.9 g, 4.94 mmol) and potassium carbonate (2.049 g, 14.82 mmol) in tetrahydrofuran (300 ml) was added bromoacetyl chloride (0.414 ml, 4.94 mmol) and the reaction was stirred at rt for 30 min. To this reaction was added 2.0M dimethylamine in THF (7.41 ml, 14.82 mmol) and the mixture was stirred overnight at rt. The solvent removed, water added (200 ml), aqueous layer extracted with dichloromethane (2 x 250 ml), organic layers adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM) to give the title compound (1g, 69%). ESIMS (M+H)+ = 293. Step D/lntermediate A153: 1-[(dimethylamino)acetyl]-Λ/5,Λ/5-dimethyl-2,3-dihydro-1/-/- indole-5,6-diamine
Figure imgf000152_0001
To an N2 degassed solution of 1-[(dimethylamino)acetyl]-N,N-dimethyl-6-nitro-2,3- dihydro-1 H-indol-5-amine (1.00 g, 3.42 mmol) and 10% Pd/C (3.64 g, 3.42 mmol) in ethanol (100 ml) was added H2 and the reaction was stirred at rt overnight on the Fisher Porter at 50 psi. The reaction was filtered through celite, rinsed with methanol (100 ml), concentrated by rotary evaporation, and high vacced prior to the next reaction to give the title compound (0.9g, 100%). ESIMS (M+H)+ = 263.
Section B: Pyrrolopyrimidine Intermediates
Intermediate B1 : 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3- c/]pyrimidin-4-yl}amino)-2-thiophenecarboxamide
Figure imgf000152_0002
A mixture of 2,4-dichloro-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidine (6.22 g, 18.2 mmol), 3-amino-2-thiophenecarboxamide (2.35 g, 16.5 mmol) and TFA (6.4 ml_, 82.6 mmol) in 2,2,2-trifluoroethanol (100 ml.) was heated at 80 0C for 3 days. The resulting mixture was allowed to cool to rt and filtered. The solids were washed with Et2O to obtain 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4- yl}amino)-2-thiophenecarboxamide as a yellow solid (4.0 g, 49%). 1H NMR (400 MHz, THF-d8) δ ppm 2.37 (s, 3H), 6.69 (d, J=4.03 Hz, 1 H), 7.48 (d, 8.42 Hz, 2H), 7.55-7.95 (m, 4H), 8.00 (d, J=8.24 Hz, 2H), 8.05 (d, J=5.49 Hz, 1 H); ESIMS (M+H)+ = 447.98.
Intermediate B2: 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrogen chloride
Figure imgf000153_0001
Step A/Intermediate B3: methyl 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3- c/]pyrimidin-4-yl}amino)-2-thiophenecarboxylate
Figure imgf000153_0002
A solution of trifluoroacetic acid (63.0 ml, 818 mmol), 2,4-dichloro-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidine (40 g, 117 mmol), and methyl 3- amino-2-thiophenecarboxylate (27.6 g, 175 mmol) in 2,2,2-trifluoroethanol (450 ml) was maintained at 9O0C for 16 hours, at which time a thick white precipitate formed. The slurry was filtered while warm and solids collected to afford methyl 3-({2-chloro-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2-thiophenecarboxylate (35.6 g, 77 mmol, 65.8 % yield). 1H NMR (400 MHz, DMSOd6) δ ppm 10.28 (s, 1 H), 7.96 (d, J=8.24 Hz, 2 H), 7.91 - 7.94 (m, 1 H), 7.86 - 7.89 (m, 1 H), 7.74 (dd, J=4.03,
0.92 Hz, 1 H), 7.44 (d, J=8.61 Hz, 2 H), 6.85 (d, J=4.03 Hz, 1 H), 3.78 (s, 3 H), 2.33 (s, 3 H).
Step B/lntermediate B2: 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one hydrogen chloride
Figure imgf000154_0001
A mixture of methyl 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin- 4-yl}amino)-2-thiophenecarboxylate (20.1 g, 43.4 mmol) and THF (1400 ml.) was treated with a solution of LiOH (10.40 g, 434 mmol) in water (200 ml_). The reaction mixture was maintained at rt for 7 h. The layers were separated, the organic layer was washed with a saturated NaCI solution (2x200 ml_), concentrated down to a thick slurry. The slurry was filtered and the solids were washed with Et2O. The filtrate was concentrated and diluted with Et2O. The resulting slurry was filtered and the solids were washed with Et2O. The filtrates were combined, concentrated onto Celite and purified by silica gel chromatography using 5-20% MeOH/THF. The second batch of solids and the product from chromatography were dissolved in warm THF (500 ml.) and filtered through pad of silica gel using 20% MeOH/THF. The filtrate was concentrated, the residue was dissolved in THF (400 ml.) and CH2CI2 (250 ml_). DMF (0.507 ml_, 6.55 mmol) was added, followed by slow addition of a solution of oxalyl chloride (2M in CH2CI2, 13.64 ml_, 27.3 mmol). The reaction mixture was stirred for 17 h, then was cooled to 0 0C and filtered. The filtrate was treated with additional oxalyl chloride solution (2M in CH2CI2, 20 ml_, 40 mmol). After 1 h the reaction mixture was cooled to 0 0C and filtered. The two batches of solids were combined to obtain 5-chloro-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one hydrogen chloride as a white solid (6.65 g, 36%). ESIMS (M+H)+ = 431.08.
Intermediate B4: 5-chloro-9-methyl-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrogen chloride
Figure imgf000154_0002
Step A/Intermediate B5: methyl 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3- c/]pyrimidin-4-yl}amino)-5-methyl-2-thiophenecarboxylate
Figure imgf000155_0001
A mixture of 2,4-dichloro-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidine (33.0 g, 96 mmol) and methyl S-amino-δ-methyl^-thiophenecarboxylate (15 g, 88 mmol) in 2,2,2-trifluoroethanol (500 ml.) was treated with TFA (33.7 ml_, 438 mmol), then heated at 80 0C. Some of the volatiles were distilled off (300 ml. total over 3 h). The resulting thick slurry was filtered hot and the solids were washed with warm TFE to obtain methyl 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-5- methyl-2-thiophenecarboxylate (16.2 g, 39%). 1H NMR (400 MHz, DMSO-c/6) δ ppm 2.38 (s, 3H), 3.80 (s, 3H), 6.83 (br s, 1 H), 7.49 (d, J=7.87 Hz, 2H), 7.70 (s, 1 H), 7.78 (d, J=3.48 Hz, 1 H), 8.01 (d, J=7.87 Hz, 2H), 10.33 (s, 1 H), a CH3 signal in missing, may overlap with DMSOd6; ESIMS (M+H)+ = 477.12.
Step B/lntermediate B4: 5-chloro-9-methyl-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one hydrogen chloride
Figure imgf000155_0002
A mixture of methyl 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-
4-yl}amino)-5-methyl-2-thiophenecarboxylate (16.2 g, 34.0 mmol) and THF (1400 ml.) was treated with a solution of LiOH (8.13 g, 340 mmol) in water (200 ml_). The resulting mixture was stirred at rt for 34 h. The layers were separated, the organic layer was washed with a saturated NaCI solution (2 x 200 ml_), dried (Na2SO4) and concentrated to about 300 ml_. The resulting slurry was filtered, the filtrate concentrated onto Celite and purified by silica gel chromatography using 0-30% MeOH/THF. The derived solids (4.22 g, 9.12 mmol) and DMF (5 drops) in tetrahydrofuran (100 ml) were stirred while 2.0M oxalyl chloride in dichloromethane (5.70 ml, 1 1.39 mmol) was added dropwise over five minutes. Stirring was continued until bubbling ceased, then the reaction was cooled to O0C and stirring stopped. The solids which precipitated were collected by vacuum filtration and washed with diethyl ether to afford 5-chloro-9-methyl-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrogen chloride (3.42 g, 7.10 mmol, 78 % yield). 1H NMR (400 MHz, DMSOd6) δ ppm 8.02 (d, J=8.43 Hz, 2 H), 7.77 (d, J=3.67 Hz, 1 H), 7.50 (d, J=8.07 Hz, 2 H), 7.14 (s, 1 H), 7.03 (d, J=3.67 Hz, 1 H), 6.66 (s, 1 H), 2.60 (s, 3 H), 2.37 (s, 3 H).
Intermediate B6: 5-chloro-3-[(4-methylphenyl)sulfonyl]-9-
(trifluoromethyl)pyrrolo[3,2-e]thieno[3\2V4,5]pyrimido[1,2-c]pyrimidin-7(3H)-one hydrogen chloride
Figure imgf000156_0001
Step A/Intermediate B7: rac syn-2,3-dibromo-4,4,4-trifluorobutanenitrile and rac anti-2,3- dibromo-4,4,4-trifluorobutanenitrile
Figure imgf000156_0002
An orange solution of (2£)-4,4,4-trifluoro-2-butenenitrile (5 g, 41.3 mmol) and Br2 (2.128 ml_, 41.3 mmol) in CCI4 (120 ml.) was heated at the reflux temperature for 3 h. The resulting mixture was diluted with H2O (5 ml.) and washed with a 5% aqueous sodium thiosulfate solution (50 ml.) and a saturated NaCI solution (50 ml_). The organic layer was dried (Na2SO4) and concentrated to obtain a mixture of rac syn-2,3-dibromo-4,4,4- trifluorobutanenitrile and rac anf/-2,3-dibromo-4,4,4-trifluorobutanenitrile as a yellow oil (10.4 g, 90%). 1H NMR (400 MHz, DMSO-d6) δ ppm 5.72-5.83 (m, 0.6H), 5.94-6.06 (m, 0.4H), 6.13-6.24 (m, 1 H).
Step B/lntermediate B8: methyl 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3- c/]pyrimidin-4-yl}amino)-5-(trifluoromethyl)-2-thiophenecarboxylate and ethyl 3-({2-chloro- 7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-5-(trifluoromethyl)-2- thiophenecarboxylate
Figure imgf000157_0001
A solution of a mixture of rac syn-2,3-dibromo-4,4,4-trifluorobutanenitrile and rac anti- 2,3-dibromo-4,4,4-trifluorobutanenitrile (10.4 g, 37.0 mmol) in EtOH (150 mL) was treated with methyl mercaptoacetate (3.31 mL, 37.0 mmol), followed by dropwise addition Of Et3N (5.16 mL, 37.0 mmol). The reaction mixture was maintained at rt for 17. Additional Et3N (10 mL, 63 mmol) was added and the reaction mixture was maintained at rt for 3 days. The resulting slurry was filtered, the filtrate was diluted with EtOAc (200 mL) and washed with a saturated NaCI solution (2x100 mL). The organic layer was concentrated onto Celite and purified by silica gel chromatography using 50-100% CH2Cl2/hexanes. The mixture of the crude products and 2,4-dichloro-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidine (5.24 g, 15.31 mmol) in a 4N HCI solution in dioxane (100 mL) was heated at 80 0C for 21 h. The resulting mixture was allowed to cool to rt and filtered. The solids were washed with Et2O to obtain a 2:1 mixture of methyl 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4- yl}amino)-5-(trifluoromethyl)-2-thiophenecarboxylate and ethyl 3-({2-chloro-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-5-(trifluoromethyl)-2- thiophenecarboxylate as a white solid (1.65 g). 1H NMR (400 MHz, DMSO-c/6) δ ppm 1.19 (t, J=IAA Hz, 1 H), 2.38 (s, 3H), 3.86 (s, 2H), 4.29 (q, J=4.28 Hz, 0.65H), 6.93-7.02 (m, 1 H), 7.49 (d, J=8.24 Hz, 2H), 7.76-7.85 (m, 1 H), 7.96-8.05 (m, 2H), 8.31 (d, J=0.73 Hz, 0.33H) 8.34 (d, J=0.73 Hz, 0.67H) 1.032-10.41 (m, 1 H); ESIMS (M+H)+ = 531.01 and 545.04.
Step C/lntermediate B6: 5-chloro-3-[(4-methylphenyl)sulfonyl]-9- (trifluoromethyl)pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one hydrogen chloride
Figure imgf000157_0002
A solution of methyl 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin- 4-yl}amino)-5-(trifluoromethyl)-2-thiophenecarboxylate (1.65 g, 3.11 mmol) in THF (130 ml.) was treated with a solution of LiOH (0.744 g, 31.1 mmol) in water (40 ml_). The resulting mixture was stirred vigorously at rt for 3.5 h, then diluted with EtOAc (100 ml_). The organic layer was washed with a saturated NaCI solution (2x50 ml_), concentrated onto Celite and purified by silica gel chromatography using 1-20% MeOH (containing 2% AcOH)/THF. The crude product was taken up into benzene and concentrated. The residue was dissolved in THF (50 ml.) and treated with a drop of DMF, followed by oxalyl chloride (0.450 ml_, 5.15 mmol). After 30 minutes at rt the reaction mixture was concentrated, taken up into benzene and concentrated again. The residue was triturated using Et2O to obtain 5-chloro-3-[(4-methylphenyl)sulfonyl]-9-(trifluoromethyl)pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one hydrogen chloride as a yellow solid (1.05 g). 1H NMR (400 MHz, THF-d8) δ ppm 2.40 (s, 3H), 2.84 (br s, 2H), 7.01 (d, J=3.66 Hz, 1 H), 7.44 (d, J=8.24 Hz, 2H), 7.72 (d, 3.85 Hz, 1 H), 7.82 (s, 1 H), 8.12 (d, 8.42 Hz, 2H); ESIMS (M+H)+ = 498.86
Intermediate B9: 5-chloro-3-[(4-methylphenyl)sulfonyl]-3H,7H-pyrrolo[3,2- e]thieno[3',4':4,5]pyrimido[1 ,2-c]pyrimidin-7-one hydrogen chloride
Figure imgf000158_0001
Step A/Intermediate B10: methyl 4-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H- pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-3-thiophenecarboxylate
Figure imgf000158_0002
A brown slurry of 2,4-dichloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidine (26.1 g, 72.5 mmol) and methyl 4-amino-3-thiophenecarboxylate (Maybridge, 11.4 g, 72.5 mmol) in 2,2,2-trifluoroethanol (400 ml.) and TFA (27.9 ml_, 363 mmol) was heated at the reflux temperature. After 40 min half of the solvent was removed via distillation and the remaining mixture was heated at 80 0C overnight. The resulting slurry was allowed to cool somewhat and was filtered warm. The solid was washed with 2,2,2- trifluoroethanol to obtain methyl 4-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3- c/]pyrimidin-4-yl}amino)-3-thiophenecarboxylate as a beige solid (14.0 g, 42%). 1H NMR (400 MHz, DMSOd6) δ ppm 2.37 (s, 3H), 3.76 (s, 3H), 6.76 (br s, 1 H), 7.48 (d, J=8.24 Hz, 2H), 7.73 (d, J=4.03 Hz, 1 H), 7.85 (d, J=3.3 Hz, 1 H), 7.99 (d, J=8.24 Hz, 2H), 8.39 (d, J=3.48 Hz, 1 H), 10.20 (s, 1 H); ESIMS (M+H)+ = 463.02.
Step B/lntermediate B1 1 : 4-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3- c/]pyrimidin-4-yl}amino)-3-thiophenecarboxylic acid
Figure imgf000159_0001
A slurry of methyl 4-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4- yl}amino)-3-thiophenecarboxylate (14.0 g, 27.2 mmol) in THF (1000 ml.) was treated with a solution of LiOH (6.52 g, 272 mmol) in water (200 ml_). The reaction mixture was stirred for 3 h. EtOAc (1000 ml.) was added and the organic layer was washed with a saturated NaCI solution (2x300 ml_), dried (Na2SO4) and concentrated. The residue was diluted with benzene and concentrated to obtain 4-({2-chloro-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-3-thiophenecarboxylic acid as a pale yellow solid (13.32 g, quantitative yield). 1H NMR (400 MHz, DMSO-c/6) δ ppm 2.36 (s, 3H), 6.62 (d, J=3.85 Hz, 1 H), 7.47 (d, J=8.06 Hz, 2H), 7.73 (dd, J=7.51 Hz, 3.66 Hz, 2H), 7.79 (d, J=3.48 Hz, 1 H), 7.98 (d, J=8.42 Hz, 2H); ESIMS (M+H)+ = 448.94.
Step C/lntermediate B9: 5-chloro-3-[(4-methylphenyl)sulfonyl]-3H,7/-/-pyrrolo[3,2- e]thieno[3',4':4,5]pyrimido[1 ,2-c]pyrimidin-7-one hydrogen chloride
Figure imgf000159_0002
To a yellow slurry of 4-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin- 4-yl}amino)-3-thiophenecarboxylic acid (13.32 g, 29.7 mmol) in THF (250 mL) were added two drops of DMF, followed by a solution of oxalyl chloride (2M in CH2CI2, 37.1 mL, 74.2 mmol). The reaction mixture was stirred at rt and cooled to 0 0C. The resulting slurry was filtered and the solids were washed with Et2O to obtain 5-chloro-3-[(4- methylphenyl)sulfonyl]-3/-/,7/-/-pyrrolo[3,2-e]thieno[3',4':4,5]pyrimido[1 ,2-c]pyrimidin-7- one hydrogen chloride as a yellow solid (12.15 g, 88%). ESIMS (M+H)+ = 430.99.
Intermediate B12: 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3- c/]pyrimidin-4-yl}amino)-2-furancarboxylic acid
Figure imgf000160_0001
Step A/Intermediate B13: 1 ,1-dimethylethyl 3-furanylcarbamate
Figure imgf000160_0002
A mixture of 3-furancarboxylic acid (17.5 g, 156 mmol), DPPA (47.3 g, 172 mmol), tBuOH (22.40 mL, 234 mmol), TEA (26.1 mL, 187 mmol) and toluene (500 mL) was heated at the reflux temperature under N2 atm. After 5 h the resulting mixture was allowed to cool to rt and filtered through pad of silica gel using toluene. The filtrate was concentrated to a slurry and filtered. The solids were washed with hexanes. The filtrate was further concentrated, the resulting slurry was filtered. The solids were washed with hexanes. The two batches were combined to afford 1 ,1-dimethylethyl 3- furanylcarbamate as a white solid (11.71 g, 41%). 1H NMR (400 MHz, DMSO-c/6) δ ppm 1.44 (s, 9H), 6.33 (s, 1 H), 7.46 (d, J= 1.47 Hz, 1 H), 7.64 (br s, 1 H), 9.24 (br s, 1 H).
Step B/lntermediate B14: methyl 3-({[(1 ,1-dimethylethyl)oxy]carbonyl}amino)-2- furancarboxylate
Figure imgf000161_0001
A solution of 1 ,1-dimethylethyl 3-furanylcarbamate (11 g, 60.0 mmol) and N, N, N1N'- tetramethylethylenediamine (22.47 ml, 150 mmol) in tetrahydrofuran (350 ml) was cooled to -780C and t-butyllithium in hexanes (88 ml, 150 mmol) was added dropwise over 10 minutes. The solution was allowed to warm to -4O0C (monitored by internal temperature sensor) for 2 hours and then methylchloroformate (5.75 ml, 74.5 mmol) was added. The reaction was warmed to O0C and maintained for 45 minutes. The reaction was diluted with saturated ammonium chloride, the organic layer washed with brine, dried over sodium sulfate, filtered, concentrated under reduced pressure, and purified by column chromatography to afford methyl 3-({[(1 ,1-dimethylethyl)oxy]carbonyl}amino)-2- furancarboxylate (7.91 g, 32.8 mmol, 54.6 % yield) as a pale yellow oil. 1H NMR (400 MHz, DMSOd6) δ ppm 8.31 (s, 1 H), 7.83 (d, J=1.81 Hz, 1 H), 7.08 (s, 1 H), 3.81 (s, 3 H), 1.47 (s, 9 H)
Step C/lntermediate B15: methyl 3-amino-2-furancarboxylate
Figure imgf000161_0002
A yellow solution of methyl 3-({[(1 ,1-dimethylethyl)oxy]carbonyl}amino)-2- furancarboxylate (7.92 g, 32.8 mmol) in CH2CI2 (100 ml.) was treated with TFA (10 ml_). The resulting deep orange solution was stirred at rt. After 5 h the reaction mixture was poured into a saturated NaHCO3 solution (400 ml_). Solid K2CO3 was added until the aqueous layer turned basic. The organic layer was washed with a saturated NaCI solution (100 ml_), concentrated onto Celite and purified by silica gel chromatography using 10-100% EtOAc/hexanes as a light brown oil (2.14 g, 46%). 1H NMR (400 MHz, DMSO-de) δ ppm 3.70 (s, 3H), 5.67 (br s, 2H), 6.18 (d, 1.83 Hz, 1 H), 7.52 (d, J=1.83 Hz, 1 H); ESIMS (M+H)+ = 142.1 1.
Step D/lntermediate B16: methyl 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7/-/- pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2-furancarboxylate
Figure imgf000162_0001
A slurry of 2,4-dichloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidine (5.19 g, 15.16 mmol) and methyl 3-amino-2-furancarboxylate (2.14 g, 15.16 mmol) in a 4N solution of HCI in dioxane (100 mL) was heated at 80 0C for 42 h and was allowed to cool to rt. EtOAc (200 mL) and a saturated NaHCO3 solution (300 mL) were added, followed by solid K2CO3 to obtain neutral pH. The organic layer was washed with a saturated NaCI solution (100 mL), dried (Na2SO4), concentrated onto Celite and purified by silica gel chromatography using 20-100% CH2CI2/hexanes to afford methyl 3-({2- chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2- furancarboxylate as a pale yellow solid (3.1 g, 46%). 1H NMR (400 MHz, DMSO-c/6) δ ppm 2.37 (s, 3H), 3.81 (s, 3H), 7.02 (d, J=4.03 Hz, 1 H), 7.23 (d, J=1.83 Hz, 1 H), 7.48 (d, 8.06 Hz, 2H), 7.76 (d, 3.85 Hz, 1 H), 7.93 (d, J=1.83 Hz, 1 H), 7.98 (s, 1 H), 8.00 (s, 1 H), 9.75 (s, 1 H); ESIMS (M+H)+ = 447.03.
Step E/lntermediate B12: 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3- c/]pyrimidin-4-yl}amino)-2-furancarboxylic acid
Figure imgf000162_0002
To a solution of methyl 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3- d]pyrimidin-4-yl}amino)-2-furancarboxylate (3.41 g, 7.63 mmol) in THF (200 mL) was added a solution of LiOH (1.827 g, 76 mmol) in water (50 mL). The resulting mixture was stirred vigorously at rt. After 10 h EtOAc (200 mL) was added. The organic layer was washed with a saturated NaCI solution (2x100 mL), concentrated onto Celite and purified by silica gel chromatography using 50-100% THF/CH2CI2, then 0-30% MeOH/THF to obtain 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2- furancarboxylic acid as a pale yellow solid (0.66 g, 20%). 1H NMR (400 MHz, DMSO-c/6) δ ppm 2.37 (s, 3H), 6.76 (d, J=3.85 Hz, 1 H), 7.25 (d, J=1.83 Hz, 1 H), 7.47 (d, J=8.06 Hz, 2H), 7.63 (d, J=1.83 Hz, 1 H), 7.72 (d, J=4.03 Hz, 1 H), 7.98 (d, J=8.42 Hz, 2H), 11.14 (br s, 1 H); ESIMS (M+H)+ = 432.97.
Intermediate B17: 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrido[3,2- cπpyrrolo[2',3':4,5]pyrimido[1,6-a]pyrimidin-7(3H)-one hydrogen chloride
Figure imgf000163_0001
Step A/Intermediate B18: 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3- c/]pyrimidin-4-yl}amino)-2-pyridinecarboxylic acid
Figure imgf000163_0002
A slurry of 2,4-dichloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidine (4.41 g, 12.89 mmol), S-amino^-pyridinecarboxylic acid (1.78 g, 12.89 mmol) and DIPEA (11.25 ml_, 64.4 mmol) in iPrOH (100 ml.) was heated at 90 0C for 5 days. The resulting mixture was allowed to cool to rt, diluted with EtOAc (200 ml.) and a 1 N aqueous HCI solution. The resulting slurry was filtered, the solids were washed with Et2O and dried in vacuum at 60 0C overnight to obtain 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H- pyrrolo[2,3-<y]pyrirrιidin-4-yl}arrιino)-2-pyridinecarboxylic acid as a light green solid (3.08 g, 54%). 1H NMR (400 MHz, DMSO-d6) δ ppm 2.38 (s, 3H), 6.83 (d, J=4.03 Hz, 1 H), 7.49 (d, J=8.06 Hz, 2H), 7.72-7.88 (m, 2H), 8.00 (d, J=8.,24 Hz, 2H), 8.42 (dd, J=4.58, 1.10 Hz, 1 H), 8.74-8.82 (m, 1 H), 1 1.83 (s, 1 H); ESIMS (M+H)+ = 444.11.
Step B/lntermediate B17: 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrido[3,2- c/]pyrrolo[2',3':4,5]pyrimido[1 ,6-a]pyrimidin-7(3H)-one hydrogen chloride
Figure imgf000163_0003
A solution of 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4- yl}amino)-2-pyridinecarboxylic acid (107 mg, 0.241 mmol) in THF (50 ml.) was treated with a drop of DMF followed by oxalyl chloride (0.2 ml_, 2.29 mmol). The reaction mixture was stirred at rt for 3 days. The resulting slurry was filtered, the solids were washed with Et2O to afford 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrido[3,2- c/]pyrrolo[2',3':4,5]pyrimido[1 ,6-a]pyrimidin-7(3/-/)-one hydrogen chloride as a yellow solid (93 mg, 83%). 1H NMR (400 MHz, THF-d8) δ ppm 2.40 (s, 3H), 4.78 (br s, 1 H), 7.16 (dd, J=3.75, 0.82 Hz, 1 H), 7.45 (d, J=8.24 Hz, 2H), 7.65-7.70 (m, 1 H), 8.04-8.16 (m, 3H), 8.72 (d, J=4.21 Hz, 1 H), 7.73 (dd, J=8.33, 4.12 Hz, 1 H); ESIMS (M+H)+ = 426.00.
Section C: Examples Example 1
3-[(2-{[4-[4-(1 -methylethyl)-1 -piperazinyl]-2-(methyloxy)phenyl]amino}-1 H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000164_0001
Step A/Intermediate C1 : 3-({2-{[4-[4-(1-methylethyl)-1-piperazinyl]-2- (methyloxy)phenyl]amino}-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4- yl}amino)-2-thiophenecarboxamide
Figure imgf000165_0001
A mixture of 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4- yl}amino)-2-thiophenecarboxamide (400 mg, 0.89 mmol) and 4-[4-(1-methylethyl)-1- piperazinyl]-2-(methyloxy)aniline (223 mg, 0.89 mmol), a 4N HCI solution in dioxane (0.9 ml_, 3.56 mmol) and 2,2,2-trifluoroethanol (7 ml.) was heated at 8O0C for 7 days. Additional 4N HCI solution in dioxane (3 ml_, 12 mmol) was added and the reaction mixture was heated at 8O0C for 7 days. THF (20 ml.) and a 27% aqueous NH4OH solution (100 ml.) were added, and the reaction mixture was maintained at rt for 2 days. The resulting mixture was diluted with EtOAc (100 ml.) and a saturated NaCI solution (100 ml_). The aqueous layer was back extracted with EtOAc (100 ml_). The organic layers were combined, dried (Na2SO4), concentrated onto Celite and purified by silica gel chromatography using 0-10% MeOH (containing 0.2% NH3)/CH2CI2 to obtain 3-({2-{[4-[4- (1-methylethyl)-1-piperazinyl]-2-(methyloxy)phenyl]amino}-7-[(4-methylphenyl)sulfonyl]- 7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2-thiophenecarboxamide as an orange solid (130 mg, 22%). 1H NMR (400 MHz, DMSO-d6) δ ppm 1.03 (d, J=6.04 Hz, 6H), 2.35 (s, 3H), 2.53-2.80 (m, 5H), 3.18 (br s, 4H), 3.81 (s, 3H), 5.76 (s, 1 H), 6.47 (d, J=3.66 Hz, 1 H), 6.60 (d, J=8.79 Hz, 1 H), 6.68 (s, 1 H), 7.28-7.42 (m, 3H), 7.46-7.82 (m, 4H), 7.92 (d, J=7.69 Hz, 2H), 8.10 (s, 1 H) 1 1.49 (s, 1 H); ESIMS (M+H)+ = 661.19.
Step B/Example 1 : 3-[(2-{[4-[4-(1 -methylethyl)-1 -piperazinyl]-2-
(methyloxy)phenyl]amino}-1 H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2- thiophenecarboxamide
A mixture of 3-({2-{[4-[4-(1-methylethyl)-1-piperazinyl]-2-(methyloxy)phenyl]amino}-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2-thiophenecarboxamide (1 12 mg, 0.17 mmol) and NaOMe (92 mg, 1.7 mmol), THF (20 mL) and MeOH (20 mL) was maintained at rt for 2 days. Additional NaOMe (108 mg, 3.4 mmol) was added and the reaction mixture was stirred for 2 days, then was heated at 60 0C for 5 h. The resulting mixture was diluted with EtOAc (50 mL), washed with a saturated NaHCOs solution (25 mL) and a saturated NaCI solution (25 mL). The aqueous layers were back extracted with EtOAc (50 mL). The organic layers were combined, concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to obtain 3-[(2-{[4-[4-(1-methylethyl)-1-piperazinyl]-2- (methyloxy)phenyl]amino}-1 H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2- thiophenecarboxamide as a yellow solid (60 mg, 70%). 1H NMR (400 MHz, DMSO-c/6) δ ppm 1.02 (d, J=6.23 Hz, 6H), 2.53-277 (m, 5H), 3.1 1 (s, 4H), 3.81 (s, 3H), 6.22 (s, 1 H), 6.49 (d, J=8.61 , 1 H), 6.63 (s, 1 H), 6.95 (s, 1 H), 7.36-7.74 (m, 4H), 7.83 (d, J=8.42 Hz, 1 H), 8.42 (d, J=4.76 Hz, 1 H), 11.31 (s, 1 H), 1 1.36 (s, 1 H); ESIMS (M+H)+ = 507.02.
Example 2
3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino}-1H- pyrrolo[2,3-cy]pyrimidin-4-yl)amino]-2 -thiophenecarboxamide
Figure imgf000166_0001
Step A/Intermediate C2: 3-({2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 H- indol-6-yl]amino}-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2- thiophenecarboxamide
Figure imgf000167_0001
A solution of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (400 mg, 0.928 mmol) and 1- [(dimethylamino)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-amine (289 mg, 1.160 mmol) in 2,2,2-trifluoroethanol was heated to 9O0C in a sealed tube for 45 minutes. The reaction was cooled, poured into DCM/saturated sodium bicarbonate and the organic layer taken to a residue under reduced pressure. Trituration from diethyl ether afforded 5-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one which was dissolved in 27% aqueous NH4OH solution (50 ml.) in THF (50 ml.) was heated at 80 0C for 17 h. The resulting mixture was diluted with EtOAc (50 ml.) and a saturated NaCI solution (100 ml_). The organic layer was washed with a saturated NaCI solution (100 ml_), dried (Na2SO4), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to obtain 3-({2-{[1- (Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2-thiophenecarboxamide (235 mg). 1H NMR (400 MHz, DMSO-d6) δ ppm 2.24 (s, 6H), 2.34 (s, 3H), 3.06-3.28 (m, 4H), 3.74 (s, 3H), 4.24 (t, J=7.87 Hz, 2H), 6.39-6.49 (m, 1 H), 7.07 (s, 1 H), 7.19-7.41 (m, 3H), 7.42-7.75 (m, 3H), 7.94 (d, J=7.87 Hz, 2H), 8.04-8.26 (m, 2H), 8.44 (s, 1 H), 1 1.49 (s, 1 H); ESIMS (M+H)+ = 661.29.
Step B/Example 2: 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
A mixture of 3-({2-{[1-(N,N-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6- yl]amino}-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2- thiophenecarboxamide (235 mg, 0.356 mmol), NaOMe (192 mg, 3.56 mmol), THF (20 ml.) and methanol (40 ml.) was heated at 80 0C for 4 h. The resulting mixture was diluted with EtOAc (100 ml_), washed with a saturated NaHCO3 solution (100 ml.) and a saturated NaCI solution (100 ml_). The aqueous layers were back extracted with EtOAc (50 ml_). The organic layers were dried (Na2SO4), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)ZCH2CI2 to obtain 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-1H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide as a yellow solid (92 mg, 51 %). 1H NMR (400 MHz, DMSO-d6) δ ppm 2.25 (s, 6H), 3.05-3.25 (m, 4 H), 3.78 (s, 3H), 4.19 (t, J=8.06 Hz, 2H), 6.23 (s, 1 H), 6.97 (s, 2H), 7.37-7.84 (m, 4H), 8.41 (d, J=4.94 Hz, 1 H), 8.59 (s, 1 H), 1 1.30 (s, 1 H), 1 1.41 (s, 1 H); ESIMS (M+H)+ = 507.27.
Example 3
3-[(2-{[5-(methyloxy)-1 -(1 -pyrrolidinylacetyl)-2,3-dihydro-1 H-indol-6-yl]amino}-1 H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000168_0001
A mixture of 5-(methyloxy)-1-(1-pyrrolidinylacetyl)-2,3-dihydro-1 H-indol-6-amine (256 mg, 0.928 mmol) and 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrogen chloride (400 mg, 0.928 mmol) in 2,2,2-trifluoroethanol (10 ml.) was heated at 80 0C for 3 h. The resulting mixture was allowed to cool to rt, diluted with CH2CI2 (100 ml_), and a NaHCO3 solution (100 ml_). The organic layer was washed with a saturated NaHCO3 solution (100 ml.) and a saturated NaCI solution (100 ml_), dried (Na2SO4) and concentrated. The residue was triturated with Et2O to afford crude 5-{[5-(methyloxy)-1-(1-pyrrolidinylacetyl)-2,3-dihydro- 1 H-indol-6-yl]amino}pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one. The solids were taken up into THF (50 ml.) and a 27% aqueous NH4OH solution (200 ml_). The reaction mixture was heated at 80 0C for 28 h. The resulting mixture was allowed to cool to rt and diluted with EtOAc (200 ml.) and a saturated NaCI solution (100 ml_). The aqueous layer was back extracted with EtOAc (200 ml_). The organic layers were combined, washed with a saturated NaCI solution (2 x 100 ml_), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2Cl2. The isolated intermediate product was dissolved in THF (10 ml.) and MeOH (10 ml_), and was treated with NaOMe (192 mg, 3.55 mmol). The reaction mixture was heated at 70 0C for 3 h. The resulting mixture was allowed to cool to rt, diluted with EtOAc (100 ml_), washed with a saturated NaHCO3 solution (50 ml.) and a saturated NaCI solution (50 ml_). The organic layer was concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to obtain 3- [(2-{[5-(methyloxy)-1-(1-pyrrolidinylacetyl)-2,3-dihydro-1 H-indol-6-yl]amino}-1 H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide as a light brown solid (55 mg). 1H NMR (400 MHz, DMSO-d6) δ ppm 1.7 (br s, 4H), 2.56 (br s, 4H), 3.14 (t, J=7.87 Hz, 2H), 3.37 (s, 2H), 3.78 (s, 3H), 4.18 (t, J=8.06 Hz, 2H), 6.23 (s, 1 H), 6.97 (s, 2H), 7.33-7.79 (m, 4H), 8.41 (d, J=4.94 Hz, 1 H), 8.58 (s, 1 H), 1 1.30 (s, 1 H), 11.41 (s, 1 H); ESIMS (M+H)+ = 533.30.
Example 4 3-{[2-({5-(methyloxy)-1-[(methyloxy)acetyl]-2,3-dihydro-1 H-indol-6-yl}amino)-1H- pyrrolo[2,3-c/]pyrimidin-4-yl]amino}-2-thiophenecarboxamide
Figure imgf000169_0001
A mixture of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one hydrogen chloride (400 mg, 0.856 mmol) and 5-(methyloxy)-1-[(methyloxy)acetyl]-2,3-dihydro-1 H-indol-6-amine (201 mg, 0.851 mmol) in THF (15 ml.) was heated at 80 0C for 17 h. The resulting mixture was diluted with THF (30 ml.) and a 27% aqueous NH4OH solution (200 ml_). The reaction mixture was heated at 80 0C. After 17 h the mixture was allowed to cool to rt and diluted with EtOAc (50 ml.) and a saturated NaCI solution (50 ml_). The organic layer was washed with a saturated NaCI solution (2x50 ml_), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)ZCH2CI2. The isolated intermediate product was dissolved in THF (6 ml.) and MeOH (6 ml.) and was treated with NaOMe (198 mg, 3.67 mmol). The reaction mixture was heated at 80 0C for 3 h. The resulting mixture was allowed to cool to rt and diluted with EtOAc (50 ml.) and a saturated NaHCO3 solution (50 ml_). The organic layer was washed with a saturated NaCI solution (50 ml_), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to obtain 3-{[2-({5- (methyloxy)-i -[(methyloxy)acetyl]-2,3-dihydro-1 H-indol-6-yl}amino)-1 H-pyrrolo[2,3- c/]pyrimidin-4-yl]amino}-2-thiophenecarboxamide (53 mg). 1H NMR (400 MHz, DMSO-c/6) δ ppm 3.14 (t, J=8.06 Hz, 2H), 3.79 (s, 3H), 4.05 (t, J=8.33 Hz, 2H), 4.18 (s, 2H), 6.18- 6.29 (m, 1 H), 6.90-7.06 (m, 2H), 7.33-7.82 (m, 4H), 8.12 (d, J=5.31 Hz, 1 H), 8.62 (s,
1 H), 1 1.32 (s, 1 H), 11.42 (s, 1 H) a CH3O signal is missing, may overlap with water peak; ESIMS (M+H)+ = 494.13.
Example 5 3-[(2-{[4-[4-(Λ/,Λ/-dimethylglycyl)-1 -piperazinyl]-2-(methyloxy)phenyl]amino}-1 H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000170_0001
Step A/Intermediate C3: 3-({2-{[4-[4-(Λ/,Λ/-dimethylglycyl)-1-piperazinyl]-2-
(methyloxy)phenyl]amino}-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4- yl}amino)-2-thiophenecarboxamide
Figure imgf000171_0001
A mixture of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrogen chloride (400 mg, 0.855 mmol) and 4-{4-[(dimethylamino)acetyl]-1-piperazinyl}-2-(methyloxy)aniline (250 mg, 0.855 mmol) in THF (50 ml.) was stirred at rt for 20 h. A 27% aqueous NH4OH solution (200 ml.) was added and the reaction mixture was heated at 80 0C for 6 h. The resulting mixture was allowed to cool to rt, and diluted with EtOAc (100 ml.) and a saturated NaCI solution (100 ml_). The organic layer was washed with a saturated NaCI solution (2x100 ml_), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)ZCH2CI2, followed by trituration using a mixture of CH2CI2 and Et2O to obtain 3-({2-{[4-[4-(Λ/,Λ/-dimethylglycyl)-1-piperazinyl]-2- (methyloxy)phenyl]amino}-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4- yl}amino)-2-thiophenecarboxamide as a yellow solid (104 mg, 17%). 1H NMR (400 MHz, DMSO-de) δ ppm 2.20 (s, 6H), 2.35 (s, 3H), 3.04-3.28 (m, 6H), 3.64 (br s, 2H), 3.73 (br s, 2H), 3.82 (s, 3H), 6.47 (d, J=3.85 Hz, 1 H), 6.64 (d, J=8.42 Hz, 1 H), 6.73 (s, 1 H), 7.29- 7.44 (m, 3H), 7.44-7.85 (m, 5H), 7.92 (d, J=7.51 Hz, 1 H), 8.11 (s, 1 H), 8.27 (br s, 1 H), 11.49 (s, 1 H); ESIMS (M+H)+ = 704.27.
Step B/Example 5: 3-[(2-{[4-[4-(Λ/,Λ/-dimethylglycyl)-1-piperazinyl]-2- (methyloxy)phenyl]amino}-1 H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2- thiophenecarboxamide A mixture of 3-({2-{[4-[4-(Λ/,Λ/-dimethylglycyl)-1-piperazinyl]-2-(methyloxy)phenyl]amino}- 7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2- thiophenecarboxamide (104 mg, 0.148 mmol), dioxane (10 mL) and a 1 N aqueous KOH solution (2.60 mL, 2.60 mmol) was heated at 80 0C for 16.5 h. The resulting mixture was allowed to cool to rt and diluted with EtOAc (100 mL). The organic layer was washed with a saturated NaHCOs solution (50 mL), a saturated NaCI solution (50 mL), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to obtain 3-[(2-{[4-[4-(Λ/,Λ/-dimethylglycyl)-1-piperazinyl]-2- (methyloxy)phenyl]amino}-1 H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2- thiophenecarboxamide as a light brown solid (38.7 mg, 48%). 1H NMR (400 MHz,
CDCI3+CD3OD) δ ppm 2.18 (s, 6H), 2.98 br s, 4H), 3.06 (s, 2H), 3.52-3.68 (m, 4H), 3.74 (s, 3H), 6.29 (d, J=3.1 1 Hz, 1 H), 6.34-6.53 (m, 2H), 6.71 (d, J=3.30 Hz, 1 H), 7.22 (d, J=5.49 Hz, 1 H), 7.96 (d, J=8.42 Hz, 1 H), 8.29 (d, J=5.49 Hz, 1 H); ESIMS (M+H)+ = 550.28.
Example 6
5-methyl-3-[(2-{[4-[4-(1-methylethyl)-1-piperazinyl]-2-(methyloxy)phenyl]amino}- 1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2 -thiophenecarboxamide
Figure imgf000172_0001
A suspension of 5-chloro-9-methyl-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (450 mg, 1.011 mmol) and [4-[4-(1- methylethyl)-1-piperazinyl]-2-(methyloxy)phenyl]amine (315 mg, 1.264 mmol) in 2,2,2- trifluoroethanol (8 ml) was warmed to 9O0C in a sealed pressure vessel for 16 hours. The solution was cooled, poured into dichloromethane/saturated sodium bicarbonate, and the organic layer was taken to a residue under reduced pressure and triturated with diethyl ether. The derived solids were mixed with a 27% aqueous NH4OH solution (190 ml.) in THF (60 ml.) and heated at 80 0C for 17 h. The resulting mixture was diluted with EtOAc (100 ml.) and the layers were separated. The aqueous layer was back extracted with EtOAc (100 ml_). The organic layers were combined, washed with a saturated NaCI solution (2 x 100 ml_), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2Cl2. The isolated intermediate product was taken up into THF (5 ml.) and MeOH (5 ml_), and was treated with NaOMe (1 15 mg, 2.134 mmol). The reaction mixture was heated at 80 0C for 1.5 h, then was diluted with EtOAc (100 ml_), washed with a saturated NaHCO3 solution (50 ml.) and a saturated NaCI solution (75 ml_). The organic layer was concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to obtain 5- methyl-3-[(2-{[4-[4-(1 -methylethyl)-1 -piperazinyl]-2-(methyloxy)phenyl]amino}-1 H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide as a yellow solid (90 mg). 1H NMR (400 MHz, DMSO-d6) δ ppm 1.01 (d, J=6.41 Hz, 6H), 2.44 (s, 3H), 2.52-2.75 m, 5H), 3.12 (br s, 4H), 3.79 (s, 3H), 6.19 (s, 1 H), 6.50 (d, J=8.79 Hz, 1 H), 6.63 (s, 1 H),
6.94 (s, 1 H), 7.38 (br s, 2H), 7.51 (s, 1 H), 7.65 (d, J=8.42 Hz, 1 H), 8.09 (s, 1 H), 11.25 (s, 1 H), 1 1.39 (s, 1 H); ESIMS (M+H)+ = 521.33.
Example 7 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino}-1H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-5-methyl-2-thiophenecarboxamide
Figure imgf000173_0001
A suspension of 5-chloro-9-methyl-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (450 mg, 1.011 mmol) and 1- [(dimethylamino)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-amine (303 mg, 1.214 mmol) in 2,2,2-trifluoroethanol (8 ml) was warmed to 9O0C in a sealed pressure vessel. After 1 hour all solids had dissolved, and the reaction was cooled to room temperature and poured into dichloromethane/saturated sodium bicarbonate. The organic layer was taken to a residue under reduced pressure and the oil was triturated with diethyl ether. The derived solids were taken up into THF (60 ml.) and a 27% aqueous NH4OH solution (190 ml_), and was heated at 80 0C for 24 h. The resulting mixture was allowed to cool to rt and diluted with EtOAc (100 ml_). The aqueous layer was back extracted with EtOAc (50 ml_). The organic layers were combined, washed with a saturated NaCI solution (2 x 100 ml_), concentrated onto Celite and purified by silica gel chromatography using 1- 10% MeOH (containing 0.2% NH3)/CH2CI2. The isolated intermediate product was dissolved in THF (5 ml.) and MeOH (5 ml_), and was treated with NaOMe (96 mg, 1.778 mmol). The reaction mixture was heated at 80 0C until all starting material was consumed as evident by LCMS. The resulting mixture was diluted with EtOAc (100 ml_), washed with a saturated NaHCOs solution (50 ml.) and a saturated NaCI solution (75 ml_). The organic layer was concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2to obtain 3-[(2-{[1- (Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}-1 /-/-pyrrolo[2,3- c/]pyrimidin-4-yl)amino]-5-methyl-2-thiophenecarboxamide as an off-white solid (62 mg). 1H NMR (400 MHz, DMSO-d6) δ ppm 2.24 (s, 6H), 2.37 (s, 3H), 3.06-3.22 (m, 4H), 3.76 (s, 3H), 4.17 (t, J=8.24 Hz, 2H), 6.16-6.27 (m, 1 H), 6.95 (s, 1 H), 6.99 (s, 1 H), 7.38 (br s, 2H), 7.74 (s, 1 H), 8.05 (s, 1 H), 8.46 (s, 1 H), 1 1.25 (s, 1 H), 11.43 (s, 1 H); ESIMS (M+H)+ = 521.26.
Example 8
5-methyl-3-[(2-{[5-(methyloxy)-1 -(1 -pyrrolidinylacetyl)-2,3-dihydro-1 H-indol-6- yl]amino}-1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000174_0001
A mixture of 5-(methyloxy)-1-(1-pyrrolidinylacetyl)-2,3-dihydro-1 H-indol-6-amine (265 mg, 0.962 mmol) and 5-chloro-9-methyl-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrogen chloride (428 mg, 0.962 mmol) in 2,2,2-trifluoroethanol (10 ml.) was heated at 80 0C for 3 h. The resulting mixture was allowed to cool to rt, diluted with CH2Cb (100 ml.) and a saturated NaHCOs solution (100 ml_). The organic layer was washed with a saturated NaCI solution (100 ml_), dried (Na2SO4) and concentrated. The residue was triturated with Et2O. The crude product was taken up into in THF (50 ml.) and a 27% aqueous NH4OH solution (200 ml_). The reaction mixture was heated at 80 0C for 28 h. The resulting mixture was allowed to cool to rt, diluted with EtOAc (200 ml.) and a saturated NaCI solution (100 ml_). The aqueous layer was back extracted with EtOAc (200 ml_). The organic layers were combined, washed with a saturated NaCI solution (2 x 100 ml_), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2. The isolated intermediate product was dissolved in in THF (10 ml.) and MeOH (10 ml.) and treated with NaOMe (202 mg, 3.73 mmol). The reaction mixture was heated at 70 0C for 3 h. The resulting mixture was allowed to cool to rt, diluted with EtOAc (100 ml.) and washed with a saturated NaHCO3 solution (50 ml_). The organic layer was diluted with a saturated NaCI solution (50 ml_). The resulting slurry was filtered and the solids were washed with Et2O to obtain 5-methyl-3-[(2-{[5-(methyloxy)-1-(1-pyrrolidinylacetyl)-2,3- dihydro-1 /-/-indol-6-yl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2- thiophenecarboxamide as a white solid (122 mg). 1H NMR (400 MHz, DMSO-c/6) δ ppm 1.69 br s, 4H), 2.37 (s, 3H), 2.53 (br s, 4H), 3.15 (t, J=8.42 Hz, 2H), 3.76 (s, 3H), 4.17 (t, J=8.06 Hz, 2H), 6.20 (s, 1 H), 6.95 (s, 1 H), 6.99 (s, 1 H), 7.38 (s, 2H), 7.73 (s, 1 H), 8.05 (s, 1 H), 8.45 (s, 1 H), 11.26 (s, 1 H), 1 1.44 (s, 1 H), a CH2 signal is missing, may overlap with water peak; ESIMS (M+H)+ = 547.29.
Example 9
3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino}-1H- pyrrolo^.S-c/lpyrimidin^-ylJaminol-S-ttrifluoromethylJ^-thiophenecarboxamide
Figure imgf000176_0001
Step A/Intermediate C4: 3-({2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 H- indol-6-yl]amino}-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-5-
(trifluoromethyl)-2-thiophenecarboxamide
Figure imgf000176_0002
A mixture of 5-chloro-3-[(4-methylphenyl)sulfonyl]-9-(trifluoromethyl)pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrogen chloride (358 mg, 0.602 mmol) and 1-[(dimethylamino)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-amine (150 mg, 0.602 mmol) in THF (50 ml.) was heated at 80 0C for 17 h. The resulting mixture was allowed to cool to rt and diluted with a 27% aqueous NH4OH solution (200 ml_). The reaction mixture was stirred at rt for 1 h, and diluted with EtOAc (100 ml.) and a saturated NaCI solution (50 ml_). The organic layer was washed with a saturated NaCI solution (2x100 ml_), dried (Na2SO4), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)ZCH2CI2 to obtain as a yellow solid (270 mg, 62%). 1H NMR (400 MHz, DMSO-d6) δ ppm 2.22 (s, 6H), 2.35 (s, 3H), 3.05-3.27 (m, 4H), 3.72 (s, 3H), 4.19 (t, 8.15 Hz, 2H), 6.47 (d, 3.85 Hz, 1 H), 7.05 (s,1 H), 7.24-7.45 (m, 3H), 7.71-8.11 (m, 4H), 8.14 (s, 1 H), 8.48 (br s, 1 H), 8.62 (s, 1 H), 11.41 (s, 1 H); ESIMS (M+H)+ = 729.36. Step B/Example 9: 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-5-(trifluoromethyl)-2- thiophenecarboxamide
A mixture of 3-[(2-{[1-(N,N-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-5-(trifluoromethyl)-2- thiophenecarboxamide (270 mg, 0.370 mmol), a 1 N aqueous KOH solution (3.70 ml_, 3.70 mmol) and dioxane (20 ml.) was heated at 80 0C for 9 h. The resulting mixture was diluted with EtOAc (100 ml_), washed with a saturated NaHCO3 solution (100 ml.) and a saturated NaCI solution (50 ml_). The organic layer was concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)ZCH2CI2 to obtain 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}- 1 H-pyrrolo^^-c/lpyrimidin^-yOaminol-S-^rifluoromethyl^-thiophenecarboxamide as a yellow solid (63 mg, 30%). 1H NMR (400 MHz, DMSO-d6) δ ppm 2.21 (s, 6H), 3.03-3.23 (m, 4H), 3.74 (s, 3H), 4.15 (t, J=8.33 Hz, 2H), 6.23 (d, J=1.47 Hz, 1 H), 6.90-7.04 (m, 2H), 7.69-8.18 (m, 3H), 8.40 (s, 1 H), 8.76 (s, 1 H), 11.24-11.47 (m, 2H); ESIMS (M+H)+ = 575.1 1.
Example 10
3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-
1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-5-(trifluoromethyl)-2- thiophenecarboxamide
Figure imgf000177_0001
Step A/Intermediate C5: 3-({2-{[1-(Λ/,Λ/-dimethylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-
7-quinolinyl]amino}-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-
5-(trifluoromethyl)-2-thiophenecarboxamide
Figure imgf000178_0001
A slurry of 5-chloro-3-[(4-methylphenyl)sulfonyl]-9-(trifluoromethyl)pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrogen chloride (390 mg, 0.656 mmol) and 1 -[(dimethylamino)acetyl]-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinamine (173 mg, 0.656 mmol) in THF (15 mL) was heated at 80 0C for 17 hours. Additional 1- [(dimethylamino)acetyl]-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinamine (27 mg, 0.102 mmol) was added and the reaction mixture was heated at 80 0C for 17 hours. THF (40 mL) was added, followed by a 27% aqueous NH4OH solution (200 mL). The reaction mixture was heated at 80 0C for 30 min and then allowed to cool to rt. EtOAc (100 mL) and a saturated NaCI solution (50 mL) were added. The organic layer was washed with a saturated NaCI solution (2 x 100 mL), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to obtain 3-({2- {[1-(Λ/,Λ/-dimethylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-5-(trifluoromethyl)-2- thiophenecarboxamide as a yellow solid (193 mg, 40%). ESIMS (M+H)+ = 743.38.
Step B/Example 10: 3-[(2-{[1 -(Λ/,Λ/-dimethylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-5-(trifluoromethyl)-2- thiophenecarboxamide
A mixture of 3-({2-{[1-(N,N-dimethylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-5- (trifluoromethyl)-2-thiophenecarboxamide (193 mg, 0.260 mmol) and a 1 N aqueous KOH solution (2.60 mL, 2.60 mmol) in dioxane (10 mL) was heated at 80 0C for 17 h. The resulting mixture was allowed to cool to rt and diluted with EtOAc (100 ml.) and a saturated NaHCOs solution (100 ml_). The organic layer was washed with a saturated NaCI solution (50 ml_), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)ZCH2CI2 to obtain 3-[(2-{[1- (Λ/,Λ/-dimethylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-1/-/- pyrrolo[2,3-(^pyrimidin-4-yl)amino]-5-(trifluoromethyl)-2-thiophenecarboxamide as a beige solid (25 mg, 16%). 1H NMR (400 MHz, THF-d8) δ ppm 1.23-1.79 (m, 8H) 2.71 (br s, 2H), 3.71 (s, 2H), 3.58-3.95 (m, 5H), 6.25 (s, 1 H), 6.84 (s, 1 H), 7.02 (s, 1 H), 7.64-8.28 (m, 4H), 8.79 (s, 1 H),11.30 (s, 1 H),1 1.41 (br s, 1 H); ESIMS (M+H)+ = 589.20.
Example 11
4-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino}-1H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-3-thiophenecarboxamide
Figure imgf000179_0001
Step A/Intermediate C6: 4-({2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 H- indol-6-yl]amino}-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-3- thiophenecarboxamide
Figure imgf000179_0002
A yellow slurry of 5-chloro-3-[(4-methylphenyl)sulfonyl]-3H,7H-pyrrolo[3,2- e]thieno[3',4':4,5]pyrimido[1 ,2-c]pyrimidin-7-one hydrogen chloride (1.2 g, 2.57 mmol) was heated in THF (500 ml.) at the reflux temperature. 1-[(Dimethylamino)acetyl]-5- (methyloxy)-2,3-dihydro-1 H-indol-6-amine (0.640 g, 2.57 mmol) was added and the reaction mixture was heated at the reflux temperature for 2 h 20 min. The resulting mixture was allowed to cool to rt and concentrated. EtOAc (200 ml_), a few ml. THF and a saturated NaHCO3 solution (100 ml.) were added. The organic layer was washed with a saturated NaCI solution (100 ml_). The aqueous layers were back extracted with a mixture of THF (20 ml.) and EtOAc (100 ml_). The organic layers were combined, dried (Na2SO4) and concentrated and twice triturated using Et2O to obtain the crude product (1 195 mg). 319 mg of this crude mixture was taken up into a 27% aqueous NH4OH solution (50 ml.) and THF (50 ml.) and was maintained at rt for 18 h. EtOAc (100 ml.) and a saturated NaCI solution (50 ml.) were added. The organic layer was concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 followed by trituration using Et2O to obtain 4-({2-{[1-(Λ/,Λ/- dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-3-thiophenecarboxamide as a white solid (75 mg). 1H NMR (400 MHz, DMSO-d6) δ ppm 2.26 (s, 6H), 2.34 (s, 3H), 3.10-3.28 (m, 4H), 3.75 (s, 3H), 4.24 (d, J=8.42 Hz, 2H), 6.41 (d, J=3.66 Hz, 1 H), 7.08 (s, 1 H), 7.25-7.39 (m, 3H), 7.66 (br s, 1 H), 7.88-8.10 (m, 3H), 8.20 (br s, 1 H), 8.25 (s, 1 H), 8.31 (d, J=3.1 1 Hz, 1 H), 8.39 (s, 1 H), 11.31 (s, 1 H): ESIMS (M+H)+ = 661.28.
Step B/Example 1 1 : 4-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-3-thiophenecarboxamide
A mixture of 4-({2-{[1-(N,N-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-3- thiophenecarboxamide (49 mg, 0.074 mmol), dioxane (5 ml.) and a 1 M aqueous KOH solution (0.7 ml.) was heated at 80 0C for 8 h and was allowed to cool to rt. EtOAc (100 ml.) was added, followed by a saturated NaHCO3 solution (50 ml_). The organic layer was washed with a saturated NaCI solution (50 ml_), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to obtain 4-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}- 1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-3-thiophenecarboxamide as a beige solid (15.1 mg, 40%). 1H NMR (400 MHz, DMSOd6) δ ppm 2.26 (s, 6H), 3.04-3.26 (m, 4H), 3.79 (s, 3H), 4.19 (t, J=8.15 Hz, 2H), 6.20 (br s, 1 H), 6.39 (br s, 1 H), 6.97 (s, 1 H), 7.63 (br s, 1 H), 7.71 (s, 1 H), 8.1 1-8.27 (m, 2H), 8.32 (d, J=3.11 , 1 H), 8.62 (s, 1 H), 1 1.16 (s, 1 H), 11.24 (s, 1 H); ESIMS (M+H)+ = 507.18.
Example 12
4-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino}-1H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-3-thiophenecarboxamide
Figure imgf000181_0001
131 mg of the crude intermediate from the synthesis of 4-({2-{[1-(Λ/,Λ/-dimethylglycyl)-5- (methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}-7-[(4-methylphenyl)sulfonyl]-7/-/- pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-3-thiophenecarboxamide was dissolved in a solution of methylamine (2N in THF, 10 ml.) and stirred at rt. After 20 h the resulting mixture was concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2Cl2. The crude product was dissolved in dioxane (10 ml.) and a 1 N aqueous KOH solution (0.845 ml_, 0.845 mmol) and was heated at 80 0C. After 2.5 h the resulting mixture was allowed to cool to rt. EtOAc (100 ml.) and a saturated NaHCO3 solution (50 ml.) was added. The organic layer was washed with a saturated NaCI solution (50 ml_), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to obtain 4-[(2-{[1- (Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}-1 /-/-pyrrolo[2,3- c/]pyrimidin-4-yl)amino]-Λ/-methyl-3-thiophenecarboxamide as a brown solid (36.1 mg, 82%). 1H NMR (400 MHz, DMSO-d6) δ ppm 2.27 (s, 6H), 2.83 (d, J=4.4 Hz, 3H), 3.05- 3.27 (m, 4H), 3.79 (s, 3H), 4.18 (t, J=8.24 Hz, 2H), 6.23 (s, 1 H), 6.85-7.05 (m, 2H), 7.71 (s, 1 H), 8.23 (s, 2H), 8.57-8.78 (m, 2H), 1 1.09 (s, 1 H), 11.25 (s, 1 H); ESIMS (M+H)+ = 521.21.
Example 13 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino}-1H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-furancarboxamide
Figure imgf000182_0001
A solution of 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4- yl}amino)-2-furancarboxylic acid (0.66 g, 1.525 mmol) in THF (50 ml.) was treated with 2 drops of DMF followed by an oxalyl chloride solution (2N in CH2CI2, 4.5 ml_, 9 mmol mmol). After 30 min the reaction mixture was concentrated. The residue was dissolved in THF and concentrated again to obtain a beige solid (650 mg). 500 mg of the crude intermediate was taken up into THF (50 ml.) and treated with 1-[(dimethylamino)acetyl]- 5-(methyloxy)-2,3-dihydro-1 H-indol-6-amine (0.404 g, 1.620 mmol). The reaction mixture was heated at 70 0C for 17 h and was allowed to cool to rt. EtOAc (200 ml.) was added, followed by a saturated NaHCO3 solution (100 ml_). The organic layer was washed with a saturated NaCI solution (100 ml_), dried (Na2SO4) and concentrated. The resulting brown oily residue was triturated with Et2O. The crude intermediate was taken up into 2,2,2-trifluoroethanol (50 ml.) and was treated with K2CO3 (550 mg, 3.98 mmol). After 24 h a 27% aqueous NH4OH solution (50 ml.) was added, followed by addition of 2,2,2- trifluoroethanol (20 ml_). The reaction mixture was stirred at rt for 20 h. The resulting slurry was filtered. The crude intermediate was dissolved in dioxane (10 ml.) and a 1 N aqueous KOH solution (2.414 ml_, 2.414 mmol) and was heated at 90 0C. After 5.5 h the resulting mixture was allowed to cool to rt. EtOAc (100 ml.) and a saturated NaHCO3 solution (50 ml.) were added. The organic layer was washed with a saturated NaCI solution (100 ml_), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to obtain 3-[(2-{[1-(Λ/,Λ/- dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}-1 /-/-pyrrolo[2,3- c/]pyrimidin-4-yl)amino]-2-furancarboxamide (39 mg, 33%). 1H NMR (400 MHz, DMSO- d6) δ ppm 2.25 (s, 6H), 3.02-3.25 (m, 4H), 3.78 (s, 3H), 4.18 (t, J=8.15Hz, 2H), 6.13-6.28 (m, 1 H), 6.86-7.05 (m, 2H), 7.33-7.87 (m, 5H), 8.57 (s, 1 H), 9.69 (s, 1 H), 1 1.29 (s, 1 H); ESIMS (M+H)+ = 491.16.
Example 14
3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-
1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-furancarboxamide
Figure imgf000183_0001
A suspension of 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4- yl}amino)-2-furancarboxylic acid (1.15 g, 2.66 mmol) in THF (100 mL) was treated with a drop of DMF and oxalyl chloride (2.66 mL, 5.31 mmol). The resulting yellow opaque mixture was stirred at rt for 3 days and concentrated. The residue was taken up into benzene and concentrated again. The derived residue was taken up into THF (50 mL) and treated with 1-[(dimethylamino)acetyl]-6-(methyloxy)-1 ,2,3,4-tetrahydro-7- quinolinamine (447 mg, 1.698 mmol). The reaction mixture was heated at 80 0C for 3 h. The resulting orange slurry was allowed to cool to rt and treated with a 27% aqueous NH4OH solution (200 mL). The reaction mixture was heated in a sealed vessel at 80 0C for 22 h. The resulting mixture was allowed to cool to rt and diluted with EtOAc (100 mL). The organic layer was washed with a saturated NaCI solution (3x100 mL), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2, followed by trituration using a mixture of THF and Et2O. The derived solids were taken up into dioxane (10 mL) and a 1 N aqueous KOH solution (3.02 mL, 3.02 mmol) and was heated at 80 0C for 11 h. The resulting mixture was allowed to cool to rt. EtOAc (50 mL) was then added. The organic layer was washed with a saturated NaCI solution (2x50 ml_), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2Cl2, followed by trituration using Et2O to obtain 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-6-(methyloxy)-1 , 2,3,4- tetrahydro-7-quinolinyl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2- furancarboxamide as a pale yellow solid (120 mg). 1H NMR (400 MHz, THF-c/8) δ ppm 1.77-1.98 (m, 2H), 2.12 (br s, 6H), 2.70 (br s, 2H), 3.26 (s, 2H), 3.62-3.78 (s, 2H), 3.85 (s, 3H), 6.24 (br s, 1 H), 6.84 (br s, 1 H), 7.00 (br s, 1 H), 7.38-7.90 (s, 5H), 8.36 (br s, 1 H), 9.71 (s, 1 H), 11.41 (br s, 1 H); ESIMS (M+H)+ = 505.21.
Example 15
3-[(2-{[2-(methyloxy)-4-(1 -propyl-4-piperidinyl)phenyl]amino}-1H-pyrrolo[2,3- c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000184_0001
A solution of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (400 mg, 0.928 mmol) and [2- (methyloxy)-4-(1-propyl-4-piperidinyl)phenyl]amine (277 mg, 1.1 14 mmol) in 2,2,2- trifluoroethanol is heated to 9O0C in a sealed pressure tube for 45 minutes. The reaction was cooled and poured into DCM/saturated sodium bicarbonate and the organic layer was taken to dark residue under reduced pressure. Trituration of the residue with diethyl ether affords 5-{[2-(methyloxy)-4-(1 -propyl-4-piperidinyl)phenyl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (245 mg, 0.381 mmol, 41.1 % yield) as a pale yellow solid. A solution of 5-{[2- (methyloxy)-4-(1-propyl-4-piperidinyl)phenyl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (245 mg, 0.381 mmol) in THF and 27% ammonium hydroxide (8OmL total) was stirred at room temperature for 72 hrs. Essentially no reaction was observed by LCMS after this time. The reaction was poured into a pressure vessel and heated at 8O0C overnight. The reaction was cooled, poured into ethyl acetate/brine, and the organic layer was dried over sodium sulfate, filtered and taken to a residue under reduced pressure to afford 3-({2-{[2-(methyloxy)-4-(1-propyl-4-piperidinyl)phenyl]amino}-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2-thiophenecarboxamide (215 mg, 0.244 mmol, 64.1 % yield) as a pale yellow solid (approximately 70% purity.) A solution of crude 3-({2-{[2-(methyloxy)-4-(1-propyl-4-piperidinyl)phenyl]amino}-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2-thiophenecarboxamide (215 mg, 0.244 mmol) and sodium methoxide (132 mg, 2.444 mmol) in THF/MeOH was warmed to 8O0C and maintained for three hours. The reaction was poured into ethyl acetate/2. ON sodium hydroxide and the organic layer was dried over sodium sulfate, filtered, stripped onto celite, and purified by column chromatography to afford 3-[(2-{[2- (methyloxy)-4-(1-propyl-4-piperidinyl)phenyl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4- yl)amino]-2-thiophenecarboxamide (77 mg, 0.152 mmol, 62.3 % yield) as a pale white solid. 1H NMR (400 MHz, DMSOd6) δ ppm 1 1.40 (s, 1 H), 11.38 (s, 1 H), 8.41 (d, J=5.50 Hz, 1 H), 8.12 (d, J=8.07 Hz, 1 H), 7.70 (d, J=5.50 Hz, 1 H), 7.58 (s, 1 H), 7.57 (bs, 2 H), 6.98 (dd, J=3.67, 2.20 Hz, 1 H), 6.89 (d, J=1.83 Hz, 1 H), 6.79 (dd, J=8.07, 1.83 Hz, 1 H), 6.23 (dd, J=3.67, 1.83 Hz, 1 H), 3.85 (s, 3 H), 2.95 (d, J=1 1.00 Hz, 2 H), 2.38 - 2.46 (m, 1 H), 2.18 - 2.31 (m, 2 H), 1.85 - 2.02 (m, 2 H), 1.60 - 1.79 (m, 4 H), 1.36 - 1.51 (m, 2 H), 0.86 (t, J=7.33 Hz, 3 H). ESIMS (M+H)+ = 506.
Example 16
3-[(2-{[5-methyl-2-(methyloxy)-4-(1 -propyl -4-piperidinyl)phenyl]amino}-1 H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000186_0001
A solution of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (400 mg, 0.928 mmol) and [5- methyl-2-(methyloxy)-4-(1-propyl-4-piperidinyl)phenyl]amine (292 mg, 1.1 14 mmol) in 2,2,2-trifluoroethanol was heated to 9O0C in a sealed pressure tube. After 45 minutes, the reaction was cooled and poured into dichloromethane/saturated sodium bicarbonate. The organic layer was taken to a residue under reduced pressure and triturated with diethyl ether to afford 5-{[5-methyl-2-(methyloxy)-4-(1-propyl-4-piperidinyl)phenyl]amino}- 3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)- one (375 mg, 0.571 mmol, 61.5 % yield) as a pale yellow solid. A solution of 5-{[5- methyl-2-(methyloxy)-4-(1-propyl-4-piperidinyl)phenyl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (275 mg, 0.419 mmol) in THF/27% ammonium hydroxide was stirred at room temperature over the weekend (ca 72 hours). Essentially no reaction observed by TLC or LCMS. The reaction was poured into a pressure flask and warmed to 8O0C for 2 hrs. TLC/LCMS retaken and indicates partial consumption of starting material (ca 50% completion). Flask was re-sealed and heated an additional 4 hours. The flask was cooled and the reaction poured into ethylacetate/saturated sodium bicarbonate. The organic layer was dried over sodium sulfate, filtered, and taken to a residue under reduced pressure to afford 3-({2-{[5-methyl-2-(methyloxy)-4-(1-propyl-4- piperidinyl)phenyl]amino}-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4- yl}amino)-2-thiophenecarboxamide (250 mg, 0.186 mmol, 44.3 % yield) as a pale yellow solid with roughly 50% purity. This residue and sodium methoxide (220 mg, 4.08 mmol) in THF/MeOH is warmed to 8O0C for three hours, cooled, and poured into EtO Ac/saturated sodium chloride (aq). The organic layer was dried over sodium sulfate, filtered, stripped onto celite, and purified by chromatography on silica gel to afford 3-[(2- {[5-methyl-2-(methyloxy)-4-(1-propyl-4-piperidinyl)phenyl]amino}-1 H-pyrrolo[2,3- d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (90 mg, 0.173 mmol, 85 % yield) as a pale grey solid. 1H NMR (400 MHz, DMSOd6) δ ppm 11.38 (s, 1 H), 11.34 (s, 1 H), 8.39 (d, J=5.13 Hz, 1 H), 7.91 (s, 1 H), 7.69 (d, J=5.13 Hz, 1 H), 7.56 (s, 2 H), 7.49 (s, 1 H), 6.93 - 7.03 (m, 1 H), 6.83 (s, 1 H), 6.23 (dd, J=3.48, 2.02 Hz, 1 H), 3.82 (s, 3 H), 2.97 (d, J=10.63 Hz, 2 H), 2.57 - 2.69 (m, 1 H), 2.24 - 2.28 (m, 2 H), 2.22 - 2.25 (m, 3 H), 1.91 - 2.04 (m, 2 H), 1.62 - 1.77 (m, 4 H), 1.39 - 1.53 (m, 2 H), 0.86 (t, J=7.33 Hz, 3 H). ESIMS (M+H)+ = 520.
Example 17
3-({2-[(2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1-piperidinyl}phenyl)amino]-1H- pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2-thiophenecarboxamide
Figure imgf000187_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (400 mg, 0.928 mmol) and (2- (methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1-piperidinyl}phenyl)amine
2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1-piperidinyl}aniline (334 mg, 1.068 mmol) in 2,2,2-trifluoroethanol was warmed to 8O0C and maintained for 45 minutes. The reaction was cooled, poured into methylene chloride/saturated sodium bicarbonate, and the organic layer was taken to a residue under reduced pressure and solids triturated with diethylether to afford 5-[(2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1- piperidinyl}phenyl)amino]-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (515 mg, 0.729 mmol, 78 % yield) as a yellow solid. A suspension of 5-[(2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1- piperidinyl}phenyl)amino]-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (515 mg, 0.729 mmol) in THF/27% ammonium hydroxide (5OmL each) was warmed at 8O0C for 5.5 hours. The reaction was diluted further with THF/27% ammonium hydroxide (75ml_ more of each) and heated for an additional 16 hours at 9O0C in a pressure vessel. The reaction was cooled and layers separated. The organic layer was taken to a residue under reduced pressure and 3-({2- [(2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1-piperidinyl}phenyl)amino]-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2-thiophenecarboxamide (400 mg, 0.359 mmol, 49.3 % yield) was isolated as a yellow solid of sufficient purity for direct use in the next step. A solution of 3-({2-[(2-(methyloxy)-4-{4-[2- (methylsulfonyl)ethyl]-1-piperidinyl}phenyl)amino]-7-[(4-methylphenyl)sulfonyl]-7H- pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2-thiophenecarboxamide (400 mg, 0.553 mmol) in dioxane/2.0N NaOH was heated in a uW vial at 12O0C for 12 minutes. The solution was poured into THF/2.0N sodium hydroxide and the organic layer was washed with brine, dried over sodium sulfate, filtered, and stripped onto celite. Purification by chromatography on SiO2 afforded a number of mixed fractions, however upon sitting at O0C a thick white precipitate formed in 6 column fractions. Filtration of these fractions and drying afforded 3-({2-[(2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1- piperidinyl}phenyl)amino]-1 H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2- thiophenecarboxamide (85 mg, 0.149 mmol, 27.0 % yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 11.34 (s, 1 H), 11.24 - 1 1.31 (m, 1 H), 8.40 (d, J=5.50 Hz, 1 H), 7.82 (d, J=8.43 Hz, 1 H), 7.66 (d, J=5.50 Hz, 1 H), 7.54 (s, 2 H), 7.48 (s, 1 H), 6.88 - 6.98 (m, 1 H), 6.62 (d, J=2.57 Hz, 1 H), 6.49 (dd, J=8.98, 2.38 Hz, 1 H), 6.20 (dd, J=3.67, 1.83 Hz, 1 H), 3.80 (s, 3 H), 3.64 (d, J=12.46 Hz, 2 H), 3.1 1 - 3.20 (m, 2 H), 2.96 (s, 3 H), 2.54 - 2.65 (m, 1 H), 1.78 (d, J=12.46 Hz, 2 H), 1.61 - 1.71 (m, 2 H), 1.41 - 1.56 (m, 2 H), 1.20 - 1.36 (m, 2 H). ESIMS (M+H)+ = 570.
Example 18 3-({2-[(2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1 - piperidinyl}phenyl)amino]-1H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2- thiophenecarboxamide
Figure imgf000189_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (300 mg, 0.696 mmol) and (2- (methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1-piperidinyl}phenyl)amine (257 mg, 0.696 mmol) in 2,2,2-trifluoroethanol was heated at 9O0C for 72 hours. Many solids were observed in solution, but when poured into methylenechloride (ca 50 ml.) and washed with saturated sodium bicarbonate all solids dissolved. The organic layer was taken to a residue under reduced pressure and triturated with diethyl ether to afford 5-[(2- (methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1-piperidinyl}phenyl)amino]-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (307 mg, 0.402 mmol, 57.8 % yield) as a bright yellow solid. This procedure was reproduced to generate additional quantitites of this material. 5-[(2-(methyloxy)-4-{4-[4- (methylsulfonyl)-1-piperazinyl]-1-piperidinyl}phenyl)amino]-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (400 mg, 0.524 mmol) was suspended between THF (150 ml.) and 27% ammonium hydroxide (150 ml.) and heated in a pressure vial for 16 hours. The reaction was cooled, and the organic layer was washed with brine, dried over sodium sulfate, filtered, and taken to a residue under reduced pressure. The residue was suspended in dioxane (7 ml.) and 2.0N NaOH solution (aqueous, 7 ml.) was added. The resulting mixture was warmed to 12O0C for 12 minutes under microwave irradiation. The solution was cooled, poured into ethyl acetate/brine, and the organic layer was dried over sodium sulfate, filtered, stripped onto celite, and purified by column chromatography to afford 3-({2-[(2- (methyloxy)-4-{4-[4-(methylsulfonyl)-1 -piperazinyl]-1 -piperidinyl}phenyl)amino]-1 H- pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2-thiophenecarboxamide (145 mg, 0.232 mmol, 44.2 % yield) as a pale yellow solid. 1H NMR (400 MHz, DMSOd6) δ ppm 11.34 (s, 1 H), 11.28 (s, 1 H), 8.39 (d, J=5.50 Hz, 1 H), 7.81 (d, J=8.80 Hz, 1 H), 7.66 (d, J=5.13 Hz, 1 H), 7.54 (s, 2 H), 7.48 (s, 1 H), 6.89 - 6.98 (m, 1 H), 6.62 (d, J=2.20 Hz, 1 H), 6.49 (dd, J=8.80, 2.57 Hz, 1 H), 6.20 (dd, J=3.48, 2.02 Hz, 1 H), 3.79 (s, 3 H), 3.68 (d, J=12.10 Hz, 2 H), 3.01 - 3.14 (m, 4 H), 2.85 (s, 3 H), 2.55 - 2.72 (m, 6 H), 2.34 - 2.45 (m, 1 H), 1.84 (d, J=11.36 Hz, 2 H), 1.44 - 1.65 (m, 2 H). ESIMS (M+H)+ = 626.
Example 19
5-methyl-3-[(2-{[2-(methyloxy)-4-(1 -propyl -4-piperidinyl)phenyl]amino}-1 H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxylic acid
Figure imgf000190_0001
A suspension of 5-chloro-9-methyl-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (450 mg, 1.011 mmol) and [2- (methyloxy)-4-(1-propyl-4-piperidinyl)phenyl]amine (301 mg, 1.214 mmol) in 2,2,2- trifluoroethanol was heated at 9O0C in a sealed pressure tube for 50 minutes. The reaction was cooled, and poured into CH2CI2/saturated sodium bicarbonate. The organic layer was taken to a residue under reduced pressure and triturated with diethyl ether to afford 9-methyl-5-{[2-(methyloxy)-4-(1-propyl-4-piperidinyl)phenyl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (356 mg, 0.542 mmol, 53.6 % yield) as a bright yellow solid. This protocol has been repeated and allowed generation of larger quantities of 9-methyl-5-{[2-(methyloxy)-4-(1- propyl-4-piperidinyl)phenyl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one. A solution of 9-methyl-5-{[2-(methyloxy)-4-(1-propyl-4-piperidinyl)phenyl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (305 mg, 0.464 mmol) and sodium amide (181 mg, 4.64 mmol) in tetrahydrofuran (25 ml) was stirred for 4 hours at room temperature. The reaction was diluted with saturated sodium bicarbonate and ethyl acetate, and the organic layer was dried over sodium sulfate and taken to a residue under reduced pressure. The residue was dissolved in 1 ,4-dioxane/2.0N sodium hydroxide and heated at 12O0C in a microwave reactor for 12 minutes. The reaction was cooled and poured into ethyl acetate/saturated sodium chloride. The organic layer was dried over sodium sulfate, filtered, stripped onto celite, and purified by column chromatography (required addition of AcOH to mobile phase for product to elute) to afford 5-methyl-3-[(2-{[2-(methyloxy)-4-(1-propyl-4- piperidinyl)phenyl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2-thiophenecarboxylic acid (32 mg, 0.061 mmol, 13.24 % yield) as a white solid. Many exchangeable protons absent/obstructed by water/DMSO: 1H NMR (400 MHz, DMSOd6) δ ppm 11.25 (s, 1 H), 8.05 (d, J=8.07 Hz, 1 H), 7.99 (s, 1 H), 7.44 (s, 1 H), 6.90 - 6.99 (m, 1 H), 6.87 (d, J=1.47 Hz, 1 H), 6.79 (d, J=9.90 Hz, 1 H), 6.23 (dd, J=3.48, 2.02 Hz, 1 H), 3.82 (s, 3 H), 2.58 - 2.74 (m, 2 H), 2.36 (s, 2 H), 1.86 (s, 2 H), 1.54 - 1.71 (m, 2 H), 0.90 (t, J=7.33 Hz, 3 H). ESIMS (M+H)+ = 521.
Example 20
5-methyl-3-[(2-{[2-(methyloxy)-4-(1 -propyl -4-piperidinyl)phenyl]amino}-1 H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000192_0001
A suspension of 9-methyl-5-{[2-(methyloxy)-4-(1-propyl-4-piperidinyl)phenyl]amino}-3- [(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)- one HCI (305 mg, 0.464 mmol) in THF (100 ml_)/27% ammonium hydroxide (100 ml.) was heated in a sealed pressure vial at 9O0C for 16 hours. Analysis by LCMS indicates only ca 10% conversion to the desired product, so the reaction was heated an additional 24 hours, at which time LCMS indicates ca 50% conversion to the desired product. The reaction was poured into THF/brine, and the organic layer was concentrated to a residue under reduced pressure, redissolved in THF (ca 20 mL) and added to a pressure vessel with fresh 27% ammonium hydroxide (200 mL) and heated for 24 hours at 9O0C. Following cooling, LCMS analysis indicated >90% consumption of starting material. The reaction was poured into ethyl acetate/ saturated sodium chloride and the organic phase was dried over sodium sulfate, filtered, and taken to a residue under reduced pressure. The residue was dissolved in dioxane (8 mL) and added to a uW vial along with 2.0N NaOH (aq, 8mL). The mixture was heated at 12O0C for 12 minutes, cooled, and poured into ethyl acetate/2. ON sodium hydroxide. The organic layer was washed with saturated aq. brine, dried over sodium sulfate, stripped onto celite, and purified by flash column chromatography to afford 5-methyl-3-[(2-{[2-(methyloxy)-4-(1-propyl-4- piperidinyl)phenyl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2- thiophenecarboxamide (92 mg, 0.177 mmol, 38% yield) as a pale yellow solid. Several protons obstructed by DMSO: 1H NMR (400 MHz, DMSO-d6) δ ppm 1 1.43 (s, 1 H), 11.35 (s, 1 H), 8.16 (s, 1 H), 8.00 (d, J=8.43 Hz, 1 H), 7.57 (s, 1 H), 7.41 (s, 2 H), 6.94 - 7.00 (m, 1 H), 6.90 (d, J=1.83 Hz, 1 H), 6.81 (dd, J=8.25, 1.65 Hz, 1 H), 6.21 (dd, J=3.48, 2.02 Hz, 1 H), 3.84 (s, 3 H), 2.95 (d, J= 1 1.00 Hz, 2 H), 2.16 - 2.29 (m, 2 H), 1.87 - 2.01 (m, 2 H), 1.57 - 1.82 (m, 4 H), 1.34 - 1.51 (m, 2 H), 0.85 (t, J=7.33 Hz, 3 H). ESIMS (M+H)+ = 520.
Example 21
3-[(2-{[5-fluoro-4-[4-(1 -methylethyl)-1 -piperazinyl]-2-(methyloxy)phenyl]amino}-1 H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000193_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (500 mg, 1.160 mmol) and [5- fluoro-4-[4-(1-methylethyl)-1-piperazinyl]-2-(methyloxy)phenyl]amine (310 mg, 1.160 mmol) in 2,2,2-trifluoroethanol (8 ml) was stirred at 9O0C in a pressure tube until all solids had dissolved (45 minutes). The vial was cooled, and the mixture poured into dichloromethane/saturated sodium bicarbonate. The organic layer was taken to a residue under reduced pressure. Trituration with diethyl ether affords 5-{[5-fluoro-4-[4- (1-methylethyl)-1-piperazinyl]-2-(methyloxy)phenyl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (535 mg, 0.808 mmol, 69.7 % yield) as a pale yellow solid. A suspension of 5-{[5-fluoro- 4-[4-(1-methylethyl)-1-piperazinyl]-2-(methyloxy)phenyl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (535 mg, 0.808 mmol) in THF/27% ammonium hydroxide was stirred at 9O0C in a pressure vessel for 16 hours. The reaction was cooled, diluted with ethyl acetate/THF/brine, and the organic layer was washed with additional brine, dried over sodium sulfate, filtered, and taken to a residue under reduced pressure. The resulting solids are suspended in 1 ,4-dioxane/2.0N sodium hydroxide, added to a uW vial, and warmed at 12O0C for 12 minutes. The resulting biphasic solution was poured into ethyl acetate/THF/brine, and the organic layer was dried over sodium sulfate, filtered, stripped onto celite, and purified by chromatography on SiO2 to afford 3-[(2-{[5-fluoro-4-[4-(1- methylethyl)-1-piperazinyl]-2-(methyloxy)phenyl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4- yl)amino]-2-thiophenecarboxamide (97 mg, 0.185 mmol, 22.87 % yield) as a pale yellow solid. 1H NMR (400 MHz, DMSOd6) δ ppm 1 1.46 (s, 1 H), 11.40 (s, 1 H), 8.43 (d, J=5.50 Hz, 1 H), 8.18 (d, J=14.66 Hz, 1 H), 7.71 (d, J=5.50 Hz, 1 H), 7.60 (s, 1 H), 7.56 (s, 2 H), 6.95 - 7.05 (m, 1 H), 6.68 (d, J=8.07 Hz, 1 H), 6.25 (dd, J=3.67, 1.83 Hz, 1 H), 3.85 (s, 3 H), 2.92 - 3.04 (m, 4 H), 2.62 - 2.75 (m, 1 H), 2.54 - 2.61 (m, 4 H), 1.00 (d, J=6.60 Hz, 6 H). ESIMS (M+H)+ = 525.
Example 22
3-[(2-{[5-chloro-4-[4-(1 -methylethyl)-1 -piperazinyl]-2-(methyloxy)phenyl]amino}-1 H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000194_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (381 mg, 0.884 mmol) and [5- chloro-4-[4-(1-methylethyl)-1-piperazinyl]-2-(methyloxy)phenyl]amine (228 mg, 0.803 mmol) in 2,2,2-trifluoroethanol (8 ml) was warmed in a sealed tube for 55 minutes at 9O0C. The solution was cooled and poured into dichloromethane/saturated sodium bicarbonate. The organic layer was taken to a residue under reduced pressure and the solids triturated with diethyl ether to afford 5-{[5-chloro-4-[4-(1-methylethyl)-1- piperazinyl]-2-(methyloxy)phenyl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (418 mg, 0.616 mmol, 77 % yield). A suspension of 5-{[5-chloro-4-[4-(1-methylethyl)-1-piperazinyl]-2- (methyloxy)phenyl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (418 mg, 0.616 mmol) in THF/27% ammonium hydroxide was warmed to 9O0C in a sealed pressure tube for 16 hours. The reaction was subsequently cooled and poured into EtOAc/THF/Brine. The organic layer was dried over sodium sulfate, filtered, and taken to a residue under reduced pressure. The solids were suspended in 1 ,4-dioxane/2.0N NaOH, added to a uW vial, and warmed at 12O0C for 12 minutes. The reaction was cooled, poured into EtOAc/THF/brine, and the organic layer was washed with additional 2.0N sodium hydroxide, dried over sodium sulfate, filtered, stripped directly onto celite, and purified by column chromatography to afford 3-[(2-{[5-chloro-4-[4-(1-methylethyl)-1-piperazinyl]-2-(methyloxy)phenyl]amino}- 1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (102 mg, 0.189 mmol, 30.6 % yield) as a pale yellow solid. 1H NMR (400 MHz, DMSOd6) δ ppm 1 1.45 (s, 1 H), 11.42 (s, 1 H), 8.40 (d, J=5.13 Hz, 1 H), 8.28 (s, 1 H), 7.70 (d, J=5.50 Hz, 1 H), 7.61 (s, 1 H), 7.57 (s, 2 H), 6.98 - 7.03 (m, 1 H), 6.82 (s, 1 H), 6.25 (dd, J=3.48, 2.02 Hz, 1 H), 3.87 (s, 3 H), 2.97 (s, 4 H), 2.63 - 2.74 (m, 1 H), 2.60 (s, 4 H), 1.00 (d, J=6.60 Hz, 6 H). ESIMS (M+H)+ = 541.
Example 23
3-[(2-{[5-methyl-4-[4-(1 -methylethyl)-1-piperazinyl]-2-(methyloxy)phenyl]amino}- 1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000196_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (500 mg, 1.070 mmol) and [5- methyl-4-[4-(1-methylethyl)-1-piperazinyl]-2-(methyloxy)phenyl]amine (423 mg, 1.605 mmol) in 2,2,2-trifluoroethanol (8 ml) was warmed at 8O0C for 45 minutes in a sealed pressure tube. The reaction was cooled and poured into methylene chloride/saturated sodium bicarbonate. The organic layer was taken to a residue under reduced pressure and the solids triturated with diethyl ether to afford 5-{[5-methyl-4-[4-(1-methylethyl)-1- piperazinyl]-2-(methyloxy)phenyl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (478 mg, 0.727 mmol, 67.9 % yield) as a bright yellow solid. A suspension of 5-{[5-methyl-4-[4-(1-methylethyl)-1-piperazinyl]- 2-(methyloxy)phenyl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (478 mg, 0.727 mmol) in THF/27% ammonium hydroxide was warmed to 8O0C in a sealed pressure vessel for 14 hours. The reaction was cooled and poured into ethyl acetate/saturated sodium chloride (aq). The organic layer was dried over sodium sulfate, filtered, taken to a residue under reduced pressure, dissolved in 1 ,4-dioxane (8 ml.) and added to a microwave vessel with 2.0N sodium hydroxide (8 ml_). The resulting mixture was heated at 12O0C for 12 minutes, cooled, and poured into ethyl acetate/THF/satu rated sodium chloride. The organic layer was dried over sodium sulfate, filtered, stripped onto celite, and purified by column chromatography to afford 3-[(2-{[5-methyl-4-[4-(1-methylethyl)-1-piperazinyl]-2- (methyloxy)phenyl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2- thiophenecarboxamide (75 mg, 0.144 mmol, 20 % yield) as a white solid. 1H NMR (400 MHz, DMSOd6) δ ppm 1 1.37 (s, 1 H), 1 1.31 (s, 1 H), 8.37 (d, J=5.50 Hz, 1 H), 7.84 (s, 1 H), 7.67 (d, J=5.50 Hz, 1 H), 7.56 (s, 2 H), 7.47 (s, 1 H), 6.91 - 7.01 (m, 1 H), 6.72 (s, 1 H), 6.21 (dd, J=3.48, 2.02 Hz, 1 H), 3.79 (s, 3 H), 2.78 - 2.88 (m, 4 H), 2.63 - 2.72 (m, 1 H), 2.53 - 2.63 (m, 4 H), 2.19 (s, 3 H), 1.00 (d, J=6.60 Hz, 6 H). ESIMS (M+H)+ = 521.
Example 24
3-[(2-{[5-(methyloxy)-1 -(1 -methyl-L-prolyl)-2,3-dihydro-1H-indol-6-yl]amino}-1H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000197_0001
A solution of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (500 mg, 1.070 mmol) and 5- (methyloxy)-1-(1-methyl-L-prolyl)-2,3-dihydro-1 H-indol-6-amine (368 mg, 1.337 mmol) in 2,2,2-trifluoroethanol (8 ml) was warmed at 9O0C in a sealed pressure vessel for 3 hours. The reaction was cooled and poured into saturated sodium bicarbonate/methylene chloride. The organic layer was taken to a residue under reduced pressure and triturated with diethyl ether, but the remaining 5-(methyloxy)-1-(1-methyl-L-prolyl)-2,3- dihydro-1 H-indol-6-amine (368 mg, 1.337 mmol) could not be removed in this manner following repeated attempts. The crude solids were stripped onto celite (from methylene chloride) and purified by chromatography on silica gel to afford 5-{[5-(methyloxy)-1-(1- methyl-L-prolyl)-2,3-dihydro-1 H-indol-6-yl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (530 mg, 0.791 mmol, 74.0 % yield) as a pale yellow solid. A solution of 5-{[5-(methyloxy)-1-(1-methyl-L-prolyl)-2,3-dihydro- 1 H-indol-6-yl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (530 mg, 0.791 mmol) in 27% ammonium hydroxide (aq) (100 ml)/Tetrahydrofuran (THF) (50 ml) was stirred at 9O0C in a sealed pressure flask for 14 hours. The flask was cooled, and its contents poured into ethyl acetate/saturated sodium chloride. The organic layer was dried over sodium sulfate, taken to a residue under reduced pressure, and reddissolved in tetrahydrofuran (15 ml.) and methanol (5 ml_). Sodium methoxide (427 mg, 7.91 mmol) was added and the resulting solution was stirred for 6 hours at 6O0C. The reaction was cooled and poured into ethyl acetate, and washed successively with 2.0N sodium hydroxide and saturated sodium chloride (aq). The organic layer was dried over sodium sulfate, filtered, stripped onto celite, and purified by column chromatography to afford 3-[(2-{[5- (methyloxy)-i -(1 -methyl-L-prolyl)-2,3-dihydro-1 H-indol-6-yl]amino}-1 H-pyrrolo[2,3- d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (125 mg, 0.235 mmol, 29.7 % yield) as a white solid. 1H NMR (400 MHz, DMSOd6) δ ppm 1 1.39 (s, 1 H), 1 1.28 (s, 1 H), 8.62 (s, 1 H), 8.38 (d, J=5.13 Hz, 1 H), 7.68 (s, 1 H), 7.59 (d, J=5.50 Hz, 1 H), 7.52 (s, 2 H), 6.89 - 6.99 (m, 2 H), 6.21 (dd, J=3.30, 1.83 Hz, 1 H), 4.04 - 4.30 (m, 2 H), 3.76 (s, 3 H), 3.08 - 3.24 (m, 3 H), 2.88 - 3.03 (m, 1 H), 2.24 (s, 3 H), 2.08 - 2.21 (m, 1 H), 1.67 - 1.83 (m, 4 H). ESIMS (M+H)+ = 533.
Example 25
3-[(2-{[5-(methyloxy)-1 -L-prolyl-2,3-dihydro-1H-indol-6-yl]amino}-1H-pyrrolo[2,3- c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000198_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrido[1 ,2-c]pyrimidin-7(3H)-one HCI (550 mg, 1.279 mmol) and1 ,1- dimethylethyl (2S)-2-{[6-amino-5-(methyloxy)-2,3-dihydro-1 H-indol-1-yl]carbonyl}-1- pyrrolidinecarboxylate (532 mg, 1.471 mmol) in 2,2,2-trifluoroethanol (8 ml) was warmed at 9O0C for 25 minutes. The solution was cooled, poured into DCM/saturated sodium bicarbonate and the organic layer dried over sodium sulfate, filtered, and taken to a residue under reduced pressure. The solids were dissolved in DCM (ca 25 ml.) and trifluoroacetic acid (0.986 ml, 12.79 mmol) was added. The solution was stirred for 3 hours and poured into saturated sodium bicarbonate. The organic layer was dried over sodium sulfate, taken to a residue under reduced pressure, and purified by chromatography to afford 5-{[5-(methyloxy)-1 -L-prolyl-2,3-dihydro-1 H-indol-6-yl]amino}- 3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrido[1 ,2-c]pyrimidin-7(3H)-one (520 mg, 0.794 mmol, 62.1 % yield) as a bright yellow solid. A solution of 5-{[5- (methyloxy)-1-L-prolyl-2,3-dihydro-1 H-indol-6-yl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (518 mg, 0.790 mmol) in tetrahydrofuran (50 ml)/27% ammonium hydroxide (aq) (100 ml) was maintained at 8O0C in a sealed pressure flask for 16 hours. The reaction was cooled, poured into ethyl acetate/saturated sodium chloride, and the organic layer was dried over sodium sulfate, filtered, and taken to a residue under reduced pressure. The solids were dissolved in 1 ,4-Dioxane (8 ml) and added to a microwave vial containing 2.0N sodium hydroxide (aq) (8 ml). The mixture was heated in a microwave reactor at 12O0C for 12 minutes, cooled, and poured into TH F/ EtO Ac/2. ON sodium hydroxide. The organic layer was washed twice with saturated sodium chloride, dried over sodium sulfate, filtered, stripped onto celite, and purified by column chromatography to afford 3- [(2-{[5-(methyloxy)-1-L-prolyl-2,3-dihydro-1 H-indol-6-yl]amino}-1 H-pyrrolo[2,3- d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (110 mg, 0.212 mmol, 26.9 % yield) as a pale yellow solid. 1H NMR (400 MHz, DMSOd6) δ ppm 11.39 (s, 1 H), 11.30 (s, 1 H), 8.60 (s, 1 H), 8.39 (d, J=5.50 Hz, 1 H), 7.67 (s, 1 H), 7.59 (d, J=5.50 Hz, 1 H), 7.53 (s, 2 H), 6.90 - 6.98 (m, 2 H), 6.21 (dd, J=3.30, 1.83 Hz, 1 H), 4.17 - 4.32 (m, 1 H), 4.00 - 4.14 (m, 1 H), 3.83 (dd, J=8.07, 5.87 Hz, 1 H), 3.77 (s, 3 H), 3.06 - 3.20 (m, 2 H), 2.87 - 3.01 (m, 1 H), 2.60 - 2.73 (m, 1 H), 1.94 - 2.12 (m, 1 H), 1.54 - 1.76 (m, 4 H). ESIMS (M+H)+ = 519.
Example 26
3-{[2-({2-(ethyloxy)-4-[4-(1 -methylethyl)-1 -piperazinyl]phenyl}amino)-1 H- pyrrolo[2,3-c/]pyrimidin-4-yl]amino}-2-thiophenecarboxamide
Figure imgf000200_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (550 mg, 1.177 mmol) and {2- (ethyloxy)-4-[4-(1-methylethyl)-1-piperazinyl]phenyl}amine (372 mg, 1.412 mmol) in 2,2,2-trifluoroethanol (8 ml) was warmed to 8O0C in a pressure tube for 1.5 hr. The solution was cooled and poured into dichloromethane/saturated sodium bicarbonate. The organic layer was taken to a residue under reduced pressure and triturated with diethyl ether to afford 5-({2-(ethyloxy)-4-[4-(1-methylethyl)-1-piperazinyl]phenyl}amino)- 3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)- one (556 mg, 0.845 mmol, 71.8 % yield) as a yellow solid. A suspension of 5-({2- (ethyloxy)-4-[4-(1-methylethyl)-1-piperazinyl]phenyl}amino)-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (0.556 g, 0.845 mmol) in tetrahydrofuran (50 ml)/27% ammonium hydroxide (aq) (100 ml) was maintained at 8O0C in a sealed pressure vessel for 16 hours. The solution was cooled, diluted with ethyl acetate, and the organic layer was washed with saturated sodium chloride, dried over sodium sulfate, filtered, and taken to a residue under reduced pressure. The residue was redissolved in 1 ,4-dioxane (8 ml) and added to a microwave vial along with 2.0N NaOH (aq) (8 ml). The resulting mixture was heated at 12O0C for 12 minutes in a microwave reactor with rapid stirring, cooled, and poured into ethylacetate/2.0N sodium hydroxide. The organic layer was washed with saturated sodium chloride, dried over sodium sulfate, filtered, stripped onto celite, and purified by column chromatography to afford 3-{[2-({2-(ethyloxy)-4-[4-(1-methylethyl)-1- piperazinyl]phenyl}amino)-1 H-pyrrolo[2,3-d]pyrimidin-4-yl]amino}-2- thiophenecarboxamide (1 15 mg, 0.221 mmol, 26.1 % yield) as a pale yellow solid. 1H NMR (400 MHz, DMSOd6) δ ppm 11.31 (s, 1 H), 11.26 (s, 1 H), 8.33 (d, J=5.31 Hz, 1 H), 7.83 (d, J=8.79 Hz, 1 H), 7.64 (d, J=5.49 Hz, 1 H), 7.52 (s, 2 H), 7.39 (s, 1 H), 6.85 6.95 (m, 1 H), 6.58 (d, J=2.56 Hz, 1 H), 6.44 (dd, J=8.88, 2.29 Hz, 1 H), 6.18 (dd, J=3.30, 1.83 Hz, 1 H), 4.04 (q, J=6.96 Hz, 2 H), 2.97 - 3.08 (m, 4 H), 2.58 - 2.70 (m, 1 H), 2.48 - 2.58 (m, 4 H), 1.28 (t, J=6.96 Hz, 3 H), 0.97 (d, J=6.41 Hz, 6 H). ESIMS (M+H)+ = 521.
Example 27
3-({2-[(5-methyl-2-(methyloxy)-4-{1 -[2-(methylsulfonyl)ethyl]-4- piperidinyl}phenyl)amino]-1H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2- thiophenecarboxamide
Figure imgf000201_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (515 mg, 1.102 mmol) and (5- methyl-2-(methyloxy)-4-{1-[2-(methylsulfonyl)ethyl]-4-piperidinyl}phenyl)amine (414 mg, 1.267 mmol) in 2,2,2-trifluoroethanol (8 ml) was warmed in a sealed pressure tube at 9O0C for 55 minutes. The solution was cooled and poured into CH2Cl2/saturated sodium bicarbonate. The organic layer was taken to a residue under reduced pressure, triturated with diethyl ether, and the solids collected by vacuum filtration to afford 5-[(5- methyl-2-(methyloxy)-4-{1-[2-(methylsulfonyl)ethyl]-4-piperidinyl}phenyl)amino]-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (565 mg, 0.784 mmol, 71.1 % yield) as a bright yellow solid. A solution of 5-[(5-methyl- 2-(methyloxy)-4-{1-[2-(methylsulfonyl)ethyl]-4-piperidinyl}phenyl)amino]-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (565 mg, 0.784 mmol) in tetrahydrofuran (50 ml)/27% ammonium hydroxide (aq) (100 ml) was warmed to 9O0C in a sealed pressure tube for 16 hours. The tube was cooled, and the reaction poured into ethyl acetate/saturated sodium chloride. The organic layer was dried over sodium sulfate, filtered, and taken to a residue under reduced pressure. The residue was suspended in 1 ,4-dioxane (8 ml) and added to a microwave-safe vial along with 2.0N NaOH (aq) (8 ml). The mixture was warmed in a microwave reactor at 12O0C for 12 minutes, cooled, and poured into ethyl acetate/THF/2.0N sodium hydroxide. The organic layer was washed twice with saturated sodium chloride, dried over sodium sulfate, filtered, stripped onto celite, and purified by column chromatography to afford 3-({2-[(5-methyl-2-(methyloxy)-4-{1-[2-(methylsulfonyl)ethyl]-4- piperidinyl}phenyl)amino]-1 H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2- thiophenecarboxamide (85 mg, 0.146 mmol, 18.58 % yield) as a yellow solid. Several Protons obstructed by H2O or DMSO: 1H NMR (400 MHz, DMSO-c/6) δ ppm 1 1.35 (s, 1 H), 11.29 - 1 1.33 (m, 1 H), 8.35 (d, J=5.31 Hz, 1 H), 7.89 (s, 1 H), 7.66 (d, J=5.31 Hz, 1 H), 7.53 (s, 2 H), 7.47 (s, 1 H), 6.91 - 6.97 (m, 1 H), 6.78 (s, 1 H), 6.15 - 6.25 (m, 1 H), 3.78 (s, 3 H), 3.02 (s, 3 H), 2.99 (d, J=10.44 Hz, 2 H), 2.71 (t, J=6.87 Hz, 2 H), 2.55 - 2.66 (m, 1 H), 2.22 (s, 3 H), 1.98 - 2.15 (m, 2 H), 1.58 - 1.70 (m, 4 H). ESIMS (M+H)+ = 584.
Example 28
3-[(2-{[5-{[3-(dimethylamino)propyl]oxy}-2-(methyloxy)phenyl]amino}-1H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2 -thiophenecarboxamide
Figure imgf000203_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (600 mg, 1.284 mmol) and (3- {[3-amino-4-(methyloxy)phenyl]oxy}propyl)dimethylamine (432 mg, 1.926 mmol) in 2,2,2- trifluoroethanol (8 ml) was warmed to 8O0C for 3 hours, cooled, and poured into saturated sodium bicarbonate/dichloromethane. The organic layer was taken to a residue under reduced pressure and triturated with diethyl ether to afford 5-{[5-{[3- (dimethylamino)propyl]oxy}-2-(methyloxy)phenyl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (465 mg, 0.752 mmol, 58.5 % yield) with roughly 70% purity, sufficient for use in the next step. A solution of 5-{[5-{[3-(dimethylamino)propyl]oxy}-2-(methyloxy)phenyl]amino}-3- [(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)- one (465 mg, 0.752 mmol) in tetrahydrofuran (75 ml)/27% ammonium hydroxide (200 ml) was maintained at 8O0C in a sealed pressure vial for 16 hours. The reaction was allowed to cool, diluted with ethyl acetate, and washed twice with saturated sodium chloride. The organic layer was dried over sodium sulfate, filtered, stripped onto celite, and purified by column chromatography to afford 3-({2-{[5-{[3- (dimethylamino)propyl]oxy}-2-(methyloxy)phenyl]amino}-7-[(4-methylphenyl)sulfonyl]-7H- pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2-thiophenecarboxamide (1 14 mg, 0.179 mmol, 23.86 % yield) as a pale yellow solid with >90% purity. A solution of 3-({2-{[5-{[3- (dimethylamino)propyl]oxy}-2-(methyloxy)phenyl]amino}-7-[(4-methylphenyl)sulfonyl]-7H- pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2-thiophenecarboxamide (1 10 mg, 0.173 mmol) in 1 ,4-dioxane (8 ml) was treated with 2.0N sodium hydroxide (aq) (8.00 ml) and warmed under microwave heating at 12O0C for 12 minutes. LCMS at this stage indicates additional starting material remains, so the vial was resubjected to 12O0C in a microwave reactor for an additional 10 minutes. The solution was cooled and poured into ethyl acetate/THF/2.0N sodium hydroxide. The organic layer was washed with saturated sodium chloride, dried over sodium sulfate, filtered, stripped onto celite, and the residue purified by column chromotagraphy to afford 3-[(2-{[5-{[3-(dimethylamino)propyl]oxy}-2- (methyloxy)phenyl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2- thiophenecarboxamide (57 mg, 0.1 18 mmol, 68.4 % yield) as a pale yellow solid. 1H NMR (400 MHz, DMSOd6) δ ppm 11.49 (s, 1 H), 11.40 (s, 1 H), 8.39 (d, J=5.49 Hz, 1 H), 8.06 (d, J=2.93 Hz, 1 H), 7.69 (d, J=5.49 Hz, 1 H), 7.57 (s, 1 H), 7.54 (s, 2 H), 6.97 - 7.05 (m, 1 H), 6.86 (d, J=8.79 Hz, 1 H), 6.42 (dd, J=8.88, 3.02 Hz, 1 H), 6.24 (dd,
J=3.11 , 1.65 Hz, 1 H), 3.89 (t, J=6.41 Hz, 2 H), 3.79 (s, 3 H), 2.30 (t, J=IAA Hz, 2 H), 2.09 (s, 6 H), 1.69 - 1.87 (m, 2 H). ESIMS (M+H)+ = 482.
Example 29 3-[(2-{[1-[3-(dimethylamino)propyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000204_0001
A solution of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (600 mg, 1.284 mmol) and 1-[3- (dimethylamino)propyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-amine (320 mg, 1.284 mmol) in 2,2,2-trifluoroethanol (8 ml) was warmed to 8O0C in a sealed pressure tube for 3 hours. The solution was cooled and poured into saturated sodium bicarbonate/methylene chloride. The organic layer was taken to a residue under reduced pressure and triturated with diethyl ether but the solids remain impure. All solids and filtrates were combined, dissolved in methylene chloride, stripped onto celite, and purified by column chromatography to afford 5-{[1-[3-(dimethylamino)propyl]-5- (methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (560 mg, 0.870 mmol, 67.8 % yield) with >95% purity as a bright yellow solid. A solution of 5-{[1-[3-(dimethylamino)propyl]-5- (methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (560 mg, 0.870 mmol)in tetrahydrofuran (THF) (75 ml)/27% ammonium hydroxide (200 ml) was warmed in a sealed pressure tube for 16 hrs. at 8O0C. The reaction was cooled and poured into ethyl acetate/brine. The organic layer was washed with brine, dried over sodium sulfate, filtered, and taken to a residue under reduced pressure. The solids were dissolved in 1 ,4-dioxane (8 ml) and placed in a uW vial along with 2.0N sodium hydroxide (aq) (8 ml). The suspension was warmed at 12O0C for 12 minutes in a microwave reactor with rapid stirring, cooled, and poured into THF/EtOAc/2.0N sodium hydroxide. The organic layer was washed with brine, dried over sodium sulfate, filtered, stripped onto celite, and purified by flash column chromatography to afford3-[(2-{[1-[3-(dimethylamino)propyl]-5- (methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2- thiophenecarboxamide (41 mg, 0.081 mmol, 9.30 % yield) as a pale yellow solid. 1H NMR (400 MHz, DMSOd6) δ ppm 11.42 (s, 1 H), 11.33 - 1 1.39 (m, 1 H), 8.41 (d, J=5.42 Hz, 1 H), 7.69 (d, J=5.42 Hz, 1 H), 7.57 (s, 2 H), 7.50 (s, 2 H), 6.99 - 7.04 (m, 1 H), 6.84 (s, 1 H), 6.25 (s, 1 H), 3.75 (s, 3 H), 3.22 (t, J=8.13 Hz, 2 H), 2.91 - 2.99 (m, 2 H), 2.84 (t, J=7.12 Hz, 2 H), 2.25 (t, J=7.02 Hz, 2 H), 2.10 (s, 6 H), 1.56 - 1.75 (m, 2 H). ESIMS (M+H)+ = 507.
Example 30 3-({2-[(5-{[3-(dimethylamino)propyl]oxy}-2-fluorophenyl)amino]-1H-pyrrolo[2,3- c/]pyrimidin-4-yl}amino)-2-thiophenecarboxamide
Figure imgf000206_0001
A solution of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (600 mg, 1.284 mmol) and {3-[(3- amino-4-fluorophenyl)oxy]propyl}dimethylamine (409 mg, 1.926 mmol) in 2,2,2- trifluoroethanol (8 ml) was warmed at 8O0C for 3 hours, cooled, and poured into saturated sodium bicarbonate/methylene chloride. The organic layer was taken to a residue under reduced pressure and the solids were triturated with diethyl ether to afford 5-[(5-{[3-(dimethylamino)propyl]oxy}-2-fluorophenyl)amino]-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (565 mg, 0.931 mmol, 72.5 % yield) as a yellow solid. A solution of 5-[(5-{[3- (dimethylamino)propyl]oxy}-2 -fluorophenyl )amino]-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (565 mg, 0.931 mmol) in tetrahydrofuran (75 ml)/27% Ammonium Hydroxide (200 ml) was warmed in a sealed pressure tube for 16 hrs. at 8O0C. The reaction was cooled and poured into ethyl acetate/brine. The organic layer was washed with brine, dried over sodium sulfate, filtered, and taken to a residue under reduced pressure. The solids were dissolved in 1 ,4-dioxane (8 ml) and placed in a microwave-safe vial along with 2.0N sodium hydroxide (aq) (8 ml). The suspension was warmed at 12O0C for 12 minutes in a microwave reactor with rapid stirring, cooled, and poured into THF/EtOAc/2.0N sodium hydroxide. The organic layer was washed with brine, dried over sodium sulfate, filtered, stripped onto celite, and purified by flash column chromatography to afford 3-({2-[(5-{[3- (dimethylamino)propyl]oxy}-2-fluorophenyl)amino]-1 H-pyrrolo[2,3-d]pyrimidin-4- yl}amino)-2-thiophenecarboxamide (165 mg, 0.351 mmol, 37.7 % yield) as a pale yellow solid. 1H NMR (400 MHz, DMSOd6) δ ppm 1 1.44 (s, 2 H), 8.51 (d, J=5.49 Hz, 1 H), 8.40 (s, 1 H), 7.69 (dd, J=6.87, 3.02 Hz, 1 H), 7.62 (d, J=5.49 Hz, 1 H), 7.55 (s, 2 H), 7.07 (dd, J=10.71 , 9.06 Hz, 1 H), 6.96 - 7.01 (m, 1 H), 6.46 - 6.55 (m, 1 H), 6.22 (dd, J=3.30, 1.65 Hz, 1 H), 3.91 (t, J=6.41 Hz, 2 H), 2.30 (t, J=7.23 Hz, 2 H), 2.09 (s, 6 H), 1.72 - 1.87 (m, 2 H). ESIMS (M+H)+ = 470.
Example 31
3-[(2-{[1-(Λ/-ethyl-Λ/-methylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000207_0001
A solution of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (600 mg, 1.284 mmol) and 1- {[ethyl(methyl)amino]acetyl}-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinamine (356 mg, 1.284 mmol) in 2,2,2-trifluoroethanol (8 ml) was warmed to 8O0C in a sealed pressure tube for 3 hours. The solution was cooled, poured into saturated sodium bicarbonate, and diluted with methylene chloride. The organic layer was dried over sodium sulfate, filtered, taken to a residue under reduced pressure, and the resulting solids triturated with diethyl ether to afford 5-{[1-(N-ethyl-N-methylglycyl)-6-(methyloxy)-1 , 2,3,4- tetrahydro-7-quinolinyl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (605 mg, 0.901 mmol, 70% yield) as a pale yellow solid. A suspension of 5-{[1-(N-ethyl-N-methylglycyl)-6-(methyloxy)- 1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (605 mg, 0.901 mmol) in tetrahydrofuran (50 ml)/27% ammonium hydroxide (150 ml) was warmed to 9O0C in a sealed pressure tube for 16 hours. The solution was cooled and poured into ethyl acetate/saturated sodium chloride. The organic layer was dried over sodium sulfate, taken to a residue under reduced pressure, dissolved in 1 ,4-dioxane (8 ml), and added to a microwave-safe vial with 2.0N NaOH (aq) (8 ml). The vial was warmed in a microwave reactor for 12 minutes at 12O0C, cooled, and poured into saturated sodium bicarbonate/ethyl acetate/THF. The organic layer was dried over sodium sulfate, filtered, stripped onto celite, and purified by column chromatography to afford 3-[(2-{[1- (N-ethyl-N-methylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-1 H- pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (125 mg, 0.234 mmol, 26% yield) as a pale yellow solid. Several protons obstructed by DMSO/H2O: 1H NMR (400 MHz, DMSOd6) δ ppm 1 1.37 (s, 2 H), 8.35 (s, 1 H), 8.34 (s, 1 H), 7.63 (d, J=5.49 Hz, 1 H), 7.59 (s, 1 H), 7.53 (s, 2 H), 6.97 (s, 1 H), 6.80 (s, 1 H), 6.18 - 6.22 (m, 1 H), 3.80 (s, 3 H), 3.67 (t, J=5.22 Hz, 2 H), 2.65 (s, 2 H), 2.28 (s, 2 H), 2.07 (s, 3 H), 1.76 - 1.91 (m, 2 H), 0.75 (s, 3 H). ESIMS (M+H)+ = 535.
Example 32
3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-3,3-dimethyl-5-(methyloxy)-2,3-dihydro-1H-indol-6- yl]amino}-1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000208_0001
A solution of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (550 mg, 1.177 mmol) and 1- [(dimethylamino)acetyl]-3,3-dimethyl-5-(methyloxy)-2,3-dihydro-1 H-indol-6-amine (375 mg, 1.353 mmol) in THF (8 ml) was warmed in a sealed pressure tube for 16 hours. The reaction was cooled, concentrated, susupended in dichloromethane (ca 25 ml.) and washed with saturated sodium bicarbonate. The organic layer was taken to a residue under reduced pressure and triturated with diethyl ether to afford 5-{[1-(N,N- dimethylglycyl)-3,3-dimethyl-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (0.627 g, 0.933 mmol, 79 % yield) as a light yellow solid. A solution of 5-{[1-(N,N- dimethylglycyl)-3,3-dimethyl-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (0.620 g, 0.923 mmol) in THF (40 ml) and 27% ammonium hydroxide (100 ml) was warmed at 8O0C in a pressure tube for 16 hours. The reaction was cooled and poured into ethyl acetate/saturated sodium bicarbonate. The organic layer was dried over sodium sulfate, filtered, and taken to a residue under reduced pressure. The residue was dissolved in 1 ,4-dioxane (8 ml) and added to a microwave-safe vial along with 2.0N NaOH (aq) (40.0 ml). The contents were warmed at 12O0C for 12 minutes in a microwave reactor, cooled, and poured into ethyl acetate/THF/saturated sodium chloride. The organic layer was dried over sodium sulfate, filtered, stripped onto celite, and purified by column chromatography. The solids collected after concentration of the column fractions contained 1-[(dimethylamino)acetyl]-3,3-dimethyl-5-(methyloxy)-2,3- dihydro-1 H-indol-6-amine. All solids were triturated with methylenechloride/diethy ether to afford pure 3-[(2-{[1-(N,N-dimethylglycyl)-3,3-dimethyl-5-(methyloxy)-2,3-dihydro-1 H- indol-6-yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (0.096 g, 0.180 mmol, 19% yield) as a pale yellow solid. 1H NMR (400 MHz, DMSO-c/6) δ ppm 11.39 (s, 1 H), 11.28 (s, 1 H), 8.51 (s, 1 H), 8.37 (d, J=5.82 Hz, 1 H), 7.70 (s, 1 H), 7.57 (d, J=5.02 Hz, 1 H), 7.52 (s, 2 H), 6.97 (s, 1 H), 6.93 - 6.97 (m, 1 H), 6.22 (dd, J=3.51 , 1.50 Hz, 1 H), 3.94 (s, 2 H), 3.80 (s, 3 H), 3.17 (s, 2 H), 2.24 (s, 6 H), 1.33 (s, 6 H). ESIMS (M+H)+ = 535.
Example 33
3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-6-(ethyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-
1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000210_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (550 mg, 1.177 mmol) and 1- [(dimethylamino)acetyl]-6-(ethyloxy)-1 ,2,3,4-tetrahydro-7-quinolinamine (326 mg, 1.177 mmol) in THF (8 ml) was warmed in a pressure vessel at 750C for 16 hours. The volatiles were removed and the solids partitioned between dichloromethane and saturated sodium bicarbonate. The organic layer was taken to a residue under reduced pressure and solids triturated with diethyl ether to afford 5-{[1-(N,N-dimethylglycyl)-6- (ethyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (0.640 g, 0.953 mmol, 81 % yield) as a yellow solid. A solution of 5-{[1 -(N, N-dimethylglycyl)-6-(ethyloxy)-1 , 2,3,4- tetrahydro-7-quinolinyl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (640 mg, 0.953 mmol) in THF (50 ml)/27% ammonium hydroxide (100 ml) was maintained at 8O0C in a sealed pressure flask for 16 hours. The reaction was cooled, diluted with ethyl acetate/THF/satu rated sodium chloride (aq) and the organic layer was dried over sodium sulfate and taken to a residue under reduced pressure. The residue was dissolved in 1 ,4-dioxane (8 ml), 2.0N NaOH (aq) (8 ml) was added, and the mixture was stirred rapidly under microwave heating at 12O0C for 12 minutes. The reaction was cooled and poured into ethyl acetate/saturated sodium bicarbonate. The organic layer was dried over sodium sulfate, filtered, stripped onto celite, and purified by column chromatograpy to afford 3-[(2-{[1- (N,N-dimethylglycyl)-6-(ethyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-1 H-pyrrolo[2,3- d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (0.075 g, 0.140 mmol, 14.73 % yield) as a pale yellow solid. 1H NMR (400 MHz, DMSOd6) δ ppm 1 1.41 (s, 2 H), 8.39 (s, 1
H), 8.34 (d, J=5.42 Hz, 1 H), 7.67 (d, J=5.42 Hz, 1 H), 7.54 - 7.56 (m, 1 H), 7.55 (s, 2 H), 6.95 - 7.06 (m, 1 H), 6.83 (s, 1 H), 6.26 (s, 1 H), 4.10 (q, J=7.02 Hz, 2 H), 3.64 - 3.77 (m, 2 H), 3.23 (s, 2 H), 2.62 - 2.73 (m, 2 H), 2.54 (s, 6 H), 1.79 - 1.94 (m, 2 H), 1.37 (t, J=6.82 Hz, 3 H). ESIMS (M+H)+ = 535.
Example 34 3-[(2-{[5-(methyloxy)-1 -(1 -methyl-D-prolyl)-2,3-dihydro-1 H-indol-6-yl]amino}-1 H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000211_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (600 mg, 1.284 mmol) and 5- (methyloxy)-1-(1-methyl-D-prolyl)-2,3-dihydro-1 H-indol-6-amine (353 mg, 1.284 mmol) in 2,2,2-trifluoroethanol (50 ml) was warmed to 8O0C and maintained for 2 hours. The solution was cooled, poured into dichloromethane, and washed with saturated sodium bicarbonate. The organic layer was taken to a residue under reduced pressure and purified via trituration with diethyl ether to afford 5-{[5-(methyloxy)-1-(1-methyl-D-prolyl)- 2,3-dihydro-1 H-indol-6-yl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (0.410 g, 0.612 mmol, 48% yield) as a dark yellow solid. A solution of 5-{[5-(methyloxy)-1-(1-methyl-D-prolyl)-2,3-dihydro-1 H- indol-6-yl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2- c]pyrimidin-7(3H)-one (410 mg, 0.612 mmol) in tetrahydrofuran (50 ml)/27% ammonium hydroxide (50.0 ml) was maintained at 8O0C in a pressure vessel for 16 hours. The vessel was cooled, diluted with tetrahydrofuran/ethyl acetate/saturated sodium chloride, and the organic layer was washed with additional water. The organic layer was dried over sodium sulfate, filtered, taken to a residue under reduced pressure, and resuspended in 1 ,4-dioxane (8 ml)/2.0N NaOH (aq) (8 ml). The suspension was warmed at 12O0C for 12 minutes in a microwave reactor, cooled, and poured into ethyl acetate/tetrahydrofuran, saturated sodium chloride (aq). The organic layer was dried over sodium sulfate, filtered, stripped onto celite, and purified by column chromatography to afford 3-[(2-{[5-(methyloxy)-1-(1-methyl-D-prolyl)-2,3-dihydro-1 H- indol-6-yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (0.090 g, 0.169 mmol, 28% yield) as a white solid. 1H NMR (400 MHz, DMSO-c/6) δ ppm 11.40 (s, 1 H), 11.29 (s, 1 H), 8.63 (s, 1 H), 8.39 (d, J=5.42 Hz, 1 H), 7.68 (s, 1 H), 7.60 (d, J=5.22 Hz, 1 H), 7.54 (s, 2 H), 6.93 - 6.99 (m, 2 H), 6.23 (s, 1 H), 4.09 - 4.26 (m, 2 H), 3.78 (s, 3 H), 3.18 - 3.24 (m, 1 H), 3.09 - 3.17 (m, 2 H), 2.93 - 3.02 (m, 1 H), 2.25 (s, 3 H), 2.1 1 - 2.21 (m, 1 H), 1.66 - 1.85 (m, 4 H). ESIMS (M+H)+ = 533.
Example 35
3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-2,3-dihydro-1H-indol-6-yl]amino}-1H-pyrrolo[2,3- c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000212_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (600 mg, 1.284 mmol) and 1- [(dimethylamino)acetyl]-2,3-dihydro-1 H-indol-6-amine (282 mg, 1.284 mmol) was maintained at 7O0C in tetrahydrofuran (20 ml) in a pressure tube for 16 hours. The reaction was cooled, opened, concentrated under reduced pressure, and redissolved in methylene chloride/2, 2, 2-trifluoroethanol (ca 15 ml_). Saturated sodium bicarbonate was added and all solids dissolve. The organic layer was dried over sodium sulfate, filtered, taken to a residue under reduced pressure, and triturated with diethyl ether to afford 5- {[1-(N,N-dimethylglycyl)-2,3-dihydro-1 H-indol-6-yl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (0.600 g, 0.978 mmol, 76 % yield) as a yellow solid. A solution of 5-{[1-(N,N- dimethylglycyl)-2,3-dihydro-1 H-indol-6-yl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (0.600 g, 0.978 mmol) in tetrahydrofuran (40 ml)/27% ammonium hydroxide (aq) (150 ml) was maintained at 8O0C in a sealed pressure tube for 12 hours. The solution was cooled and poured into ethyl acetate/saturated sodium chloride (aq). The organic layer was dried over sodium sulfate, filtered, stripped onto celite, and purified by column chromatography to afford 3- ({2-{[1-(Λ/,Λ/-dimethylglycyl)-2,3-dihydro-1H-indol-6-yl]amino}-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2-thiophenecarboxamide of sufficient purifty for use in the next transformation. The solid was dissolved in 1 ,4- dioxane (8 ml)/2.0N NaOH (aq) (40.0 ml) and maintained at 12O0C for 12 minutes under microwave heating. The solution was cooled, poured into ethyl acetate/THF/saturated sodium chloride (aq) and the organic layer was dried over sodium sulfate, filtered, stripped onto celite, and purified by column chromatography to afford 3-[(2-{[1 -(N1N- dimethylglycyl)-2,3-dihydro-1 H-indol-6-yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]- 2-thiophenecarboxamide (0.075 g, 0.157 mmol, 16% yield) as a yellow solid. 1H NMR (400 MHz, DMSOd6) δ ppm 11.46 (s, 1 H), 11.34 - 1 1.39 (m, 1 H), 9.09 (s, 1 H), 8.74 (d, J=5.02 Hz, 1 H), 8.22 (s, 1 H), 7.88 (d, J=7.83 Hz, 1 H), 7.69 (d, J=5.42 Hz, 1 H), 7.55 (s, 2 H), 7.12 (d, J=8.23 Hz, 1 H), 6.99 (dd, J=3.31 , 2.51 Hz, 1 H), 6.24 (dd, J=3.51 , 1.91 Hz, 1 H), 4.16 (t, J=8.13 Hz, 2 H), 3.23 (s, 2 H), 3.07 (t, J=9.13 Hz, 2 H), 2.29 (s, 6 H). ESIMS (M+H)+ = 477.
Example 36
3-[(2-{[5-chloro-1-(Λ/,Λ/-dimethylglycyl)-2,3-dihydro-1H-indol-6-yl]amino}-1H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000213_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one HCI (800 mg, 1.712 mmol) and 5- chloro-1-[(dimethylamino)acetyl]-2,3-dihydro-1 H-indol-6-amine (499 mg, 1.969 mmol) in tetrahydrofuran (20 ml) was maintained at 8O0C for 24 hours. The solution was cooled, all solvents evaporated, and the solids were partioned between methylene chloride/trifluoroethanol/saturated sodium bicarbonate. The organic layer was dried over sodium sulfate, filtered, taken to a solid under reduced pressure, and the solids were triturated with diethyl ether to afford 5-{[5-chloro-1-(N,N-dimethylglycyl)-2,3-dihydro-1 H- indol-6-yl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2- c]pyrimidin-7(3H)-one (0.920 g, 1.419 mmol, 83 % yield) as a green-yellow solid. A solution of 5-{[5-chloro-1-(N,N-dimethylglycyl)-2,3-dihydro-1 H-indol-6-yl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (770 mg, 1.188 mmol) in tetrahydrofuran (40 ml)/27% ammonium hydroxide (aq) (150 ml) was maintained in a pressure tube at 8O0C for 12 hours. The solution was cooled, opened cautiously, and poured into ethyl acetate/saturated sodium chloride (aq). The organic layer was dried over sodium sulfate, taken to a residue under reduced pressure, dissolved in 1 ,4-dioxane (8 ml), and added to a microwave vial with 2.0N NaOH (aq) (40.0 ml). The vial was maintained under microwave heating at 12O0C for 12 minutes, cooled, poured into ethyl acetate/tetrahydrofuran/saturated sodium bicarbonate, and the organic layer was dried over sodium sulfate, filtered, stripped onto celite, and purified by column chromatography to afford 3-[(2-{[5-chloro-1-(N,N-dimethylglycyl)-2,3-dihydro-1 H- indol-6-yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (0.040 g, 0.078 mmol, 7% yield). 1H NMR (400 MHz, DMSOd6) δ ppm 11.42 (s, 1 H), 11.28 - 11.35 (m, 1 H), 8.37 (s, 1 H), 8.28 - 8.35 (m, 2 H), 7.56 - 7.60 (m, 1 H), 7.55 (s, 2 H), 7.35 (s, 1 H), 6.97 (s, 1 H), 6.23 (s, 1 H), 4.21 (t, J=8.63 Hz, 2 H), 3.29 (s, 2 H), 3.08 - 3.20 (m, 2 H), 2.28 (s, 6 H). ESIMS (M+H)+ = 511.
Example 37: Λ/-methyl-3-[(2-{[4-[4-(1 -methylethyl)-1 -piperazinyl]-2-(methyloxy)phenyl]amino}- 1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2- thiophenecarboxamide
Figure imgf000215_0001
Step A/Intermediate C7: 5-{[4-[4-(1-methylethyl)-1-piperazinyl]-2- (methyloxy)phenyl]amino}pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)- one
Figure imgf000215_0002
A slurry of 4-[4-(1-methylethyl)-1-piperazinyl]-2-(methyloxy)aniline (3.18 g, 12.76 mmol) and 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2- c]pyrimidin-7(3H)-one (5.5 g, 12.76 mmol) in 2,2,2-trifluoroethanol (200 ml.) was heated at 80 0C for 1.5 h, then was allowed to cool to rt. The resulting mixture was diluted with EtOAc (200 ml_), washed with a saturated NaHCOs solution (100 ml.) and a saturated NaCI solution (100 ml_). The aqueous layers were back extracted with EtOAc (200 ml_). The organic layers were combined and concentrated. The residue was triturated using Et2O to give 5-{[4-[4-(1-methylethyl)-1-piperazinyl]-2-
(methyloxy)phenyl]amino}pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)- one as a yellow solid. ESIMS (M+H)+ = 644.32.
Step B/Example 37: Λ/-methyl-3-[(2-{[4-[4-(1-methylethyl)-1-piperazinyl]-2- (methyloxy)phenyl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2- thiophenecarboxamide
A mixture of 5-{[4-[4-(1-methylethyl)-1-piperazinyl]-2-(methyloxy)phenyl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one (150 mg, 0.23mmol) and methylamine 2.0M/THF (1 1.65ml, 23.30 mmol)) in tetrahydrofuran (THF) (20ml) was stirred at rt for three days.. Next, the crude reaction was adsorbed on silica gel and purified by LC (DCM to 10% MeOH/DCM). ESIMS (M+H)+ = 675. The isolated amide was redissolved in 1 ,4-dioxane (10 ml), potassium hydroxide (8.72 ml, 8.72 mmol) was added and the reaction was heated in the microwave at 12O0C for 20 min. The resulting solution was adsorbed onto silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford Λ/-methyl-3-[(2-{[4-[4-(1-methylethyl)- 1-piperazinyl]-2-(methyloxy)phenyl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2- thiophenecarboxamide (0.035g, 30% over two steps). ESIMS (M+H)+ = 521. 1 H NMR (400 MHz, DMSOd6) δ ppm 1.00 (d, J=6.23 Hz, 6 H) 2.54 - 2.60 (m, 4 H) 2.64 - 2.72 (m, 1 H) 2.77 (d, J=4.77 Hz, 3 H) 3.05 - 3.13 (m, 4 H) 3.79 (s, 3 H) 6.23 (dd, J=3.48, 2.02 Hz, 1 H) 6.46 (dd, J=8.80, 2.57 Hz, 1 H) 6.61 (d, J=2.57 Hz, 1 H) 6.88 - 6.97 (m, 1 H) 7.49 (s, 1 H) 7.63 (d, J=5.50 Hz, 1 H) 7.81 (d, J=8.80 Hz, 1 H) 8.06 - 8.14 (m, 1 H) 8.40 (d, J=5.13 Hz, 1 H) 11.26 (s, 1 H) 1 1.28 - 1 1.30 (m, 1 H).
Example 38:
Λ/-methyl-3-[(2-{[2-(methyloxy)-4-(1 -propyl -4-piperidinyl)phenyl]amino}-1 H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2 -thiophenecarboxamide
Figure imgf000216_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one hydrochloride (109 mg, 0.23 mmol) and [2-(methyloxy)-4-(1 -propyl-4-piperidinyl)phenyl]amine (64 mg, 0.26 mmol) in trifluoroethanol (22 ml) was heated at 9O0C for 3 hrs. The reaction was concentrated by rotary evaporation, solids triturated from ethyl acetate/hexanes, and dried under house vacuum prior to next step. ESIMS (M+H)+ = 643. The solids were then added to tetrahydrofuran (50 ml) followed by methylamine 2.0M/THF (11.67 ml_, 23.34 mmol) and the resulting mixture was stirred at rt for 72 hrs. Next, the crude reaction was adsorbed to silica gel and purified by LC (DCM to 5% MeOH/DCM over 30 min). ESIMS (M+H)+ = 674. The isolated amide was dissolved in 1 ,4-dioxane (10 ml), potassium hydroxide (1.039 ml_, 1.039 mmol) was added and the reaction was heated in the microwave at 12O0C for 20 min. The resulting solution was adsorbed onto silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford the N- methyl-3-[(2-{[2-(methyloxy)-4-(1-propyl-4-piperidinyl)phenyl]amino}-1 /-/-pyrrolo[2,3- c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (0.018 g, 15% over three steps).
ESIMS (M+H)+ = 520. 1 H NMR (400 MHz, DMSO-d6) δ ppm 0.86 (t, J=7.33 Hz, 3 H) 1.49-1.93 (s, 6 H) 2.74 (d, J=4.40 Hz, 4 H) 3.82 (s, 3 H) 6.22-6.25 (m, 1 H) 6.76 (d, J=8.24 Hz, 1 H) 6.85 (s, 1 H) 6.93 - 6.99 (m, 1 H) 7.58 (s, 1 H) 7.65 (d, J=5.49 Hz, 1 H) 8.06 - 8.14 (m, 2 H) 8.37 (d, J=5.31 Hz, 1 H) 11.27 (s, 1 H) 1 1.35 (s, 1 H)
Example 39:
3-[(2-{[4-[4-(1 -methylethyl)-1 -piperazinyl]-2-(methyloxy)phenyl]amino}-1 H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-propyl-2-thiophenecarboxamide
Figure imgf000217_0001
A mixture of 5-{[4-[4-(1-methylethyl)-1-piperazinyl]-2-(methyloxy)phenyl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one (200 mg, 0.311 mmol) and propylamine (2.55 ml_, 31.1 mmol) in tetrahydrofuran (THF) (20ml) was stirred at rt for 36 hrs. Next, the crude reaction was adsorbed to silica gel and purified by LC (DCM to 5% MeOH/DCM). ESIMS (M+H)+ = 703. The isolated pure amide was redissolved in 1 ,4-dioxane (10 ml), potassium hydroxide (2.276 ml_, 2.276 mmol) was added and the reaction was heated in the microwave at 120C for 20 min. The resulting solution was adsorbed onto silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford 3-[(2-{[4-[4-(1-methylethyl)-1-piperazinyl]-2- (methyloxy)phenyl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-propyl-2- thiophenecarboxamide (0.035g, 22% over two steps). ESIMS (M+H)+ = 549. 1H NMR (400 MHz, DMSOd6) δ ppm 0.84 (t, J=7.42 Hz, 3 H) 0.97 (d, J=6.59 Hz, 6 H) 1.45 - 1.54 (m, 2 H) 2.51 - 2.57 (m, 4 H) 2.59 - 2.67 (m, 1 H) 3.03 - 3.09 (m, 4 H) 3.13 - 3.21 (m, 2 H) 3.76 (s, 3 H) 6.18-6.21 (m, 1 H) 6.44 (dd, J=8.79, 2.20 Hz, 1 H) 6.57-6.60 (m, 1 H) 6.88-6.91 (m, 1 H) 7.45 (s, 1 H) 7.61 (d, J=5.49 Hz, 1 H) 7.79 (d, J=8.79 Hz, 1 H) 8.13 (t, J=5.77 Hz, 1 H) 8.37 (d, J=5.49 Hz, 1 H) 11.22 (s, 1 H) 1 1.25 (s, 1 H).
Example 40:
3-[(2-{[2-(methyloxy)-4-(1 -propyl-4-piperidinyl)phenyl]amino}-1H-pyrrolo[2,3- c/]pyrimidin-4-yl)amino]-Λ/-propyl-2-thiophenecarboxamide
Figure imgf000218_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one hydrochloride (137 mg, 0.29 mmol) and [2-(methyloxy)-4-(1-propyl-4-piperidinyl)phenyl]amine (80 mg, 0.32 mmol) in 2,2,2-trifluoroethanol (27 ml) was heated at 9O0C for 3 hrs. The reaction was concentrated by rotary evaporation, solids triturated from ethyl acetate/hexanes, and dried under house vacuum prior to next step. ESIMS (M+H)+ = 643. The solids were then added to tetrahydrofuran (50 ml) followed by propylamine (2.56 ml_, 31.1 mmol) and the resulting mixture was stirred at rt for 36 hrs. Next, the crude reaction was adsorbed to silica gel and purified by LC (DCM to 5% MeOH/DCM). ESIMS (M+H)+ = 702. The isolated amide was redissolved in 1 ,4-Dioxane (10 ml), potassium hydroxide (1.995 ml_, 1.995 mmol) was added and the reaction was heated in the microwave at 120C for 20 min. The resulting solution was adsorbed onto silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford 3-[(2-{[2-(methyloxy)-4-(1-propyl- 4-piperidinyl)phenyl]amino}-1 H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-propyl-2- thiophenecarboxamide (0.035 g, 22% over three steps). ESIMS (M+H)+ = 548. 1 H NMR (400 MHz, DMSOd6) δ ppm 0.81 - 0.91 (m, 6 H) 1.47 - 2.03 (m, 9 H) 2.90-3.05 (m, 2H) 3.13 - 3.22 (m, 2 H) 3.82 (s, 3 H) 6.21 - 6.27 (m, 1 H) 6.77 (d, J=8.06 Hz, 1 H) 6.85 (s, 1 H) 6.94 - 6.98 (m, 1 H) 7.58 (s, 1 H) 7.65 (d, J=5.49 Hz, 1 H) 8.10 - 8.21 (m, 2 H) 8.36 (d, J=5.31 Hz, 1 H) 11.26 (s, 1 H) 1 1.35 (s, 1 H).
Example 41 :
3-[(2-{[1 -(N,N-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-1 H- pyrrolo[2,3-d]pyrimidin-4-yl)amino]-N-methyl-2-thiophenecarboxamide
Figure imgf000219_0001
A mixture of 5-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}- 3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)- one (300 mg, 0.44mmol, generated as in Example 2) and methylamine 2.0M/THF (11.65 mL, 23.30 mmol) in tetrahydrofuran (THF) (20ml) was stirred at rt for three days.. Next, the crude reaction was adsorbed to silica gel and purified by LC (DCM to 5% MeOH/DCM). ESIMS (M+H)+ = 675. The isolated pure amide was redissolved in 1 ,4- dioxane (10 ml), potassium hydroxide (4.08 ml_, 4.08 mmol) was added and the reaction was heated in the microwave at 120C for 20 min. The resulting solution was adsorbed onto silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford 3-[(2-{[1 -(N1N- dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-1 H-pyrrolo[2,3- d]pyrimidin-4-yl)amino]-N-methyl-2-thiophenecarboxamide (0.055g, 24% over two steps). ESIMS (M+H)+ = 521. 1 H NMR (400 MHz, DMSOd6) δ ppm 2.23 (s, 6 H) 2.76 (d, J=4.77 Hz, 3 H) 3.11 (t, J=8.07 Hz, 2 H) 3.16 (s, 2 H) 3.76 (s, 3 H) 4.16 (t, J=8.43 Hz, 2 H) 6.23-6.26 (m, 1 H) 6.91 - 6.96 (m, 2 H) 7.57 (d, J=5.50 Hz, 1 H) 7.68 (s, 1 H) 8.08- 8.1 1 (m, 1 H) 8.38 (d, J=5.50 Hz, 1 H) 8.56 (s, 1 H) 1 1.27 (s, 1 H) 11.30 (s, 1 H).
Example 42:
3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino}-1H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-propyl-2-thiophenecarboxamide
Figure imgf000220_0001
A mixture of 5-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1/-/-indol-6-yl]amino}- 3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)- one (300 mg, 0.44mmol, generated as in Example 2) and propylamine (1.916 ml_, 23.30 mmol) in tetrahydrofuran (THF) (20ml) was stirred at rt for two days. Next, the crude reaction was adsorbed to silica gel and purified by LC (DCM to 5% MeOH/DCM). ESIMS (M+H)+ = 703. The isolated amide was redissolved in 1 ,4-dioxane (10 ml), potassium hydroxide (7.68 mL, 7.68 mmol) was added and the reaction was heated in the microwave at 12O0C for 20 min. The resulting solution was adsorbed onto silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5- (methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/- propyl-2-thiophenecarboxamide (0.08Og, 33% over two steps). ESIMS (M+H)+ = 549. 1 H NMR (400 MHz, DMSOd6) δ ppm 0.84 (t, J=7.42 Hz, 3 H) 1.44 - 1.55 (m, 2 H) 2.20 (s, 6 H) 3.03 - 3.23 (m, 6 H) 3.73 (s, 3 H) 4.14 (t, J=8.15 Hz, 2 H) 6.19 - 6.24 (m, 1 H) 6.88 - 6.99 (m, 2 H) 7.54 (d, J=6.04 Hz, 1 H) 7.64 (s, 1 H) 8.09 - 8.17 (m, 1 H) 8.35 (d, J=5.68 Hz, 1 H) 8.54 (s, 1 H) 1 1.21 - 1 1.32 (m, 2 H).
Example 43:
Λ/-methyl-3-[(2-{[6-(methyloxy)-1 -(1 -methyl-L-prolyl)-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000221_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one hydrochloride (300 mg, 0.64 mmol) and 6-(methyloxy)-1-(1-methyl-L-prolyl)-1 ,2,3,4-tetrahydro-7-quinolinamine
(223 mg, 0.77 mmol) in THF (20 ml) was heated at 650C for 4 hrs. The reaction was diluted with ethyl acetate (200 ml) and washed with saturated NaHCOs (300 ml). Organic layer was removed, concentrated by rotary evaporation, solids triturated from ethyl acetate/hexanes, and dried under house vacuum prior to next step. ESIMS (M+H)+ = 684. The solids were then added to tetrahydrofuran (THF) (100 ml) followed by methylamine 2.0M/THF (25.6 ml_, 51.2 mmol) and the resulting mixture was stirred at rt overnight. The crude reaction was adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM). ESIMS (M+H)+ = 715. The isolated amide was redissolved in 1 ,4- Dioxane (6 ml), potassium hydroxide (6.29 ml_, 6.29 mmol) added and the reaction was heated in the microwave at 12O0C for 20 min. The resulting solution was adsorbed onto silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford Λ/-methyl-3-[(2-{[6- (methyloxy)-i -(1 -methyl-L-prolyl)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-1 /-/-pyrrolo[2,3- c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (0.05 g, 14% over three steps). ESIMS (M+H)+ = 561. 1 H NMR (400 MHz, DMSOd6) δ ppm 1.48 - 1.98 (m, 8 H) 2.06 (s, 3 H) 2.10 - 2.21 (m, 1 H) 2.57 - 2.74 (m, 2 H) 2.78 (d, J=4.01 Hz, 2 H) 2.86 - 2.95 (m, 1 H) 3.42 - 3.54 (m, 1 H) 3.87 (s, 3 H) 3.93 - 4.02 (m, 1 H) 6.30 (s, 1 H) 6.88 (s, 1 H) 7.01- 7.04 (m, 1 H) 7.62 - 7.71 (m, 2 H) 8.14-8.18 (m, 1 H) 8.30 - 8.41 (m, 2 H) 1 1.32-1 1.35(m, 2 H).
Example 44:
3-[(2-{[6-(methyloxy)-1 -(1 -methyl-L-prolyl)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}- 1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000222_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one hydrochloride (300 mg, 0.64 mmol) and 6-(methyloxy)-1-(1-methyl-L-prolyl)-1 ,2,3,4-tetrahydro-7-quinolinamine (223 mg, 0.77 mmol) in THF (20 ml) was heated at 650C for 4 hrs. The reaction was diluted with ethyl acetate (200 ml) and washed with saturated NaHCO3 (300 ml). Organic layer was removed, concentrated by rotary evaporation, solids triturated from ethyl acetate/hexanes, and dried under house vacuum prior to next step. ESIMS (M+H)+ = 684. The solids were then added to tetrahydrofuran (THF) (50 ml) followed by ammonium hydroxide (199 ml_, 51 18 mmol) and the resulting mixture was stirred at rt for three days. The crude reaction was adsorbed to silica gel and purified by LC (DCM to 5% MeOH/DCM). ESIMS (M+H)+ = 701. To the isolated amide in tetrahydrofuran (10 ml) and methanol (2 ml) was added sodium methoxide (46.3 mg, 0.856 mmol) and the reaction was heated at 6O0C for 2 hrs. The reaction was adsorbed to silica gel and purified by LC (DCM to 5% MeOH/DCM + 0.1% NH4OH) to afford 3-[(2-{[6-(methyloxy)- 1-(1-methyl-L-prolyl)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4- yl)amino]-2-thiophenecarboxamide (0.013 g, 3.7% over three steps). ESIMS (M+H)+ = 547. 1 H NMR (400 MHz, DMSOd6) δ ppm 1.47 - 1.92 (m, 5 H) 2.07 (s, 3 H) 2.15 - 2.20 (m, 1 H) 2.55 - 2.91 (m, 4 H) 3.39-3.55 (m, 2 H) 3.88 (2, 3 H) 3.94-4.02 (m, 1 H) 6.27 (dd, J=3.21 , 1.81 Hz, 1 H) 6.89 (s, 1 H) 7.02-7.44 (m, 1 H) 7.51-7.63 (m, 2H) 7.66 (s, 1 H) 7.69 (d, J=5.42 Hz, 1 H) 8.33 - 8.37 (m, 1 H) 8.39 (d, J=5.42 Hz, 1 H) 1 1.34 (s, 1 H) 11.43 (s, 1 H)
Example 45: 3-[(2-{[1-(Λ/,Λ/-dimethyl-L-alanyl)-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino}- 1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2-thiophenecarboxamide
Figure imgf000223_0001
A mixture 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2- c]pyrimidin-7(3/-/)-one hydrochloride (500 mg, 1.07 mmols) and 1-[(2S)-2- (dimethylamino)propanoyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-amine (336 mg, 1.276 mmol) in tetrahydrofuran (50 ml) was heated at 650C for 4 hrs. The reaction was diluted with ethyl acetate (200 ml) and washed with saturated NaHCO3 (300 ml). The organic layer was removed, concentrated, solids triturated from ethyl acetate/hexanes, and dried under house vacuum. ESIMS (M+H)+ = 658. A solution of 5-{[1-(Λ/,Λ/-dimethyl-L-alanyl)- 5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one (450 mg, 0.672 mmol) and methylamine 2.0M/THF (16.80 ml, 33.6 mmol) in tetrahydrofuran (50 ml) was stirred at rt overnight. The crude reaction was adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM). ESIMS (M+H)+ = 701. The isolated amide was dissolved in 1 ,4- dioxane (10 ml), potassium hydroxide (2.497 ml, 12.49 mmol) was added and the reaction was heated in the microwave at 12O0C for 20 min. The resulting solution was adsorbed onto silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford 3-[(2- {[1-(Λ/,Λ/-dimethyl-L-alanyl)-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}-1 /-/- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2-thiophenecarboxamide (0.100g, 18% over three steps). ESIMS (M+H)+ = 521. 1H NMR (400 MHz, DMSOd6) δ ppm 1.04 (d, J=6.62 Hz, 3 H) 2.20 (s, 6 H) 2.76 - 2.80 (m, 3 H) 3.09 - 3.17 (m, 2 H) 3.55 - 3.63 (m, 1 H) 3.77 (s, 3 H) 4.1 1 - 4.21 (m, 1 H) 4.36 - 4.43 (m, 1 H) 6.26 (dd, J=3.61 , 1.81 Hz, 1 H) 6.94 - 7.00 (m, 2 H) 7.57 (d, J=5.62 Hz, 1 H) 7.69 (s, 1 H) 8.09 - 8.16 (m, 1 H) 8.39 (d, J=5.42 Hz, 1 H) 8.57 (s, 1 H) 1 1.28 (s, 1 H) 11.32 (s, 1 H).
Example 46:
3-[(2-{[1-(Λ/,Λ/-dimethyl-L-alanyl)-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino}-
1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000224_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one hydrochloride (222 mg, 0.48 mmol) and 1 -[(2S)-2-(dimethylamino)propanoyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-amine (149 mg, 0.57 mmol) in THF (50 ml) was heated at 650C for 4 hrs. The reaction was diluted with ethyl acetate (200 ml) and washed with saturated NaHCO3 (300 ml). Organic layer was removed, concentrated by rotary evaporation, solids triturated from ethyl acetate/hexanes, and dried under house vacuum prior to next step. ESIMS (M+H)+ = 658. A solution of 5-{[1-(N,N-dimethyl-L-alanyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2- c]pyrimidin-7(3H)-one (200 mg, 0.304 mmol) and potassium carbonate (840 mg, 6.08 mmol) in 2,2,2-trifluoroethanol (100 ml) was stirred at rt overnight. The crude reaction was concentrated, diluted with ethyl acetate (200 ml) and washed with water ( 300 ml). The organic layer was removed, concentrated, and maintained under vacuum overnight. ESIMS (M+H)+ = 758. The solids were then added to tetrahydrofuran (50 ml) and trifluoroethanol (25 ml) followed by 27% ammonium hydroxide (103 ml, 2639 mmol) and the resulting mixture was stirred overnight at rt. The crude reaction was adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM). ESIMS (M+H)+ = 675. To the isolated amide in tetrahydrofuran (25 ml) and methanol (5 ml) was added sodium methoxide (46.3 mg, 0.856 mmol) and the reaction was heated at 6O0C for 2 hrs. The reaction was adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford 3-[(2-{[1 -(Λ/,Λ/-dimethyl-L-alanyl)-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}- 1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (0.076 g, 31% over four steps). ESIMS (M+H)+ = 521. 1H NMR (400 MHz, DMSOd6) δ ppm 1.04 (d, J=6.62 Hz, 3 H) 2.20 (s, 6 H) 3.08 - 3.18 (m, 2 H) 3.55 - 3.64 (m, 1 H) 3.77 (s, 3 H) 4.11 - 4.22 (m, 1 H) 4.36 - 4.46 (m, 1 H) 6.22 (dd, J=3.51 , 1.91 Hz, 1 H) 6.94 - 7.00 (m, 2 H) 7.50-7.62 (m, 3 H) 7.70 (s, 1 H) 8.39 (d, J=5.42 Hz, 1 H) 8.57 (s, 1 H) 11.28 (s, 1 H) 1 1.40 (s, 1 H)
Example 47:
3-[(2-{[1-(Λ/,Λ/-dimethyl-D-alanyl)-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino}- 1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2-thiophenecarboxamide
Figure imgf000225_0001
A mixture 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2- c]pyrimidin-7(3/-/)-one hydrochloride (250 mg, 0.54 mmols) and 1-[(2R)-2- (dimethylamino)propanoyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-amine (153 mg, 0.58 mmol) in tetrahydrofuran (50 ml) was heated at 650C for 4 hrs. The reaction was diluted with ethyl acetate (200 ml) and washed with saturated NaHCOs ( 300 ml). The organic layer was removed, concentrated, solids triturated from ethyl acetate/hexanes, and dried under house vacuum. ESIMS (M+H)+ = 658. A solution of 5-{[1-(N,N-dimethyl-D- alanyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (200 mg, 0.304 mmol) and methylamine 2.0M/THF (15.20 ml, 30.4 mmol) in tetrahydrofuran (50 ml) was stirred at rt overnight. The crude reaction was adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM). ESIMS (M+H)+ = 689. The isolated amide was redissolved in tetrahydrofuran (20 ml) and methanol (5 ml), followed by the addition of sodium methoxide (157 mg, 2.90 mmol). After heating at 6O0C for 3 hrs, the resulting solution was adsorbed onto silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford 3-[(2-{[1-(Λ/,Λ/-dimethyl-D-alanyl)-5-(methyloxy)-2,3-dihydro-1 H- indol-6-yl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2- thiophenecarboxamide (0.100g, 35% over three steps). ESIMS (M+H)+ = 535. 1H NMR (400 MHz, DMSOd6) δ ppm 1.04 (d, J=6.62 Hz, 3 H) 2.20 (s, 6 H) 2.77 - 2.80 (m, 3 H) 3.08 - 3.18 (m, 2 H) 3.55 - 3.64 (m, 1 H) 3.77 (s, 3 H) 4.09 - 4.21 (m, 1 H) 4.36 - 4.48 (m, 1 H) 6.26 (d, J=3.41 Hz, 1 H) 6.93 - 7.00 (m, 2 H) 7.57 (d, J=5.62 Hz, 1 H) 7.69 (s, 1 H) 8.06 - 8.17 (m, 1 H) 8.39 (d, J=5.42 Hz, 1 H) 8.57 (s, 1 H) 11.32 (s, 2 H).
Example 48:
3-[(2-{[1-(Λ/,Λ/-dimethyl-D-alanyl)-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino}-
1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000226_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one hydrochloride (250 mg, 0.54 mmol) and 1 -[(2R)-2-(dimethylamino)propanoyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-amine (153 mg, 0.58 mmol) in THF (50 ml) was heated at 650C for 4 hrs. The reaction was diluted with ethyl acetate (200 ml) and washed with saturated NaHCO3 (300 ml). The organic layer was removed, concentrated by rotary evaporation, solids triturated from ethyl acetate/hexanes, and dried under house vacuum prior to next step. ESIMS (M+H)+ = 658. A solution of 5-{[1-(N,N-dimethyl-D-alanyl)-5-(methyloxy)-2,3-dihydro- 1 H-indol-6-yl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (200 mg, 0.304 mmol) and potassium carbonate (840 mg, 6.08 mmol) in 2,2,2-trifluoroethanol (100 ml) was stirred at rt overnight. The crude reaction was concentrated, diluted with ethyl acetate (200 ml) and washed with water ( 300 ml). The organic layer was removed, concentrated, and dried under high vacuum overnight. ESIMS (M+H)+ = 758. The solids were then added to tetrahydrofuran (50 ml) and trifluoroethanol (25 ml) followed by 27% ammonium hydroxide (103 ml, 2639 mmol) and the resulting mixture was stirred overnight at 5OC. The organic layer was adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford 3-[(2-{[1-(Λ/,Λ/-dimethyl-D-alanyl)-5-(methyloxy)-2,3-dihydro-1 H- indol-6-yl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (0.130 g, 47% over three steps). ESIMS (M+H)+ = 521. 1H NMR (400 MHz, DMSOd6) δ ppm 1.04 (d, J=6.42 Hz, 3 H) 2.20 (s, 6 H) 3.08 - 3.18 (m, 2 H) 3.56 - 3.64 (m, 1 H) 3.77 (s, 3 H) 4.1 1 - 4.22 (m, 1 H) 4.36 - 4.46 (m, 1 H) 6.22 (dd, J=3.41 , 1.81 Hz, 1 H) 6.92 - 7.00 (m, 2 H) 7.51 - 7.61 (m, 3 H) 7.70 (s, 1 H) 8.39 (d, J=5.42 Hz, 1 H) 8.57 (s, 1 H) 11.28 (s, 1 H) 1 1.40 (s, 1 H).
Example 49
3-({2-[(5-methyl-2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1 - piperazinyl}phenyl)amino]-1H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2- thiophenecarboxamide
Figure imgf000228_0001
Step A\ Intermediate C28: 5-[(5-methyl-2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1- piperazinyl}phenyl)amino]-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one
Figure imgf000228_0002
To a suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one HCI salt (0.5 g, 1.07 mmol) in 2,2,2- trifluoroethanol (15 mL) was added 5-methyl-2-(methyloxy)-4-{4-[2- (methylsulfonyl)ethyl]-1-piperazinyl}aniline (0.419 g, 1.28 mmol). The resulting slurry was stirred at rt for 4 h. The reaction mixture was diluted with dichloromethane and saturated sodium bicarbonate as to adjust the aqueous layer to pH >10. The organic layer was washed with water and saturated brine solution and dried over sodium sulfate. Solvents were removed under reduced pressure to afford a green solid that was purified via trituration from diethyl ether to afford 5-[(5-methyl-2-(methyloxy)-4-{4-[2- (methylsulfonyl)ethyl]-1-piperazinyl}phenyl)amino]-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (760 mg, 1.05 mmol) as a green solid (90% clean) (ESIMS (M+H)+ =722)
Step B\ Intermediate C9: 3-({2-[(5-methyl-2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1- piperazinyl}phenyl)amino]-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4- yl}amino)-2-thiophenecarboxamide
Figure imgf000229_0001
To a solution of : 5-[(5-methyl-2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1- piperazinyl}phenyl)amino]-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (760 mg, 1.05 mmol) in THF (50 ml.) was added ammonium hydroxide (27 % aqueous) (>100 ml.) The resulting mixture was let stir at 9O0C in a pressure vessel for 8 h at which time it was diluted with ethyl acetate and washed with a saturated sodium bicarbonate solution, water and a saturated brine solution. Organics were dried over sodium sulfate and solvents removed under reduced pressure. Purification via column chromatography afforded a yellow solid, 3- ({2-[(5-methyl-2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1-piperazinyl}phenyl)amino]-7- [(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2- thiophenecarboxamide (291 mg, 0.0.395 mmol)). ESIMS(M+H)+ = 739 Step C\ Example 49: 3-({2-[(5-methyl-2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1- piperazinyl}phenyl)amino]-1/-/-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2- thiophenecarboxamide
Figure imgf000230_0001
3-({2-[(5-methyl-2-(methyloxy)-4-{4-[2-(methylsulfonyl)ethyl]-1-piperazinyl}phenyl)amino]- 7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2- thiophenecarboxamide (291 mg, 0.0.395 mmol) was dissolved in 1 ,4-dioxane (10 ml.) and transferred to a 20 ml microwave vessel. A solution of 6M NaOH (3 ml) and water (3 ml) was added and reaction heated in microwave at 12O0C for 25 minutes. Resulting brown solution was diluted with ethyl acetate and washed with a saturated sodium bicarbonate solution, water and a saturated brine solution. Organics were dried over sodium sulfate and solvents removed under reduced pressure to afford a yellow solid which was purified via chromatography on Siθ2 to afford 3-({2-[(5-methyl-2-(methyloxy)- 4-{4-[2-(methylsulfonyl)ethyl]-1-piperazinyl}phenyl)amino]-1 /-/-pyrrolo[2,3-c/]pyrimidin-4- yl}amino)-2-thiophenecarboxamide (105 mg, 0.108 mmol) as a yellow solid. 1 H NMR (400 MHz, DMSOd6) δ ppm 2.22 (s, 3 H) 2.56 - 2.68 (m, 4 H) 2.75 - 2.82 (m, 2 H) 2.83 - 2.91 (m, 4 H) 3.08 (s, 3 H) 3.33 - 3.37 (m, 2 H) 3.82 (s, 3 H) 6.20 - 6.27 (m, 1 H) 6.73 (s, 1 H) 6.95 - 7.02 (m, 1 H) 7.49 (s, 1 H) 7.52 - 7.67 (m, 2 H) 7.69 (d, J=5.40 Hz, 1 H) 7.89 (s, 1 H) 8.35 - 8.42 (m, 1 H) 1 1.29 - 11.36 (m, 1 H) 11.39 (s, 1 H) ESIMS(M+H)+ = 585
Example 50 3-({2-[(2-(ethyloxy)-5-methyl-4-{4-[2-(methylsulfonyl)ethyl]-1- piperazinyl}phenyl)amino]-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3- c/]pyrimidin-4-yl}amino)-2-thiophenecarboxamide
NH2
N
) N
O=S=O
N2679-66-1
Step AMntermediate C10: 5-[(2-(ethyloxy)-5-methyl-4-{4-[2-(methylsulfonyl)ethyl]-1- piperazinyl}phenyl)amino]-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one
Figure imgf000231_0001
To a suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one HCI salt (0.5 g, 1.07 mmol) in 2,2,2- trifluoroethanol (15 ml.) was added 2-(ethyloxy)-5-methyl-4-{4-[2-(methylsulfonyl)ethyl]- 1-piperazinyl}aniline (0.435 g, 1.28 mmol). The resulting slurry was stirred at rt in a pressure vial for 16 h. The reaction mixture was diluted with dichloromethane and saturated sodium bicarbonate as to adjust the aqueous layer to pH >10. The organic layer was washed with water and saturated brine solution and dried over sodium sulfate. Solvents were removed under reduced pressure to afford a yellow solid that was purified via trituration from diethyl ether to afford 5-[(2-(ethyloxy)-5-methyl-4-{4-[2- (methylsulfonyl)ethyl]-1-piperazinyl}phenyl)amino]-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one (700 mg, 0.951 mmol) as a green solid. (ESIMS (M+H)+ =736)
Step B\ Example 50: 3-({2-[(2-(ethyloxy)-5-methyl-4-{4-[2-(methylsulfonyl)ethyl]-1- piperazinyl}phenyl)amino]-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4- yl}amino)-2-thiophenecarboxamide
Figure imgf000232_0001
To a solution of 5-[(2-(ethyloxy)-5-methyl-4-{4-[2-(methylsulfonyl)ethyl]-1- piperazinyl}phenyl)amino]-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one (700 mg, 0.951 mmol) in THF (50 ml.) was added ammonium hydroxide (27 % aqueous) (>100 ml.) The resulting mixture was let stir at 9O0C in a pressure vessel for -24 h at which time it was diluted with ethyl acetate and washed with a saturated sodium bicarbonate solution, water and a saturated brine solution. Organics were dried over sodium sulfate and solvents removed under reduced pressure. Purification via column chromatography afforded a yellow solid containing 3-({2-[(2-(ethyloxy)-5-methyl-4-{4-[2-(methylsulfonyl)ethyl]-1- piperazinyl}phenyl)amino]-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4- yl}amino)-2-thiophenecarboxamide (70% pure, 312 mg, 0.414 mmol) This material was dissolved in 1 ,4-dioxane (10 ml.) and transferred to a 20 ml microwave vessel. A solution of 6M NaOH (3 ml) and water (3 ml) was added and the reaction was heated in a microwave at 12O0C for 20 minutes. Resulting brown solution was diluted with ethyl acetate and washed with a saturated sodium bicarbonate solution, water and a saturated brine solution. Organics were dried over sodium sulfate and solvents removed under reduced pressure to afford a yellow solid which was purified via chromatography on SiO2 to afford 3-({2-[(2-(ethyloxy)-5-methyl-4-{4-[2-(methylsulfonyl)ethyl]-1 - piperazinyl}phenyl)amino]-1 H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2- thiophenecarboxamide (185 mg, 0.309 mmol) as a light brown solid. 1H NMR (400 MHz, DMSO-de) δ ppm 1.34 (t, J=6.97 Hz, 3 H) 2.21 (s, 3 H) 2.54 - 2.68 (m, 4 H) 2.78 (t, J=6.62 Hz, 2 H) 2.82 - 2.90 (m, 4 H) 3.08 (s, 3 H) 3.34 - 3.39 (m, 2 H) (partly under water peak) 4.08 (q, J=6.86 Hz, 2 H) 6.25 (dd, J=3.36, 1.76 Hz, 1 H) 6.72 (s, 1 H) 7.00 (dd, J=3.26, 2.26 Hz, 1 H) 7.42 (s, 1 H) 7.47 - 7.67 (m, 2 H) 7.71 (d, J=5.32 Hz, 1 H) 7.93 (s, 1 H) 8.36 (d, J=5.42 Hz, 1 H) 11.34 (s, 1 H) 1 1.40 (s, 1 H) ESIMS(M+H)+ = 599
Example 51
3-({2-[(5-ethyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1 -piperazinyl]-1 - piperidinyl}phenyl)amino]-1H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2- thiophenecarboxamide
Figure imgf000234_0001
Step A\ Intermediate C1 1 : 3-({2-[(5-ethyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1- piperazinyl]-1-piperidinyl}phenyl)amino]-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3- c/]pyrimidin-4-yl}amino)-2-thiophenecarboxamide
Figure imgf000234_0002
To a suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one HCI salt (0.5 g, 1.07 mmol) in tetrahydrofuran (50 mL) was added 5-ethyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1- piperazinyl]-1-piperidinyl}aniline (0.508 g, 1.28 mmol). The resulting slurry was stirred at rt in a pressure vial for 16 h. Analysis by LCMS indicated all starting material had been consumed so ammonium hydroxide (27 % in water) (100 ml) was added and the reaction was heated to 800C in a pressure vessel for 16 hours. The resulting solution was cooled to room temperature and diluted with ethyl acetate and washed with saturated sodium bicarbonate solution, water and saturated brine solution and dried over sodium sulfate. Solvents were removed under reduced pressure to afford a yellow solid that was purified via column chromatography to afford 3-({2-[(5-ethyl-2-(methyloxy)-4-{4- [4-(methylsulfonyl)-1-piperazinyl]-1-piperidinyl}phenyl)amino]-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2-thiophenecarboxamide (0.575 g, 0.712 mmol) as a yellow solid. (90% clean) (ESIMS (M+H)+ =809)
Step B\ Example 51 : 3-({2-[(5-ethyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1- piperazinyl]-1-piperidinyl}phenyl)amino]-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2- thiophenecarboxamide:
Figure imgf000235_0001
A solution of 3-({2-[(5-ethyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1- piperidinyl}phenyl)amino]-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4- yl}amino)-2-thiophenecarboxamide (0.575 g, 0.712 mmol) in 1 ,4-dioxane (10 ml.) was transferred to a 20 ml microwave vessel. A solution of 6M NaOH (3 ml) and water (3 ml.) was added and reaction heated in microwave at 12O0C for 10 minutes. Microwave heating in 10 min intervals was continued until LCMS analysis indicated starting material consumed (-40 min). Resulting brown solution was diluted with ethyl acetate and washed with a saturated sodium bicarbonate solution, water and a saturated brine solution. Organics were dried over sodium sulfate and solvents removed under reduced pressure to afford an off white solid. Attempts to redissolve the compound in organic solvents were not successful so the solid was filtered. This material was then dissolved in a DMSO-MeOH solution and purified by HPLC (acetonitrile/ water with TFA). Fractions containing clean product were combined and neutralized with potassium carbonate. The resulting solution was diluted with EtOAc and washed with a saturated sodium bicarbonate solution, water and a saturated brine solution. Organics were dried over sodium sulfate and solvents removed under reduced pressure to afford 3-({2-[(5- ethyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1-piperidinyl}phenyl)amino]- 1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2-thiophenecarboxamide (88 mg, 0.135 mmol) as an off-white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.17 (t, J=7.46 Hz, 3 H) 1.53 - 1.67 (m, 2 H) 1.79 - 1.90 (m, 2 H) 2.35 - 2.47 (m, 1 H) 2.58 - 2.76 (m, 8 H) 2.88 (s, 3 H) 3.03 (d, J=11.35 Hz, 2 H) 3.08 - 3.16 (m, 4 H) 3.81 (s, 3 H) 6.24 (d, J=3.39 Hz, 1 H) 6.77 (s, 1 H) 6.99 (d, J=3.30 Hz, 1 H) 7.44 - 7.63 (m, 3 H) 7.67 (d, J=5.31 Hz, 1 H) 7.93 (s, 1 H) 8.37 (d, J=5.31 Hz, 1 H) 11.25 - 1 1.36 (m, 1 H) 11.39 (s, 1 H) ESIMS(M+H)+ = 654
Example 52
3-[(2-{[4-{4-[(3/?)-3-(dimethylamino)-1 -pyrrolidinyl]-1-piperidinyl}-2- (methyloxy)phenyl]amino}-1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2- thiophenecarboxamide
Figure imgf000236_0001
Step A\ Intermediate C12: 5-{[4-{4-[(3R)-3-(dimethylamino)-1-pyrrolidinyl]-1-piperidinyl}- 2-(methyloxy)phenyl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one
Figure imgf000237_0001
To a suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one HCI salt (0.5 g, 1.07 mmol) in 2,2,2- trifluoroethanol (15 mL) was added (3R)-1-{1-[4-amino-3-(methyloxy)phenyl]-4- piperidinyl}-N,N-dimethyl-3-pyrrolidinamine (0.406 g, 1.27 mmol). The resulting slurry was stirred at rt for 3 h. Reaction mixture was diluted with dichloromethane and saturated sodium bicarbonate as to adjust the aqueous layer to pH >10. The organic layer was washed with water and saturated brine solution and dried over sodium sulfate. Solvents were removed under reduced pressure to afford a yellow solid that was purified via trituration from diethyl ether to afford 5-{[4-{4-[(3R)-3-(dimethylamino)-1-pyrrolidinyl]- 1-piperidinyl}-2-(methyloxy)phenyl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (680 mg, 0.954 mmol) as a green yellow solid. (ESIMS (M+H)+ = 713
Step B\ Example 52: 3-[(2-{[4-{4-[(3R)-3-(dimethylamino)-1-pyrrolidinyl]-1-piperidinyl}-2-
(methyloxy)phenyl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2- thiophenecarboxamide
Figure imgf000238_0001
/ N — /
To a solution of 5-{[4-{4-[(3R)-3-(dimethylamino)-1-pyrrolidinyl]-1-piperidinyl}-2- (methyloxy)phenyl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (680 mg, 0.954 mmol) in THF (50 ml.) was added ammonium hydroxide (27 % aqueous) (>100 ml.) The resulting mixture was let stir at 8O0C in a pressure vessel for 16 h at which time it was diluted with ethyl acetate and washed with a saturated sodium bicarbonate solution, water and a saturated brine solution. Organics were dried over sodium sulfate and solvents removed under reduced pressure. Purification via column chromatography afforded a yellow solid containing 3-({2-[(2-(ethyloxy)-5-methyl-4-{4-[2-(methylsulfonyl)ethyl]-1- piperazinyl}phenyl)amino]-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4- yl}amino)-2-thiophenecarboxamide (90% pure, 123 mg, 0.169 mmol) This material was dissolved in 1 ,4-dioxane (5 ml.) and transferred to a 20 ml microwave vessel. A solution of 6M NaOH (1.5 ml) and water (1.5 ml) was added and reaction heated in microwave at 12O0C for 16 minutes. Resulting brown solution was diluted with ethyl acetate and washed with a saturated sodium bicarbonate solution, water and a saturated brine solution. Organics were dried over sodium sulfate and solvents removed under reduced pressure to afford a yellow solid which was purified via chromatography on Siθ2 to afford 3-[(2-{[4-{4-[(3R)-3-(dimethylamino)-1 -pyrrolidinyl]-1 -piperidinyl}-2- (methyloxy)phenyl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2- thiophenecarboxamide (52 mg, 0.090 mmol) as a light brown solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.43 - 1.64 (m, 3 H) 1.78 - 1.96 (m, 3 H) 2.10 (s, 6 H) 2.31 (d, J=7.93 Hz, 1 H) 2.44 - 2.49 (m, 1 H)(hidden by DMSO peak) 2.59 - 2.77 (m, 4 H) 2.81 (t, J=7.88 Hz, 1 H) 3.17 (d, J=5.02 Hz, 1 H) 3.58 (d, J=12.34 Hz, 2 H) 3.80 (s, 3 H) 6.21 (dd, J=3.26, 1.86 Hz, 1 H) 6.50 (dd, J=8.68, 2.26 Hz, 1 H) 6.63 (d, J=2.31 Hz, 1 H) 6.95 (dd, J=3.26, 2.36 Hz, 1 H) 7.46 - 7.63 (m, 3 H) 7.68 (d, J=5.42 Hz, 1 H) 7.80 (d, J=8.63 Hz, 1 H) 8.39 (d, J=5.32 Hz, 1 H) 11.30 (s, 1 H) 1 1.36 (s, 1 H) ESIMS(M+H)+ = 576
Example 53
3-{[2-({2-(methyloxy)-4-[4-(4-morpholinyl)-1 -piperidinyl]phenyl}amino)-1H- pyrrolo[2,3-c/]pyrimidin-4-yl]amino}-2-thiophenecarboxamide
Figure imgf000239_0001
Step A\ Intermediate C13: 3-({2-({2-(methyloxy)-4-[4-(4-morpholinyl)-1- piperidinyl]phenyl}amino)-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4- yl}amino)-2-thiophenecarboxamide
Figure imgf000240_0001
To a suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one HCI salt (0.5 g, 1.07 mmol) in tetrahydrafuran (50 ml.) was added 2-(methyloxy)-4-[4-(4-morpholinyl)-1- piperidinyl]aniline (0.342 g, 1.17 mmol) — for synthesis see: Garcia-Echeverria, Carlos; Kanazawa, Takanori; Kawahara, Eiji; Masuya, Keiichi; Matsuura, Naoko; Miyake, Takahiro; Ohmori, Osamu; Umemura, Ichiro. Preparation of novel 2,4- di(phenylamino)pyrimidines useful in the treatment of neoplastic diseases, inflammatory and immune system disorders. PCT Int. Appl. (2004), 185 pp. CODEN: PIXXD2 WO 2004080980 A1 20040923 CAN 141 :296041 AN 2004:780679 CAPLUS. The resulting slurry was stirred at rt for 4 h. Ammonium hydroxide (27 % aqueous) (>100 ml.) was added and the resulting mixture was let stir at 8O0C in a pressure vessel for 2 days at which time it was diluted with ethyl acetate and washed with a saturated sodium bicarbonate solution, water and a saturated brine solution. Organics were dried over sodium sulfate and solvents removed under reduced pressure. Purification via column chromatography afforded a yellow solid containing 3-({2-({2-(methyloxy)-4-[4-(4- morpholinyl)-1-piperidinyl]phenyl}amino)-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3- c/]pyrimidin-4-yl}amino)-2-thiophenecarboxamide (283 mg, 0.403 mmol) as a yellow solid. (ESIMS (M+H)+ = 703
Step B\ Example 53: 3-{[2-({2-(methyloxy)-4-[4-(4-morpholinyl)-1- piperidinyl]phenyl}amino)-1 H-pyrrolo[2,3-d]pyrimidin-4-yl]amino}-2- thiophenecarboxamide
Figure imgf000241_0001
3-({2-({2-(methyloxy)-4-[4-(4-morpholinyl)-1-piperidinyl]phenyl}amino)-7-[(4- methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2-thiophenecarboxamide (283 mg, 0.403 mmol) was dissolved in 1 ,4-dioxane (10 ml.) and transferred to a 20 ml microwave vessel. A solution of 6M NaOH (3 ml) and water (3 ml) was added and the reaction heated in microwave at 12O0C for 15 minutes. Resulting brown solution was diluted with ethyl acetate and washed with a saturated sodium bicarbonate solution, water and a saturated brine solution. Organics were dried over sodium sulfate and solvents removed under reduced pressure to afford a yellow solid which was purified via chromatography on SiO2 to afford 3-{[2-({2-(methyloxy)-4-[4-(4-morpholinyl)-1- piperidinyl]phenyl}amino)-1 H-pyrrolo[2,3-d]pyrimidin-4-yl]amino}-2- thiophenecarboxamide (88 mg, 0.160 mmol) as a tan solid. 1H NMR (400 MHz, DMSO- de) δ ppm 1.52 (dd, J=11.67, 3.07 Hz, 2 H) 1.88 (d, J=1 1.54 Hz, 2 H) 2.21 - 2.31 (m, 1 H) 2.65 (t, J=11.35 Hz, 3 H) 3.17 - 3.19 (m, 2 H) 3.54 - 3.63 (m, 5 H) 3.69 (d, J=12.27 Hz, 2 H) 3.81 (s, 3 H) 6.18 - 6.25 (m, 1 H) 6.50 (dd, J=8.70, 2.29 Hz, 1 H) 6.64 (d, J=2.20 Hz, 1 H) 6.91 - 6.99 (m, 1 H) 7.45 - 7.64 (m, 3 H) 7.68 (d, J=5.40 Hz, 1 H) 7.81 (d, J=8.70 Hz, 1 H) 8.39 (d, J=5.40 Hz, 1 H) 1 1.30 (s, 1 H) 11.36 (s, 1 H) ESIMS(M+H)+ = 549
Example 54 3-({2-[(5-methyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1- piperidinyl}phenyl)amino]-1H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2- thiophenecarboxamide
Figure imgf000242_0001
Step A\ Intermediate C14: 5-[(5-methyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1- piperazinyl]-1-piperidinyl}phenyl)amino]-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one
Figure imgf000242_0002
To a suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one HCI salt (0.450 g, 1.07 mmol) in 2,2,2-trifluoroethanol (50 ml.) was added 5-methyl-2-(methyloxy)-4-{4-[4- (methylsulfonyl)-1-piperazinyl]-1-piperidinyl}aniline (0.384 g, 1.00 mmol). The resulting slurry was stirred at rt for 3 h. The reaction mixture was diluted with dichloromethane and saturated sodium bicarbonate as to adjust the aqueous layer to pH >10. The organic layer was washed with water and saturated brine solution and dried over sodium sulfate. Solvents were removed under reduced pressure to afford a yellow solid that was purified via trituration from diethyl ether to afford 5-[(5-methyl-2-(methyloxy)-4-{4-[4- (methylsulfonyl)-1-piperazinyl]-1-piperidinyl}phenyl)amino]-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (618 mg, 0.79 mmol) as a yellow solid. (ESIMS (M+H)+ = 713
Step B\ Example C15: 3-({2-[(5-methyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1- piperazinyl]-1-piperidinyl}phenyl)amino]-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3- c/]pyrimidin-4-yl}amino)-2-thiophenecarboxamide
Figure imgf000243_0001
To a solution of 5-[(5-methyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1- piperidinyl}phenyl)amino]-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one (618 mg, 0.79 mmol) in THF (50 ml.) was added ammonium hydroxide (27 % aqueous) (>150 ml.) The resulting mixture was let stir at 9O0C in a pressure vessel for 16 h at which time it was diluted with ethyl acetate and washed with a saturated sodium bicarbonate solution, water and a saturated brine solution. Organics were dried over sodium sulfate and solvents removed under reduced pressure. Purification via column chromatography afforded a yellow solid, 3- ({2-[(5-methyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1- piperidinyl}phenyl)amino]-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4- yl}amino)-2-thiophenecarboxamide (384 mg, 0.961 mmol). (ESIMS (M+H)+ = 777
Step C\ Example 54: 3-({2-[(5-methyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1- piperazinyl]-1-piperidinyl}phenyl)amino]-1 H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2- thiophenecarboxamide
3-({2-[(5-methyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1- piperidinyl}phenyl)amino]-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4- yl}amino)-2-thiophenecarboxamide (384 mg, 0.961 mmol) was dissolved in 1 ,4-dioxane (6 ml.) and transferred to a 20 ml microwave vessel. A solution of 6M NaOH (2 ml) and water (2 ml) was added and the reaction heated in microwave at 12O0C for 35 minutes. Resulting brown solution was diluted with ethyl acetate and washed with a saturated sodium bicarbonate solution, water and a saturated brine solution. Organics were dried over sodium sulfate and solvents removed under reduced pressure to afford a yellow solid which was purified via chromatography on SiO2 to afford 3-({2-[(5-methyl-2- (methyloxy)-4-{4-[4-(methylsulfonyl)-1 -piperazinyl]-1 -piperidinyl}phenyl)amino]-1 H- pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2 -thiophenecarboxamide (159 mg, 0.249 mmol) as an off-whte solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.54 - 1.68 (m, 2 H) 1.86 (d, J=10.99 Hz, 2 H) 2.21 (s, 3 H) 2.37 - 2.47 (m, 1 H) 2.59 - 2.72 (m, 6 H) 2.88 (s, 3 H) 3.06 - 3.16 (m, 6 H) 3.81 (s, 3 H) 6.24 (dd, J=3.16, 1.79 Hz, 1 H) 6.72 (s, 1 H) 6.95 - 7.02 (m, 1 H) 7.44 - 7.66 (m, 3 H) 7.69 (d, J=5.40 Hz, 1 H) 7.87 (s, 1 H) 8.38 (d, J=5.40 Hz, 1 H) 11.32 (s, 1 H) 1 1.39 (s, 1 H). ESIMS(M+H)+ = 640
Example 55 3-{[2-({2-(methyloxy)-4-[4-(2-methylpropyl)-1 -piperazinyl]phenyl}amino)-1 H- pyrrolo[2,3-c/]pyrimidin-4-yl]amino}-2 -thiophenecarboxamide
Figure imgf000245_0001
To a suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one HCI salt (0.5 g, 1.07 mmol) in tetrahydrofuran (50 ml.) was added 2-(methyloxy)-4-[4-(2-methylpropyl)-1- piperazinyl]aniline (0.295 g, 1.12 mmol). The resulting slurry was stirred at rt for 4 h. Ammonium hydroxide (27 % aqueous) (>100 ml.) was added and the resulting mixture was let stir at 8O0C in a pressure vessel for 8 h at which time it was diluted with ethyl acetate and washed with a saturated sodium bicarbonate solution, water and a saturated brine solution. Organics were dried over sodium sulfate and solvents removed under reduced pressure. Purification via column chromatography afforded a yellow solid containing -50% 3-({2-({2-(methyloxy)-4-[4-(2-methylpropyl)-1 - piperazinyl]phenyl}amino)-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4- yl}amino)-2-thiophenecarboxamide (323 mg, 0.239 mmol) . This material was dissolved in 1 ,4-dioxane (10 ml.) and transferred to a 20 ml microwave vessel. A solution of 6M NaOH (3 ml) and water (3 ml) was added and the reaction heated in microwave at 12O0C for 15 minutes. Resulting brown solution was diluted with ethyl acetate and washed with a saturated sodium bicarbonate solution, water and a saturated brine solution. Organics were dried over sodium sulfate and solvents removed under reduced pressure to afford a yellow solid which was purified via chromatography on SiO2 to afford 3-{[2-({2- (methyloxy)-4-[4-(2-methylpropyl)-1-piperazinyl]phenyl}amino)-1 /-/-pyrrolo[2,3- c/]pyrimidin-4-yl]amino}-2-thiophenecarboxamide (1 14 mg, 0.219 mmol) as a tan solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 0.89 (d, J=6.50 Hz, 6 H) 1.75 - 1.87 (m, 1 H) 2.10 (d, J=7.32 Hz, 2 H) 3.08 - 3.15 (m, 4 H) 3.82 (s, 3 H) 6.22 (dd, J=3.16, 1.79 Hz, 1 H) 6.45 - 6.53 (m, 1 H) 6.64 (d, J=2.29 Hz, 1 H) 6.92 - 6.99 (m, 1 H) 7.46 - 7.64 (m, 3 H) 7.68 (d, J=5.40 Hz, 1 H) 7.85 (d, J=8.70 Hz, 1 H) 8.42 (d, J=5.40 Hz, 1 H) 11.27 - 11.40 (m, 2 H) **4H missing, assumed to be hidden under DMSO peak @2.5. ESIMS(M+H)+ = 521
Example 56
3-[(2-{[4-(1 ,4'-bipiperidin-1 '-yl)-2-(methyloxy)phenyl]amino}-1 H-pyrrolo[2,3- d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000246_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrochloride (600 mg, 1.39 mmol) and [4-(1 ,4'-bipiperidin-1'-yl)-2-(methyloxy)phenyl]amine (484 mg, 1.70 mmol) in THF (50 ml) was heated at 850C for 2 hrs. The reaction was diluted with ethyl acetate (100 ml) and washed with saturated NaHCO3 (100 ml). The organic layer was removed, concentrated by rotary evaporation, solids were triturated from ethyl acetate/hexanes, and dried under house vacuum. A solution of 5-{[4-(1 ,4'-bipiperidin-1'-yl)-2- (methyloxy)phenyl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo
[3,2e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one in tetrahydrofuran (50 ml), and ammonium hydroxide (28% in water, 50 ml.) was stirred at 850C overnight, the solvent was removed and dried under house vacuum. A solution of 3-({2-{[4-(1 ,4'-bipiperidin-1'- yl)-2-(methyloxy)phenyl]amino}-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin- 4-yl}amino)-2-thiophenecarboxamide in THF (25 ml_)/MeOH (25 ml.) was treated with NaOMe (0.23 g) and the resulting mixture was stirred at 7O0C for 4 hours, the solvent was removed to yield the crude product which was adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford 3-[(2-{[4-(1 ,4'-bipiperidin-1'-yl)-2- (methyloxy)phenyl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2- thiophenecarboxamide (0.078 g, 33%). ESIMS (M+H)+ = 547. 1H NMR (400 MHz, DMSOd6) δ ppm 1.68 (br. s., 7 H), 1.99 (br. s., 2 H), 2.24 (br. s., 3 H), 2.63 (br. s., 2 H), 2.93 (br. s., 2 H), 3.78 (br. s., 6 H), 6.18 (br. s., 1 H), 6.49 (br. s., 1 H), 6.61 (br. s., 1 H), 6.92 (br. s., 1 H), 7.08 (br. s., 1 H), 7.45 (d, J=19.04 Hz, 2 H), 7.64 (s., 1 H), 7.82 (s., 1 H), 8.37 (s., 1 H), 1 1.32 (s., 2 H).
Example 57: 3-[(2-{[4-(1 ,4'-bipiperidin-1l-yl)-5-methyl-2-(methyloxy)phenyl]amino}-1H- pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2 -thiophenecarboxamide
Figure imgf000247_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrochloride (600 mg, 1.39 mmol) and [4-(1 ,4'-bipiperidin-1'-yl)-5-methyl-2-(methyloxy)phenyl]amine (507 mg, 1.70 mmol) in THF (50 ml) was heated at 850C for 0.5 hrs. The reaction was diluted with ethyl acetate (100 ml) and washed with saturated NaHCO3 (100 ml). The organic layer was removed, concentrated by rotary evaporation, solids triturated from ethyl acetate/hexanes, and dried under house vacuum prior to next step. A solution of 5-{[4- (1 ,4'-bipiperidin-1'-yl)-5-methyl-2-(methyloxy)phenyl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one in tetrahydrofuran (50 ml) and 27% ammonium hydroxide (50 ml.) was stirred at 850C overnight. The solvent was removed and dried under house vacuum. A solution of 3-({2- {[4-(1 ,4'-bipiperidin-1'-yl)-5-methyl-2-(methyloxy)phenyl]amino}-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2-thiophenecarboxamide in THF (25 ml_)/MeOH (25 mL) was treated with NaOMe (0.82g) and the resulting mixture was stirred at 7O0C for 4 hours. The solvent was removed to yield the crude product which was adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford 3-[(2-{[4-(1 ,4'-bipiperidin-1 '-yl)-5-methyl-2-(methyloxy)phenyl]amino}-1 H- pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (0.12 g, 42%). ESIMS (M+H)+ = 561. 1H NMR (400 MHz, DMSOd6) δ ppm 1.58 (br. s., 8 H), 1.91 (br. s., 3 H), 2.21 (s, 3 H), 2.29 (s, 3 H), 2.65 (br. s., 4 H), 3.10 (br. s., 2 H), 3.81 (s, 2 H), 6.24 (dd, J=3.44, 1.88 Hz, 1 H), 6.71 (s, 1 H), 6.99 (dd, J=3.30, 2.29 Hz, 1 H), 7.11 (d, J=7.88 Hz, 1 H), 7.49 (d, J=3.48 Hz, 2 H), 7.69 (d, J=5.50 Hz, 1 H), 7.88 (s, 1 H), 8.38 (d, J=5.32 Hz, 1 H), 11.39 (s, 2 H).
Example 58 3-[(2-{[1 -(N-ethyl-N-methylglycyl)-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino}- 1H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000248_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrochloride (400 mg, 0.95 mmol) and 1-{[ethyl(methyl)amino]acetyl}-5-(methyloxy)-2,3-dihydro-1 H-indol-6-amine (300 mg, 1.1 mmol) in THF (30 ml) was heated at 650C overnight. The reaction was diluted with ethyl acetate (100 ml) and washed with saturated NaHCO3 (100 ml). The organic layer was removed, concentrated by rotary evaporation, solids triturated from ethyl acetate/hexanes, and dried under house vacuum. A solution of 5-{[1-(N-ethyl-N-methylglycyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one was dissolved in tetrahydrofuran (50 ml) and 27% ammonium hydroxide (50 ml.) and the resulting mixture was stirred at 850C overnight. The solvent was removed and dried under house vacuum prior to next step. A solution of 3-({2-{[1-(N-ethyl-N-methylglycyl)-5- (methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-7-[(4-methylphenyl)sulfonyl]-7H- pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2-thiophenecarboxamide in THF (25 ml.) and MeOH (5 ml.) was treated with NaOMe (0.12g) and the resulting mixture was stirred at 7O0C for 2 hours. The solvent was removed to yield the crude product. Next, the crude product was adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford 3-[(2- {[1 -(N-ethyl-N-methylglycyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-1 H- pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (0.030 g, 49%). ESIMS (M+H)+ = 521. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.01 (t, 3 H), 2.24 (s, 3 H), 2.43 - 2.49 (m, 2 H), 3.13 (t, J=8.06 Hz, 2 H), 3.25 (s, 2 H), 3.78 (s, 3 H), 4.21 (t, J=8.19 Hz, 2 H), 6.23 ( s., 1 H), 6.97 (s, 2 H), 7.61 (d, J=5.31 Hz, 3 H), 7.71 (s, 1 H), 8.40 (d, J=5.31 Hz, 1 H), 8.58 (s, 1 H), 1 1.30 ( s., 1 H), 11.41 (s, 1 H).
Example 59
3-[(2-{[5-(methyloxy)-1 -(N-methyl-N-propylglycyl)-2,3-dihydro-1 H-indol-6-yl]amino}- 1H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000249_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrochloride (0.5 mg, 1.20 mmol) and 5-(methyloxy)-1-{[methyl(propyl)amino]acetyl}-2,3-dihydro-1 H-indol-6-amine (350 mg, 1.4 mmol) in THF (30 ml) was heated at 650C overnight. The reaction was diluted with ethyl acetate (100 ml) and washed with saturated NaHCOs (100 ml). Organic layer was removed, concentrated by rotary evaporation, solids triturated from ethyl acetate/hexanes, and dried under house vacuum. A solution of 5-{[5-(methyloxy)-1 -(N-methyl-N-propylglycyl)-2,3-dihydro-1 H-indol-6-yl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one in tetrahydrofuran (35 ml) and 27% ammonium hydroxide (40 ml.) and the resulting mixture was stirred at 850C overnight, The solvent was removed and dried under house vacuum. A solution of 3-({2-{[5-(methyloxy)-1-(N-methyl-N-propylglycyl)-2,3-dihydro-1 H- indol-6-yl]amino}-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2- thiophenecarboxamide in THF (25 ml.) and MeOH (5 ml.) was treated with NaOMe (0.12g) and the resulting mixture was stirred at 7O0C for 2 hours. The solvent was removed to yield the crude product which was adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford 3-[(2-{[5-(methyloxy)-1-(N-methyl-N-propylglycyl)- 2,3-dihydro-1 H-indol-6-yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2- thiophenecarboxamide (0.11 g, 43%). ESIMS (M+H)+ = 535. 1H NMR (400 MHz, DMSO-de) δ ppm 0.81 - 0.91 (m, 3 H), 1.52 (d, J=6.50 Hz, 2 H), 2.38 - 2.47 (m, 3 H), 2.56 - 2.67 (m, 2 H), 3.16 (s., 2 H), 3.60 (br. s., 2 H), 3.78 (s, 3 H), 4.18 (t, J=7.78 Hz, 2 H), 6.23 (br. s., 1 H), 6.98 (d, J=7.33 Hz, 2 H), 7.62 (d, J=5.04 Hz, 3 H), 7.73 (s, 1 H), 8.40 (d, J=5.13 Hz, 1 H), 8.57 (s, 1 H), 11.29 (s., 1 H), 1 1.42 (s., 1 H).
Example 60
3-[(2-{[1-{N-methyl-N-[2-(methyloxy)ethyl]glycyl}-5-(methyloxy)-2,3-dihydro-1 H- indol-6-yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000251_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrochloride (0.45 mg, 1.0 mmol) and 1 -({methyl[2-(methyloxy)ethyl]amino}acetyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6- amine (350 mg, 1.2 mmol) in THF (30 ml) was heated at 7O0C overnight. The reaction was diluted with ethyl acetate (100 ml) and washed with saturated NaHCOs (100 ml). The organic layer was removed, concentrated by rotary evaporation, solids triturated from ethyl acetate/hexanes, and dried under house vacuum. A solution of 5-{[1-{N- methyl-N-[2-(methyloxy)ethyl]glycyl}-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-3- [(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)- one in tetrahydrofuran (35 ml) and 27% ammonium hydroxide (4OmL) was stirred at 850C overnight. The solvent was removed and dried under house vacuum. A solution of 3-({2-{[1-{N-methyl-N-[2-(methyloxy)ethyl]glycyl}-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2- thiophenecarboxamide in THF (25 ml.) and MeOH (5 ml.) was treated with NaOMe (0.3Og) and the resulting mixture was stirred at 7O0C for 2 hours. The solvent was removed to yield the crude product which was adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford 3-[(2-{[1-{N-methyl-N-[2-(methyloxy)ethyl]glycyl}-5- (methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2- thiophenecarboxamide (0.16 g, 51%). ESIMS (M+H)+ = 551. 1H NMR (400 MHz, DMSO- d6) δ ppm 2.30 ( s., 3 H), 2.65 ( s., 2 H), 3.07 - 3.17 (m, 2 H), 3.22 ( s., 3 H), 3.32 (br. s., 2 H), 3.43 (br. s., 2 H), 3.78 (s., 3 H), 4.18 (s., 2 H), 6.24 ( s., 1 H), 6.96 ( s., 2 H), 7.52 - 7.65 (m, 3 H), 7.70 ( s., 1 H), 8.41 (s., 1 H), 8.59 ( s., 1 H), 11.31 ( s., 1 H), 1 1.42 ( s., 1 H).
Example 61 3-[(2-{[1 -[(3-hydroxy-1 -piperidinyl)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrochloride (0.45 mg, 1.0 mmol) and 1-{2-[6-amino-5-(methyloxy)-2,3-dihydro-1 H-indol-1-yl]-2-oxoethyl}-3-piperidinol (350 mg, 1.1 mmol) in THF (30 ml) was heated at 7O0C overnight. The reaction was diluted with ethyl acetate (100 ml) and washed with saturated NaHCO3 (100 ml). The organic layer was removed, concentrated by rotary evaporation, solids triturated from ethyl acetate/hexanes, and dried under house vacuum. A solution of
5-{[1-[(3-hydroxy-1-piperidinyl)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-3- [(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)- one in tetrahydrofuran (35 ml) and 27% ammonium hydroxide (40 ml.) was stirred at 850C overnight. The solvent was removed and the residue was dried under house vacuum. A solution of 3-({2-{[1-[(3-hydroxy-1-piperidinyl)acetyl]-5-(methyloxy)-2,3- dihydro-1 H-indol-6-yl]amino}-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4- yl}amino)-2-thiophenecarboxamide in THF (2ml_), water (2ml_) and MeOH (5 ml.) was treated with K2CO3 (0.8Og) and the resulting mixture was stirred at 8O0C for 3 hours. The K2CO3 was filtered out and the solvent was removed to yield the crude product which was adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford 3-[(2-{[1 -[(3-hydroxy-1 -piperidinyl)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}- 1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (0.10 g, 61%). ESIMS (M+H)+ = 563. 1H NMR (400 MHz, DMSOd6) δ ppm 1.06 (d, 1 H), 1.42 (d, J=1 1.90 Hz, 1 H), 1.61 (d, J=13.19 Hz, 1 H), 1.78 (d, J=8.79 Hz, 1 H), 1.91 (t, J=9.71 Hz, 1 H), 2.02 (t, J=10.26 Hz, 1 H), 2.69 (d, J=10.62 Hz, 1 H), 2.89 (d, J=7.05 Hz, 1 H), 3.12 (t, J=8.19 Hz, 2 H), 3.23 (d, J=5.77 Hz, 2 H), 3.48 (dd, J=8.84, 4.44 Hz, 1 H), 3.78 (s, 3 H), 4.20 (t, J=6.78 Hz, 2 H), 4.63 (d, J=4.76 Hz, 1 H), 6.24 ( s., 1 H), 6.96 (s, 2 H), 7.62 (d, J=5.40 Hz, 3 H), 7.70 (s, 1 H), 8.42 (d, J=5.31 Hz, 1 H), 8.60 (s, 1 H), 11.31 ( s., 1 H), 1 1.42 (s, 1 H).
Example 62
3-[(2-{[1 -[(3-hydroxy-1 -pyrrolidinyl)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000253_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrochloride (0.45 mg, 1.0 mmol) and 1-{2-[6-amino-5-(methyloxy)-2,3-dihydro-1 H-indol-1-yl]-2-oxoethyl}-3-pyrrolidinol (330 mg, 1.1 mmol) in THF (30 ml) was heated at 7O0C overnight. The reaction was diluted with ethyl acetate (100 ml) and washed with saturated NaHCO3 (100 ml). The organic layer was removed, concentrated by rotary evaporation, solids triturated from ethyl acetate/hexanes, and dried under house vacuum prior to next step. Next , a solution of 5-{[1 -[(3-hydroxy-1 -pyrrolidinyl)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2- c]pyrimidin-7(3H)-one in tetrahydrofuran (35 ml), followed by ammonium hydroxide (28% in water), and the resulting mixture was stirred at 850C overnight, the solvent was removed and dried under house vacuum. A solution of 3-({2-{[1-[(3-hydroxy-1- pyrrolidinyl)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2-thiophenecarboxamide in THF (2 ml_), water (2 ml_), and MeOH (5 ml.) was treated withK2CO3 (0.8Og) and the resulting mixture was stirred at 8O0C for 3 hours. The K2CO3 was filtered out, and the solvent was removed to yield the crude product which was adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford 3-[(2-{[1-[(3-hydroxy-1- pyrrolidinyl)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-1 H-pyrrolo[2,3- d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (0.09 g, 57%). ESIMS (M+H)+ = 549. 1H NMR (400 MHz, DMSOd6) δ ppm 1.55 (dd, 1 H), 1.98 (dd, J=13.05, 6.91 Hz, 1 H), 2.45 (dd, J=9.52, 3.20 Hz, 1 H), 2.53 - 2.61 (m, 1 H), 2.71 (q, J=7.60 Hz, 1 H), 2.83 (dd, J=9.39, 6.18 Hz, 1 H), 3.14 (q, J=8.12 Hz, 2 H), 3.37 (br. s., 3 H), 3.78 (s, 3 H), 4.17 (t, J=8.38 Hz, 2 H), 4.73 (d, J=4.21 Hz, 1 H), 6.24 (br. s., 1 H), 6.96 (s, 2 H), 7.51 - 7.65 (m, 3 H), 7.70 (s, 1 H), 8.41 (d, J=5.40 Hz, 1 H), 8.59 (s, 1 H), 11.31 ( s., 1 H), 1 1.41 (s, 1 H).
Example 63
3-[(2-{[1 -[(3-hydroxy-1 -pyrrolidinyl)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-N-methyl-2- thiophenecarboxamide
Figure imgf000254_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrochloride (0.45 mg, 1.0 mmol) and 1-{2-[6-amino-5-(methyloxy)-2,3-dihydro-1 H-indol-1-yl]-2-oxoethyl}-3-pyrrolidinol (330 mg, 1.1 mmol) in THF (30 ml) was heated at 7O0C overnight. The reaction was diluted with ethyl acetate (100 ml) and washed with saturated NaHCOs (100 ml). The organic layer was removed, concentrated by rotary evaporation, solids triturated from ethyl acetate/hexanes, and dried under house vacuum. Next, a solution of 5-{[1-[(3- hydroxy-1-pyrrolidinyl)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one in tetrahydrofuran (35 ml) and methyl amine (2M in THF, 30 ml.) was stirred at room temperature overnight. The solvent was removed and the resulting residue was dried under house vacuum. A solution of 3-({2-{[1-[(3-hydroxy-1-pyrrolidinyl)acetyl]-5- (methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-7-[(4-methylphenyl)sulfonyl]-7H- pyrrolo[2,3-d]pyrimidin-4-yl}amino)-N-methyl-2-thiophenecarboxamide in THF (20 ml.) was treated with NaOMe (0.4Og) and stirred at 7O0C for 2 hours. The solvent was removed and the residue was redissolved in 50 ml of DCM and washed with 50 ml of sat. NH4CI. The organic solvent was removed under reduced pressure to yield the crude product which was adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford 3-[(2-{[1 -[(3-hydroxy-1 -pyrrolidinyl)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-N-methyl-2-thiophenecarboxamide (0.056 g, 21% ). ESIMS (M+H)+ = 563. 1H NMR (400 MHz, DMSOd6) δ ppm 1.56 (dd, 1 H), 1.98 (dd, J=12.82, 7.05 Hz, 1 H), 2.46 (dd, J=9.61 , 3.20 Hz, 1 H), 2.54 - 2.63 (m, 1 H), 2.68 - 2.75 (m, 1 H), 2.79 (d, J=4.39 Hz, 3 H), 2.85 (d, J=4.21 Hz, 1 H), 3.13 (t, J=8.06 Hz, 2 H), 3.38 (d, J=6.59 Hz, 3 H), 3.78 (s, 3 H), 4.16 (t, J=8.38 Hz, 2 H), 4.74 (br. s., 1 H), 6.27 (br. s., 1 H), 6.96 (s, 2 H), 7.60 (d, J=5.40 Hz, 1 H), 7.70 (s, 1 H), 8.13 (d, J=4.49 Hz, 1 H), 8.42 (d, J=5.31 Hz, 1 H), 8.59 (s, 1 H), 1 1.32 (d, J=9.71 Hz, 2 H).
Example 64
3-[(2-{[1 -[(3-hydroxy-1 -piperidinyl)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-N-methyl-2- thiophenecarboxamide
Figure imgf000256_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrochloride (0.45 mg, 1.0 mmol) and 1-{2-[6-amino-5-(methyloxy)-2,3-dihydro-1 H-indol-1-yl]-2-oxoethyl}-3-piperidinol (350 mg, 1.1 mmol) in THF (30 ml) was heated at 7O0C overnight. The reaction was diluted with ethyl acetate (100 ml) and washed with saturated NaHCO3 (100 ml). The organic layer was removed, concentrated by rotary evaporation, solids triturated from ethyl acetate/hexanes, and dried under house vacuum. A solution of 5-{[1-[(3-hydroxy-1- piperidinyl)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one in tetrahydrofuran (35 ml) and methyl amine (2M in THF, 30 ml.) was stirred at room temperature overnight. The solvent was removed and dried under house vacuum. A solution of 3-({2-{[1-[(3-hydroxy-1-piperidinyl)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol- 6-yl]amino}-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-N- methyl-2 thiophenecarboxamide in THF (2 ml_), water (2 ml_), and MeOH (5 ml.) was treated with K2CO3 (0.6Og) and the resulting mixture was stirred at 8O0C for 3 hours. The K2CO3 was filtered off and the solvent was removed to yield the crude product, which was adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford 3-[(2-{[1 -[(3-hydroxy-1 -piperidinyl)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}- 1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-N-methyl-2-thiophenecarboxamide (0.16 g, 61%). ESIMS (M+H)+ = 577. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.08 ( s., 1 H), 1.42 (d, J=11.72 Hz, 1 H), 1.61 (d, J=12.91 Hz, 1 H), 1.77 (br. s., 1 H), 1.91 (t, J=9.57 Hz, 1 H), 2.02 (t, J=10.30 Hz, 1 H), 2.69 (d, J=10.71 Hz, 1 H), 2.79 (d, J=4.30 Hz, 3 H), 2.90 (br. s., 1 H), 3.12 (t, J=8.10 Hz, 2 H), 3.23 (d, J=5.13 Hz, 2 H), 3.48 (d, J=4.67 Hz, 1 H), 3.78 (s, 3 H), 4.14 - 4.27 (m, 2 H), 4.62 (d, J=4.67 Hz, 1 H), 6.27 ( s., 1 H), 6.96 ( s., 2 H), 7.60 (d, J=5.31 Hz, 1 H), 7.70 (s, 1 H), 8.12 (d, J=4.30 Hz, 1 H), 8.42 (d, J=5.31 Hz, 1 H), 8.58 (s, 1 H), 11.31 (d, J=10.44 Hz, 2 H).
Example 65
N-methyl-3-[(2-{[1-{N-methyl-N-[2-(methyloxy)ethyl]glycyl}-5-(methyloxy)-2,3- dihydro-1 H-indol-6-yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2- thiophenecarboxamide
Figure imgf000257_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrochloride (450 mg, 1.0 mmol) and 1 -({methyl[2-(methyloxy)ethyl]amino}acetyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6- amine (350 mg, 1.2 mmol) in THF (30 ml) was heated at 7O0C overnight. The reaction was diluted with ethyl acetate (100 ml) and washed with saturated NaHCO3 (100 ml). The organic layer was removed, concentrated by rotary evaporation, solids triturated from ethyl acetate/hexanes, and dried under house vacuum. A solution of 5-{[1-{N- methyl-N-[2-(methyloxy)ethyl]glycyl}-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-3- [(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)- one in tetrahydrofuran (35 ml) and methyl amine (2M in THF, 30 ml.) was stirred at room temperature overnight. The solvent was removed and dried under house vacuum prior. A solution of N-methyl-3-({2-{[1-{N-methyl-N-[2-(methyloxy)ethyl]glycyl}-5-(methyloxy)- 2,3-dihydro-1 H-indol-6-yl]amino}-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin- 4-yl}amino)-2-thiophenecarboxamide THF (20 ml.) and MeOH (20 ml.) was treated with NaOMe (0.4g) and stirred at 7O0C for 2 hours. The solvent was removed, the residue was redissolved in 50 ml of DCM, washed with 50 ml of sat.NH4CI, and the organic solvent was removed to yield the crude product which was adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford N-methyl-3-[(2-{[1-{N-methyl-N-[2- (methyloxy)ethyl]glycyl}-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-1 H-pyrrolo[2,3- d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (0.15 g, 38%). ESIMS (M+H)+ = 565. 1H NMR (400 MHz, DMSOd6) δ ppm 2.30 (s, 3 H), 2.65 (t, J=5.59 Hz, 2 H), 2.79 (d, J=4.21 Hz, 2 H), 2.93 - 3.00 (m, 1 H), 3.12 (t, J=7.97 Hz, 1 H), 3.22 (d, J=3.57 Hz, 3 H), 3.27 (s, 1 H), 3.32 (s, 1 H), 3.36 (br. s., 1 H), 3.43 (d, J=2.66 Hz, 2 H), 3.71 (s, 1 H), 3.78 (s, 2 H), 4.02 - 4.09 (m, 1 H), 4.18 (t, J=8.06 Hz, 1 H), 6.28 (s., 1 H), 6.96 (br. s., 2 H), 7.56 (s, 1 H), 7.60 (d, J=5.31 Hz, 1 H), 7.70 (s, 1 H), 8.42 (d, J=5.40 Hz, 1 H), 8.60 (s, 1 H), 11.31 ( s., 1 H), 11.34 (s, 1 H)
Example 66
N-methyl-3-[(2-{[5-(methyloxy)-1 -(N-methyl-N-propylglycyl)-2,3-dihydro-1H-indol-6- yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000258_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrochloride (500mg, 1.2 mmol) and 5-(methyloxy)-1-{[methyl(propyl)amino]acetyl}-2,3-dihydro-1 H-indol-6-amine (380 mg, 1.4 mmol) in THF (30 ml) was heated at 7O0C overnight. The reaction was diluted with ethyl acetate (100 ml) and washed with saturated NaHCOs (100 ml). The organic layer was removed, concentrated by rotary evaporation, solids triturated from ethyl acetate/hexanes, and dried under house vacuum. A solution of 5-{[5-(methyloxy)-1-(N-methyl-N-propylglycyl)-2,3-dihydro-1 H-indol-6-yl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one in tetrahydrofuran (35 ml) and methyl amine (2M in THF, 30 ml.) was stirred at room temperature overnight. The solvent was removed and the resulting residue was dried under house vacuum. Next, a solution of N-methyl-3-({2-{[5-(methyloxy)-1-(N-methyl-N- propylglycyl)-2,3-dihydro-1 H-indol-6-yl]amino}-7-[(4-methylphenyl)sulfonyl]-7H- pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2-thiophenecarboxamide in THF (20 mL) and MeOH (20 mL) was treated with sodium methoxide (0.3Og) stirred at 7O0C for 2 hours. The solvent was removed, the residue was redissolved in 50 ml of DCM, washed with 50 ml of sat. NH4CI, and the organic solvent was removed to yield the crude product which was adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford N-methyl-3-[(2-{[5-(methyloxy)-1-(N-methyl-N-propylglycyl)-2,3- dihydro-1 H-indol-6-yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-2- thiophenecarboxamide (0.22 g, 81%). ESIMS (M+H)+ = 549. 1H NMR (400 MHz, DMSOd6) δ ppm 0.84 (t, J=7.23 Hz, 3 H), 1.38 - 1.49 (m, 2 H), 2.23 (s, 3 H), 2.38 (t, J=7.10 Hz, 2 H), 2.79 (d, J=4.21 Hz, 3 H), 3.13 (t, J=8.10 Hz, 1 H), 3.19 (s, 1 H), 3.24 (s, 1 H), 3.35 (s, 2 H), 3.71 (s, 1 H), 3.78 (s, 2 H), 4.21 (t, J=8.19 Hz, 1 H), 6.27 (br. s., 1 H), 6.96 (s, 2 H), 7.59 (d, J=5.31 Hz, 1 H), 7.70 (s, 1 H), 8.12 (d, J=4.30 Hz, 1 H), 8.41 (d, J=5.31 Hz, 1 H), 8.58 (s, 1 H), 11.32 (d, J=13.09 Hz, 2 H).
Example 67
3-[(2-{[1 -(N-ethyl-N-methylglycyl)-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino}- 1H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-N-methyl-2 -thiophenecarboxamide
Figure imgf000259_0001
A suspension of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrochloride (400 mg, 0.9 mmol) and 1-{[ethyl(methyl)amino]acetyl}-5-(methyloxy)-2,3-dihydro-1 H-indol-6-amine (300 mg, 1.1 mmol) in THF (30 ml) was heated at 7O0C overnight. The reaction was diluted with ethyl acetate (100 ml) and washed with saturated NaHCO3 (100 ml). The organic layer was removed, concentrated by rotary evaporation, solids triturated from ethyl acetate/hexanes, and dried under house vacuum. A solution of 5-{[1-(N-ethyl-N-methylglycyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one in tetrahydrofuran (35 ml) and methyl amine (2M in THF, 50 ml.) was stirred at room temperature overnight. The solvent was removed and dried under house vacuum. A solution of 3-({2-{[1 -(N-ethyl-N-methylglycyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-N-methyl- 2-thiophenecarboxamide in THF (20 ml.) and MeOH (20 ml.) was treated with sodium methoxide (0.35g) and stirred at 7O0C for 2 hours. The solvent was removed, the residue was redissolved in 50 ml of DCM, washed with 50 ml of sat.NH4CI, and the organic solvent was removed to yield the crude product which was adsorbed to silica gel and purified by LC (DCM to 10% MeOH/DCM) to afford 3-[(2-{[1-(N-ethyl-N-methylglycyl)-5- (methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-N- methyl-2-thiophenecarboxamide (0.20 g, 57%). ESIMS (M+H)+ = 535. 1H NMR (400 MHz, DMSO-de) δ ppm 0.98 (t, J=7.00 Hz, 3 H), 2.20 (s, 3 H), 2.47 (dd, J=4.17, 2.06 Hz, 2 H), 2.76 (d, J=4.40 Hz, 3 H), 3.10 (t, J=8.15 Hz, 2 H), 3.21 (s, 2 H), 3.75 (s, 3 H), 4.18 (t, J=8.19 Hz, 2 H), 6.25 (br. s., 1 H), 6.93 (s, 2 H), 7.56 (d, J=5.40 Hz, 1 H), 7.67 (s, 1 H), 8.09 (d, J=4.30 Hz, 1 H), 8.39 (d, J=5.40 Hz, 1 H), 8.57 (s, 1 H), 11.29 (d, J=13.09 Hz, 2 H)
Example 68
3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}- 1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000261_0001
A mixture of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido [1 ,2-c]pyrimidin-7(3/-/)-one hydrochloride (1.00 g, 2.14 mmol) and 1-[(dimethylamino) acetyl]-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinamine (700 mg, 2.66 mmol) was dissolved in 2,2,2-trifluoroethanol (30 ml.) and heated in a sealed tube overnight. The reaction was concentrated, added to a sealed tube, diluted with tetrahydrofuran (20 ml_), treated with excess 28% aqueous ammonia (25 ml_), and stirred overnight at 90 0C. The reaction was cooled, the organic layer was separated, dried over sodium sulfate, filtered, concentrated, and subjected to purification via flash silica gel chromatography (0 to 20% methanol/dichloromethane, with -1 % ammonium hydroxide). The product obtained, 3- ({2-{[1-(Λ/,Λ/-dimethylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2-thiophenecarboxamide, (ESIMS (M+H)+ = 675), was dissolved in THF (10 ml_), 1 ,4-dioxane (30 ml_), and treated with excess potassium hydroxide (3.0 g dissolved in 5 ml. of water), and stirred at 100 0C for 6 h. The reaction was cooled, diluted with water and EtOAc, and transferred to a separatory funnel and shaken. Brine was added to assist in the separation of layers. The organic layer was separated, dried over sodium sulfate, filtered, and subjected to purification via flash silica gel column chromatography (0 to 20% methanol/dichloromethane, spiked with -1 % ammonium hydroxide). Relevant fractions were combined, concentrated, and suspended in diethyl ether/ethyl acetate. The solids obtained were filtered to obtain 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-6-(methyloxy)-1 ,2,3,4- tetrahydro-7-quinolinyl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2- thiophenecarboxamide (259 mg, 0.47 mmol, 21 % over 3 steps). 1H NMR (400 MHz, DMSOd6) δ ppm 1.83 - 1.94 (m, 2 H), 2.12 (s, 6 H), 2.64 - 2.75 (m, 2 H), 3.25 (s, 2 H), 3.67 - 3.77 (m, 2 H), 3.84 (s, 3 H), 6.21 - 6.28 (m, 1 H), 6.84 (s, 1 H), 7.01 (s, 1 H), 7.51 - 7.70 (m, 4 H), 8.33 - 8.44 (m, 2 H), 1 1.36 - 1 1.46 (m, 2 H); ESIMS (M+H)+ = 521.
Example 69
3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-4,4-dimethyl-6-(methyloxy)-1,2,3,4-tetrahydro-7- quinolinyl]amino}-1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000262_0001
A mixture of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido [1 ,2-c]pyrimidin-7(3/-/)-one hydrochloride (90% pure, 603 mg, 1.16 mmol) and 1-
[(dimethylamino) acetyl]-4,4-dimethyl-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinamine (367 mg, 1.26 mmol) was dissolved in 2,2,2-trifluoroethanol (25 ml.) and refluxed for 3 h. The reaction was cooled, diluted with excess saturated aqueous sodium bicarbonate, and then extracted with dichloromethane. The organic layer was separated, dried over magnesium sulfate, filtered, and concentrated. The crude material 5-{[1-(Λ/,Λ/- dimethylglycyl)-4,4-dimethyl-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo [3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one was divided approximately into two equal parts. One part (assumed -0.58 mmol) was taken in a sealed tube, diluted with tetrahydrofuran (10 ml_), treated with excess 28% aqueous ammonia (10 ml_), and stirred overnight at 50 0C. The reaction was cooled, the organic layer was separated, dried over magnesium sulfate, filtered, concentrated, and subjected to purification via flash silica gel chromatography (dichloromethane to 8:1 :1 mixture of dichloromethane:methanol:2M ammonia in methanol). The product obtained, 3-({2-{[1-(Λ/,Λ/-dimethylglycyl)-4,4- dimethyl-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2-thiophenecarboxamide, was dissolved in THF (10 ml_), MeOH (10 ml_), treated with excess sodium methoxide (200 mg, 3.70 mmol), and stirred at 70 0C for 2 h. The reaction was cooled, diluted with water and EtOAc, and transferred to a separatory funnel and shaken. Brine was added to assist in the separation of layers. The organic layer was separated, dried over sodium sulfate, filtered, and subjected to purification via flash silica gel column chromatography (dichloromethane to 8:1 :1 mixture of dichloromethane:methanol :2M ammonia in methanol). Relevant fractions were combined, concentrated, suspended in diethyl ether/ethyl acetate. The solids were filtered to obtain 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-4,4- dimethyl-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin- 4-yl)amino]-2-thiophenecarboxamide (1 10 mg, 0.200 mmol, -34% over three steps). 1H NMR (400 MHz, DMSO-d6) δ ppm 1.29 (s, 6 H), 1.69 - 1.77 (m, 2 H), 2.1 1 (s, 6 H), 3.27 (s, 2 H), 3.73 - 3.80 (m, 2 H), 3.88 (s, 3 H), 6.25 (s, 1 H), 6.96 (s, 1 H), 7.02 (s, 1 H), 7.51 - 7.72 (m, 4 H), 8.34 (s, 1 H), 8.39 (d, J=5.3 Hz, 1 H), 1 1.42 (s, 2 H); ESIMS (M+H)+ = 549.
Example 70
3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-4,4-dimethyl-6-(methyloxy)-1,2,3,4-tetrahydro-7- quinolinyl]amino}-1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2- thiophenecarboxamide
Figure imgf000263_0001
A part of the crude material 5-{[1-(Λ/,Λ/-dimethylglycyl)-4,4-dimethyl-6-(methyloxy)- 1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo [3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one (assumed -0.58 mmol) obtained from Example 69 was taken in a sealed tube, diluted with tetrahydrofuran (10 ml_), treated with excess methyl amine (10 ml_, 20 mmol, 2M in THF), and stirred overnight at 50 0C. The reaction was cooled, concentrated, and subjected to purification via flash silica gel chromatography (dichloromethane to 8:1 :1 mixture of dichloromethane:methanol:2M ammonia in methanol). The product obtained, 3-({2-{[1- (Λ/,Λ/-dimethylglycyl)-4,4-dimethyl-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}- 7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-Λ/-methyl-2- thiophenecarboxamide, was characterized by LC (ESIMS (M+H)+ = 717), dissolved in THF (3 ml_), MeOH (6 ml_), treated with excess sodium methoxide (200 mg, 3.70 mmol), and stirred at 70 0C for 2 h. The reaction was cooled, diluted with water, EtOAc, and transferred to a separatory funnel and shaken. Brine was added to assist in the separation of layers. The organic layer was separated, dried over sodium sulfate, filtered, and subjected to purification via flash silica gel column chromatography (dichloromethane to 8:1 :1 mixture of dichloromethane:methanol:2M ammonia in methanol). Relevant fractions were combined, concentrated, and subjected to further purification under reverse phase HPLC (10 to 70% acetonitrile/water with 0.05% trifluoroacetic acid). Relevant fractions were collected and treated with excess saturated aqueous sodium bicarbonate and extracted with dichloromethane. The organic layer was dried over magnesium sulfate, filtered, concentrated, suspended in diethyl ether/ethyl acetate. The solids obtained were filtered to obtain 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-4,4- dimethyl-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin- 4-yl)amino]-Λ/-methyl-2-thiophenecarboxamide (29 mg, 0.05 mmol, -9% over three steps) 1H NMR (400 MHz, DMSO-d6) δ ppm 1.29 (s, 6 H), 1.68 - 1.78 (m, 2 H), 2.1 1 (s, 6 H), 2.79 (d, J=4.6 Hz, 3 H), 3.28 (s, 2 H), 3.73 - 3.80 (m, 2 H), 3.88 (s, 3 H), 6.29 (s, 1 H), 6.96 (s, 1 H), 7.02 (s, 1 H), 7.63 - 7.68 (m, 2 H), 8.13 - 8.19 (m, 1 H), 8.32 - 8.35 (m, 1 H), 8.39 (d, J=5.3 Hz, 1 H), 11.33 (s, 1 H), 11.42 (br. s., 1 H); ESIMS (M+H)+ = 563.
Example 71
3-[(2-{[6-(methyloxy)-1 -(1 -pyrrolidinylacetyl)-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000265_0001
A mixture of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido [1 ,2-c]pyrimidin-7(3/-/)-one hydrochloride (730 mg, 2.08 mmol) and 6-(methyloxy)-1-(1- pyrrolidinylacetyl)-1 ,2,3,4-tetrahydro-7-quinolinamine (730 mg, 2.52 mmol) were mixed in tetrahydrofuran (-25 ml.) and heated at 90 0C in a sealed tube overnight. The reaction was concentrated, the crude material was diluted with tetrahydrofuran (-20 ml), treated with excess 28% aqueous ammonia (-25 ml_), and heated in a sealed tube overnight at 80 0C. The mixture was cooled; the organic layer was separated, dried over sodium sulfate, and subjected to purification by flash silica gel chromatography (0 to 20% methanol/dichloromethane, spiked with -1 % ammonium hydroxide). Relevant fractions were mixed and concentrated; the solids obtained were triturated with dichloromethane/ hexanes/diethylether. The mixture was filtered and the residue, 3-({2-{[6-(methyloxy)-1- (1-pyrrolidinylacetyl)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-7-[(4-methylphenyl)sulfonyl]- 7/-/-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2-thiophenecarboxamide (ESIMS (M+H)+ = 701 , 95% pure). The residue was diluted with tetrahydrofuran (15 ml_), methanol (20 ml_), treated with excess sodium methoxide (1.4 g, 26 mmol) and stirred at 80 0C for 2 h. The reaction was cooled, diluted with water and EtOAc. Brine was added to assist in the separation of layers. The organic layer was separated, dried over sodium sulfate, filtered, adsorbed on silica gel and purified by flash silica gel chromatography (0 to 20% methanol/dichloromethane, with -1 % ammonium hydroxide). Fractions, which seemed clean were concentrated, triturated with dichloromethane/hexanes, and filtered. 1 H NMR of the solids obtained revealed the presence of an impurity (-35%) which was presumed to be p-toluenesulfonic acid. The solids were taken up in dichloromethane with slight amounts of methanol, washed with 4 N aqueous sodium hydroxide, and the aqueous layer was back-extracted with THF (brine was added to assist in separation of layers). The organic layers were combined, dried over magnesium sulfate, and concentrated. 1 H NMR of the solids obtained revealed that p-tosic acid (~5%) was still present. At this point the solids were dissolved in methanol/dichloromethane, adsorbed over silica gel, purified via flash silica gel chromatography (0 to 20% 2M ammonia in MeOHZCH2CI2). Relevant fractions were concentrated and the solids obtained triturated to provide 3-[(2-{[6-(methyloxy)-1 -(1 -pyrrolidinylacetyl)-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide (250 mg, 0.46 mmol, 22% over three steps). 1H NMR (400 MHz, DMSO-c/6) δ ppm 1.45 - 1.57 (m, 4 H), 1.84 - 1.94 (m, 2 H), 2.36 - 2.48 (m, 4 H), 2.70 (s, 2 H), 3.35 - 3.46 (m, 2 H), 3.69 - 3.76 (m, 2 H), 3.85 (s, 3 H), 6.26 (s, 1 H), 6.85 (s, 1 H), 7.01 (s, 1 H), 7.62 (s, 4 H), 8.38 - 8.49 (m, 2 H), 11.37 - 1 1.47 (m, 2 H); ESIMS (M+H)+ = 547.
Example 72
3-[(2-{[4,4-dimethyl-6-(methyloxy)-1 -(1 -pyrrol idinylacetyl)-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000266_0001
A mixture of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido [1 ,2-c]pyrimidin-7(3H)-one hydrochloride (90% pure, 600 mg, 1.16 mmol) and 4,4- dimethyl-6-(methyloxy)-1-(1-pyrrolidinylacetyl)-1 ,2,3,4-tetrahydro-7-quinolinamine (395 mg, 1.24 mmol) was taken in 2,2,2-trifluoroethanol (25 ml.) and refluxed for 3 h. The reaction was cooled, diluted with excess saturated aqueous sodium bicarbonate, and then extracted with dichloromethane. The organic layer was separated, dried over magnesium sulfate, filtered, and concentrated. The crude material 5-{[4,4-dimethyl-6- (methyloxy)-i -(1 -pyrrolidinylacetyl)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one was divided approximately into two equal parts. One part (assumed -0.58 mmol) was added to a sealed tube, diluted with tetrahydrofuran (10 ml_), treated with excess 28% aqueous ammonia (10 ml_), and stirred overnight at 50 0C. The reaction was cooled, the organic layer was separated, dried over magnesium sulfate, filtered, concentrated, and subjected to purification via flash silica gel chromatography (dichloromethane to 8:1 :1 mixture of dichloromethane:methanol:2M ammonia in methanol). The product obtained, 3-({2-{[4,4-dimethyl-6-(methyloxy)-1-(1-pyrrolidinylacetyl)-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2- thiophenecarboxamide (ESIMS (M+H)+ = 729) was dissolved in THF (3 ml_), MeOH (6 ml_), treated with excess sodium methoxide (200 mg, 3.70 mmol), and stirred at 70 0C for 2 h. The reaction was cooled, diluted with water, EtOAc, and transferred to a separatory funnel and shaken. Brine was added to assist in the separation of layers. The organic layer was separated, dried over sodium sulfate, filtered, and subjected to purification via flash silica gel column chromatography (dichloromethane to 8:1 :1 mixture of dichloromethane:methanol:2M ammonia in methanol). Relevant fractions were combined, concentrated, and suspended in diethyl ether/ethyl acetate. The solids obtained were filtered to obtain 3-[(2-{[4,4-dimethyl-6-(methyloxy)-1-(1- pyrrolidinylacetyl)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4- yl)amino]-2-thiophenecarboxamide (82 mg, 0.14 mmol, -24% over three steps). 1H NMR (400 MHz, DMSO-d6) δ ppm 1.28 (s, 6 H), 1.49 (br. s., 4 H), 1.70 - 1.76 (m, 2 H), 2.38 (br. s., 4 H), 3.36 (br. s., 2 H), 3.77 (t, J=6.1 Hz, 2 H), 3.89 (s, 3 H), 6.24 - 6.26 (m, 1 H), 6.96 (s, 1 H), 7.01 (s, 1 H), 7.53 - 7.69 (m, 4 H), 8.36 - 8.43 (m, 2 H), 11.39 - 1 1.46 (m, 2 H); ESIMS (M+H)+ = 575.
Example 73
3-[(2-{[4,4-dimethyl-6-(methyloxy)-1 -(1 -pyrrol idinylacetyl)-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2- thiophenecarboxamide
Figure imgf000268_0001
A part of the crude material 5-{[4,4-dimethyl-6-(methyloxy)-1-(1-pyrrolidinylacetyl)- 1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one (assumed -0.58 mmol-see Example 72) was added to a sealed tube, diluted with tetrahydrofuran (5 ml_), treated with excess methyl amine (10 ml_, 20 mmol, 2M in tetrahydrofuran), and stirred overnight at 50 0C. The reaction was cooled, concentrated, and subjected to purification via flash silica gel chromatography (dichloromethane to 8:1 :1 mixture of dichloromethane:methanol:2M ammonia in methanol). The product obtained, 3-({2-{[4,4-dimethyl-6-(methyloxy)-1-(1- pyrrolidinylacetyl)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-7-[(4-methylphenyl)sulfonyl]-7/-/- pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-Λ/-methyl-2-thiophenecarboxamide (ESIMS (M+H)+ = 743) was dissolved in THF (5 ml_), MeOH (10 ml_), treated with excess sodium methoxide (200 mg, 3.70 mmol), and stirred at 70 0C for 2 h. The reaction was cooled, diluted with water, EtOAc, and transferred to a separatory funnel and shaken. Brine was added to assist in the separation of layers. The organic layer was separated, dried over sodium sulfate, filtered, and subjected to purification via flash silica gel column chromatography (dichloromethane to 8:1 :1 mixture of dichloromethane:methanol:2M ammonia in methanol). Relevant fractions were combined, concentrated, and subjected to further purification under reverse phase HPLC (10 to 70% acetonitrile/water with 0.05% trifluoroacetic acid). Relevant fractions were collected, treated with excess potassium carbonate, and extracted with dichloromethane. The organic layer was dried over magnesium sulfate, filtered, concentrated, suspended in diethyl ether/ethyl acetate. The solids obtained were filtered to obtain 3-[(2-{[4,4-dimethyl-6-(methyloxy)-1-(1- pyrrolidinylacetyl)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-1 H-pyrrolo[2,3-c/]pyrimidin-4- yl)amino]-Λ/-methyl-2-thiophenecarboxamide (43 mg, 0.07 mmol, -13% over three steps). 1H NMR (400 MHz, DMSOd6) δ ppm 1.28 (s, 6 H), 1.50 (br. s., 4 H), 1.69 - 1.75 (m, 2 H), 2.40 (br. s., 4 H), 2.79 (d, J=4.4 Hz, 3 H), 3.38 (br. s, 2 H), 3.73 - 3.81 (m, 2 H), 3.89 (s, 3 H), 6.27 - 6.31 (m, 1 H), 6.95 - 7.03 (m, 2 H), 7.62 - 7.66 (m, 2 H), 8.16 (s, 1 H), 8.35 - 8.43 (m, 2 H), 11.34 (s, 1 H), 11.43 (br. s., 1 H); ESIMS (M+H)+ = 589.
Example 74
3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino}-1H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-pyridinecarboxamide
Figure imgf000269_0001
Step A/Intermediate C16: 3-({2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1/-/- indol-6-yl]amino}-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2- pyridinecarboxamide
Figure imgf000269_0002
A solution of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrido[3,2- d]pyrrolo[2',3':4,5]pyrimido[1 ,6-a]pyrimidin-7(3H)-one hydrogen chloride (69 mg, 0.149 mmol) and 1-[(dimethylamino)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-amine (37.2 mg, 0.149 mmol) in THF (6 ml.) was stirred at rt for 4 h. The resulting orange slurry was treated with a 27% aqueous NH4OH solution (50 ml_). The resulting clear mixture was stirred at rt. After 2 h EtOAc (100 ml.) and a saturated NaCI solution (50 ml.) were added. The organic layer was washed with a saturated NaCI solution (50 ml_). The aqueous layers were back extracted with CHCI3 (100 ml_). The combined organic layers were concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3VCH2CI2. The product was triturated using CH2CI2 and Et2O to afford 3-({2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}-7- [(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2-pyridinecarboxamide as a white solid (42.5 mg, 43%). 1H NMR (400 MHz, DMSO-d6) δ ppm 2.23 (s, 6H), 2.35 (s, 3H), 3.15-3.27 (m, 4H), 3.74 (s, 3H), 4.24 (t, J=7.89 Hz, 2H), 6.44-6.55 (m, 1 H), 7.09 (s, 1 H), 7.20-7.50 (m, 4H), 7.88-8.1 1 (m, 3H), 8.16 (br s, 1 H), 8.22 (s, 1 H), 8.52 (s, 2H), 9.18 (d, J=7.89 Hz, 1 H), 12.59 (s, 1 H); ESIMS (M+H)+ = 656.35.
Step B/Example 73: 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-pyridinecarboxamide
A mixture of 3-({2-{[1-(N,N-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2- pyridinecarboxamide (40 mg, 0.061 mmol) and a 2M aqueous NaOH solution (1 ml.) in dioxane (5 ml.) was heated at 90 0C. After 2.5 h the reaction mixture was allowed to cool to rt and maintained overnight. The resulting slurry was filtered, the solids were washed with dioxane and E2O to obtain 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro- 1 H-indol-6-yl]amino}-1 H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-pyridinecarboxamide as a yellow solid (17.7 mg, 58%). 1H NMR (400 MHz, THF-d8) δ ppm 2.30 (s, 6H), 3.04-3.21 (m, 4H), 3.87 (s, 3H), 4.25 (t, J=8.31 Hz, 2H), 6.47 (d, J=3.16 Hz, 1 H), 6.83 (s, 1 H), 6.90 (d, J=3.02 Hz, 1 H), 7.13 (s, 1 H), 7.33-7.50 (m, 2H), 8.07 (d, J=4.26 Hz, 1 H), 8.31 (s, 1 H), 9.36 (s, 1 H), 9.66 (d, J=8.65 Hz, 1 H), 10.67 (s, 1 H), 12.55 (s, 1 H); ESIMS (M+H)+ = 502.35.
Example 75
3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino}-1H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2-pyridinecarboxamide
Figure imgf000271_0001
A solution of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrido[3,2- d]pyrrolo[2',3':4,5]pyrimido[1 ,6-a]pyrimidin-7(3H)-one hydrogen chloride (10 mg, 0.022 mmol) and 1-[(dimethylamino)acetyl]-5-(methyloxy)-2,3-dihydro-1 H-indol-6-amine (5.39 mg, 0.022 mmol) in 2,2,2-trifluoroethanol (1 ml.) was stirred at rt. After 1 h a 2M MeNH2 solution in THF (4 ml.) was added and the reaction mixture was maintained at rt for 18 h. A 2N aqueous NaOH solution (1 ml.) was added and the reaction mixture was heated at 80 0C for 4 h. The resulting mixture was allowed to cool to rt. EtOAc (50 ml.) was added and the layers were separated. The organic layer was washed with a saturated NaCI solution (50 ml_), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3VCH2CI2 to afford 3-[(2-{[1-(Λ/,Λ/- dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino}-1/-/-pyrrolo[2,3- c/]pyrimidin-4-yl)amino]-Λ/-methyl-2-pyridinecarboxamide as a white solid (5 mg, 44%). 1H NMR (400 MHz, THF-d8) δ ppm 2.30 (s, 6H), 2.96 (d, J=4.94 Hz, 3H), 3.06-3.18 (m, 4H), 3.87 (s, 3H), 4.24 (t, J=8.51 Hz, 2H), 6.52 (dd, J=3.11 , 1.83 Hz, 1 H), 6.82 (s, 1 H), 6.89-6.96 (m, 1 H), 7.36-7.45 (m, 2H), 8.05 (d. J=3.66 Hz, 1 H), 8.79 (d, 4.94 Hz, 1 H), 9.41 (s, 1 H), 9.65 (d, J=8.79 Hz, 1 H), 10.76 (s, 1 H), 12.59 (s, 1 H); ESIMS (M+H)+ = 516.16.
Example 76
3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino}-5- methyl-1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2-thiophenecarboxamide
Figure imgf000272_0001
Step AMntermediate C17: 2,4-dichloro-5-iodo-1 /-/-pyrrolo[2,3-c/]pyrimidine
Figure imgf000272_0002
2,4-dichloro-1 H-pyrrolo[2,3-d]pyrimidine (0.5 g, 2.66 mmol) was dissolved in anhydrous DMF (20 mL) and N-iodo succinimide (718 mg, 3.19 mmol) was added. After stirring for 45 minutes at room temperature, the reaction mixture was poured into water (400 mL) and extracted with EtOAc (2x500 mL) Organics were combined and washed with brine (500 mL) and dried over sodium sulfate. Solvents were removed under reduced pressure and purified via column chromatography to afford 2,4-dichloro-5-iodo-1 H- pyrrolo[2,3-c/]pyrimidine (660 mg, 2.103 mmol) as a yellow solid. ESIMS (M+H)+ =315
Step B/lntermediate C18: 2,4-dichloro-5-iodo-7-[(4-methylphenyl)sulfonyl]-7H- pyrrolo[2,3-c/]pyrimidine
Figure imgf000272_0003
2,4-dichloro-5-iodo-1 /-/-pyrrolo[2,3-c/]pyrimidine (660 mg, 2.103 mmol) and p- toluenesulfonyl chloride (441 mg, 2.31 mmol) were combined in dry dichloromethane (20 ml). Diisopropylethylamine (0.734 ml_, 4.21 mmol) was added and the reaction mixture was let stir at rt for 18 h. The reaction was then diluted with dichloromethane and the organic layer was washed with a saturated solution of sodium bicarbonate, water and a saturated solution of brine, then dried over sodium sulfate. Solvents were removed under reduced pressure and the residue purified by chromatography on SiO2 to afford 2,4-dichloro-5-iodo-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidine (901 mg, 1.94 mmol) as a yellow solid. (ESIMS (M+H)+ =469)
Step C/lntermediate C19: 2,4-dichloro-5-methyl-7-[(4-methylphenyl)sulfonyl]-7H- pyrrolo[2,3-c/]pyrimidine
Figure imgf000273_0001
2,4-dichloro-5-iodo-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidine (800 mg, 1.71 mmol) and Pd(PPh3)4 (40 mg, 0.034 mmol) were combined in dry THF (20 ml) and a 2M solution of methylzinc chloride in THF (1.28 ml, 2.56 mmol) was added under N2 .
The reaction mixture was let stir at 6O0C for 18 h at which time the reaction was diluted with ethyl acetate and the organic layer was washed with a saturated solution of sodium bicarbonate, water and a saturated brine solution, then dried over sodium sulfate. Solvents were removed under reduced pressure and the residue purified by chromatography on SiO2 to afford 2,4-dichloro-5-methyl-7-[(4-methylphenyl)sulfonyl]-7/-/- pyrrolo[2,3-c/]pyrimidine (487 mg, 1.37 mmol) as a tan solid. This reaction was repeated to synthesize sufficient material as needed for subsequent reactions. (ESIMS(M+H)+ =357)
Step D/lntermediate C20: 5-chloro-1-methyl-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one HCI salt
Figure imgf000274_0001
To a suspension of 2,4-dichloro-5-methyl-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3- c/]pyrimidine (1.27 g, 3.57 mmol) and methyl 3-amino-2-thiophenecarboxylate (0.841 g, 5.35 mmol) in TFE (20 ml.) was added trifluoroacetic acid (2.03 g, 17.83 mmol) and the mixture was heated at 850C for -30 h. At this time the reaction was terminated and the solids filtered and washed with TFE. Residual solvent was removed under reduced pressure to afford a solid containing about 90% methyl 3-({2-chloro-5-methyl-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2-thiophenecarboxylate (614 mg, 1.29 mmol). This material (and 423 mg of material obtained following the procedure for the reaction described above) was dissolved in THF (50 ml.) and a solution of lithium hydroxide (521 mg, 21.74 mmol) in water (15 ml) was added. Additional water (50 ml) and THF (35 ml) were added in efforts to solublize the reactants. The reaction was let stir at rt for 50 h at which time an additional 5 eq of LiOH was added. After several hours of stirring, little change occurred so reaction was worked up by diluting with EtOAc and washing with a saturated solution of NaCI. The organics were concentrated in vacuo to give a white residue which was dissolved in THF (10 ml) and DMF (few drops) and a solution of 2M oxalyl chloride in dichloromethane (0.960 ml_, 1.92 mmol) was added dropwise. LCMS analysis indicated some starting material remained so an additional 0.5 eq of oxalyl chloride was added. The reaction mixture was then concentrated in vacuo to afford 5-chloro-1-methyl-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one HCI salt (754 mg, 1.695 mmol) (-70% clean). (ESIMS (M+H)+ = 445)
Step E/ Intermediate C21 : 3-({2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 H- indol-6-yl]amino}-5-methyl-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4- yl}amino)-Λ/-methyl-2-thiophenecarboxamide
Figure imgf000275_0001
To a flask containing 5-chloro-1-methyl-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3/-/)-one HCI salt (747 mg, 1.09 mmol) in 2,2,2-trifluoroethanol (20 ml.) was added 1-[(dimethylamino)acetyl]-5-(methyloxy)-2,3- dihydro-1 H-indol-6-amine (352 mg, 1.41 mmol). The resulting slurry was stirred at 600C for 6 h and then rt for 16h, followed by 2h at 400C. Solvents were removed under reduced pressure to afford a green residue. Half of this material was dissolved in a 2M solution of methyl amine in THF (15 ml). The reaction was let stir at rt for 48 h, at which time the reaction mixture was diluted with ethyl acetate and washed with a saturated solution of sodium bicarbonate, water and saturated brine solution. Organics were dried over sodium sulfate and solvents removed under reduced pressure to afford a solid that was purified via chromatography on Siθ2 to afford 3-({2-{[1-(Λ/,Λ/-dimethylglycyl)-5- (methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}-5-methyl-7-[(4-methylphenyl)sulfonyl]-7/-/- pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-Λ/-methyl-2-thiophenecarboxamide (292 mg, 0.424 mmol). (ESIMS (M+H)+ = 689)
Step F/Example 76: 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-5-methyl-1 H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2- thiophenecarboxamide
3-({2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}-5-methyl- 7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-Λ/-methyl-2- thiophenecarboxamide (292 mg, 0.424 mmol) was dissolved in dioxane (6 ml.) and 2N NaOH (2 ml.) was added in a microwave safe vessel. The reaction was then heated in a microwave at 12O0C for 15 min. Reaction was diluted with EtOAc and the organic layer was washed with water and a saturated brine solution and dried over sodium sulfate. Solvents were removed under reduced pressure and the residue purified by chromatography on SiO2 and then reverse phase HPLC to afford 3-[(2-{[1-(Λ/,Λ/- dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}-5-methyl-1 /-/-pyrrolo[2,3- c/]pyrimidin-4-yl)amino]-Λ/-methyl-2-thiophenecarboxamide (52 mg, 0.097 mmol) as a tan solid. 1H NMR (400 MHz, DMSOd6) δ ppm 2.25 (s, 6 H) 2.76 (d, J=4.39 Hz, 3 H) 3.13 (t, J=8.24 Hz, 2 H) 3.18 (s, 2 H) 3.77 (s, 3 H) 4.18 (t, J=8.24 Hz, 2 H) 6.66 (s, 1 H) 6.96 (s, 1 H) 7.53 (d, J=5.31 Hz, 1 H) 7.62 (s, 1 H) 8.05 (d, J=4.58 Hz, 1 H) 8.44 (d, J=5.49 Hz, 1 H) 8.53 (s, 1 H) 10.89 (s, 1 H) 1 1.15 (s, 1 H) (3H hidden under DMSO peak at 2.5). (ESIMS (M+H)+ = 535
Example 77
3-[(2-{[5-[(Λ/,Λ/-dimethylglycyl)amino]-2-(methyloxy)phenyl]amino}-1H-pyrrolo[2,3- c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000276_0001
A slurry of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrogen chloride (400 mg, 0.856 mmol) and N1-[3-amino-4-(methyloxy)phenyl]-N2,N2-dimethylglycinamide hydrogen chloride (222 mg, 0.856 mmol) in 2,2,2-trifluoroethanol (40 ml.) was heated at 50 0C for 3 days. LCMS indicated incomplete reaction, therefore more N1-[3-amino-4-
(methyloxy)phenyl]-N2,N2-dimethylglycinamide hydrogen chloride (120 mg, 0.46 mmol) was added and the mixture was heated at 80 0C for 4 h. After allowing to cool to rt THF (50 mL) and a 27% aqueous NH4OH solution (150 mL) were added. The reaction mixture was heated at 80 0C for 18 h, allowed to cool to rt and diluted with CHCI3 (200 mL). The organic layer was washed with a saturated NaCI solution (100 mL), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)ZCH2CI2 to obtain 3-[(2-{[5-[(Λ/,Λ/-dimethylglycyl)amino]-2- (methyloxy)phenyl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2- thiophenecarboxamide as a white solid (86 mg, 21 %). 1H NMR (400 MHz, DMSO-c/6) δ ppm 2.27 (s, 6H), 3.03 (s, 2H), 3.83 (s, 3H), 6.26 (dd, J=3.30, 1.83 Hz, 1 H), 6.96 (d, J=8.79 Hz, 1 H), 7.01 (dd, J=3.30, 2.38 Hz, 1 H), 7.37 (dd, J=8.79, 2.56 Hz, 1 H), 7.58 (br s, 1 H), 7.66 (d, J=5.31 Hz, 1 H), 7.69 (s, 1 H), 8.16 (d, J=2.56 Hz, 1 H), 8.41 (d, J=5.49 Hz, 1 H), 9.42 (s, 1 H), 11.38 (s, 1 H), 1 1.44 (s, 1 H); ESIMS (M+H)+ = 484.14.
Example 78
3-[(2-{[5-[(Λ/,Λ/-dimethylglycyl)amino]-2-(methyloxy)phenyl]amino}-1H-pyrrolo[2,3- c/]pyrimidin-4-yl)amino]-Λ/-methyl-2-thiophenecarboxamide
Figure imgf000277_0001
Step A/Intermediate C22: 3-({2-{[5-[(Λ/,Λ/-dimethylglycyl)amino]-2-
(methyloxy)phenyl]amino}-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4- yl}amino)-Λ/-methyl-2-thiophenecarboxamide
Figure imgf000277_0002
A slurry of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrogen chloride (400 mg, 0.856 mmol) and N1-[3-amino-4-(methyloxy)phenyl]-N2,N2-dimethylglycinamide hydrogen chloride (222 mg, 0.856 mmol) in 2,2,2-trifluoroethanol (40 ml.) was heated at 50 0C for 3 days. LCMS indicated starting material remained, therefore more N1-[3-amino-4-
(methyloxy)phenyl]-N2,N2-dimethylglycinamide hydrogen chloride (120 mg, 0.46 mmol) was added. The reaction mixture was heated at 80 0C for 4 h. A 2M MeNH2 solution in THF (10 ml.) was added and the mixture was heated at 80 0C. After 40 h the resulting mixture was allowed to cool to rt, concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to afford 3-({2-{[5- [(Λ/,Λ/-dimethylglycyl)amino]-2-(methyloxy)phenyl]amino}-7-[(4-methylphenyl)sulfonyl]- 7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-Λ/-methyl-2-thiophenecarboxamide as a yellow solid (436 mg, 79%). ESIMS (M+H)+ = 649.20.
Step B/Example 78: 3-[(2-{[5-[(Λ/,Λ/-dimethylglycyl)amino]-2-(methyloxy)phenyl]amino}- 1 H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2-thiophenecarboxamide
A mixture of 3-({2-{[5-[(N,N-dimethylglycyl)amino]-2-(methyloxy)phenyl]amino}-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-N-methyl-2- thiophenecarboxamide (434 mg, 0.669 mmol) and a 1 M aqueous KOH solution (6.69 ml_, 6.69 mmol) in dioxane (20 ml.) was heated at 80 0C for 7 h. The resulting mixture was allowed to cool to rt. EtOAc (50 ml.) and a saturated NaHCO3 solution (50 ml.) were added. The organic layer was washed with a saturated NaCI solution (50 ml_), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2. The product was dissolved in a mixture of CH2CI2 and Et2O and concentrated again to afford 3-[(2-{[5-[(Λ/,Λ/-dimethylglycyl)amino]-2- (methyloxy)phenyl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2- thiophenecarboxamide (160 mg, 48%). 1H NMR (400 MHz, DMSO-c/6) δ ppm 2.27 (s, 6H), 2.79 (d, J=4.40 Hz, 3H), 3.03 (s, 2H), 3.83 (s, 3H), 6.60 (dd, J=3.39, 1.74 Hz, 1 H), 6.96 (d, J=8.79 Hz, 1 H), 6.99-7.05 (m, 1 H), 7.37 (dd, J=8.70, 2.47 Hz, 1 H), 7.65 (d, J=5.31 Hz, 1 H), 7.69 (s, 1 H), 8.08-8.21 (m, 2H), 8.41 (d, J=5.49 Hz, 1 H), 9.42 (s, 1 H), 11.35 (s, 1 H), 1 1.38 (br s, 1 H); ESIMS (M+H)+ = 495.13. Example 79
3-[(2-{[1 -(Λ/,Λ/-dimethylglycyl)-6-methyl-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-1 H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000279_0001
Step A/Intermediate C23: 3-({2-{[1-(Λ/,Λ/-dimethylglycyl)-6-methyl-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2- thiophenecarboxamide
Figure imgf000279_0002
A slurry of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrogen chloride (300 mg, 0.642 mmol) and 1-[(dimethylamino)acetyl]-6-methyl-1 ,2,3,4-tetrahydro-7-quinolinamine (159 mg, 0.642 mmol) in THF (50 ml.) was heated at 70 0C for 20 h. The resulting mixture was allowed to cool to rt and treated with a 27% aqueous NH4OH solution (100 ml_). The reaction mixture was stirred at rt for 3 days. LCMS indicated incomplete reaction, therefore more 27% aqueous NH4OH solution (100 ml.) was added. The mixture was heated at 80 0C for 40 h, then was allowed to cool to rt. EtOAc (200 ml.) and a saturated NaCI solution (100 ml.) were added. The organic layer was washed with a saturated NaCI solution, concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH(containing 0.2% NH3)/CH2CI2 to afford 3-({2-{[1-(Λ/,Λ/-dimethylglycyl)-6- methyl-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-7-[(4-methylphenyl)sulfonyl]-7/-/- pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-2-thiophenecarboxamide as a beige solid (55 mg, 13%). ESIMS (M+H)+ = 659.1 1.
Step B/Example 79: 3-[(2-{[1 -(Λ/,Λ/-dimethylglycyl)-6-methyl-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
A mixture of 3-({2-{[1-(N,N-dimethylglycyl)-6-methyl-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-2- thiophenecarboxamide (53 mg, 0.080 mmol) and a 1 M aqueous KOH solution (0.805 ml_, 0.805 mmol) in dioxane (10 ml.) was heated at 80 0C for 1 1 h. LCMS indicated incomplete reaction, therefore more 1 M aqueous KOH solution (0.805 ml_, 0.805 mmol) was added. The mixture was heated at 80 0C for 7 h, then was allowed to cool to rt. EtOAc (50 ml.) and a saturated NaHCO3 solution (50 ml.) were added. The organic layer was washed with a saturated NaCI solution (50 ml_), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)ZCH2CI2. The product was taken up into a mixture of CH2CI2 and hexanes and concentrated to obtain 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-6-methyl-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-1 /-/- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide as a white solid (40 mg, 99%). 1H NMR (400 MHz, DMSO-d6) δ ppm 1.84-41.96 (m, 2H), 2.12 (s, 6H), 2.17 (s,
3H), 2.71 (t, J=6.41 Hz, 2H), 3.19 (s, 2H), 3.73 (t, J=5.95 Hz, 2H), 6.20 (dd, J=3.48, 1.83 Hz, 1 H), 6.89-7.97 (m, 1 H), 7.02 (s, 1 H), 7.36-7.69 (m, 3H), 7.76 (br s, 1 H), 8.17 (s, 1 H), 8.28 (d, J=5.31 Hz, 1 H), 11.24 (s, 1 H), 11.38 (s, 1 H). ESIMS (M+H)+ = 504.87.
Example 80
3-[(2-{[1 -(Λ/,Λ/-dimethylglycyl)-6-methyl-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-1 H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2 -thiophenecarboxamide
Figure imgf000281_0001
Step A/Intermediate C24: 3-({2-{[1-(Λ/,Λ/-dimethylglycyl)-6-methyl-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-/\/- methyl-2-thiophenecarboxamide
Figure imgf000281_0002
A slurry of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrogen chloride (300 mg, 0.642 mmol) and 1-[(dimethylamino)acetyl]-6-methyl-1 ,2,3,4-tetrahydro-7-quinolinamine (159 mg, 0.642 mmol) in THF (50 ml.) was heated at 70 0C. After 20 h a 2M MeNH2 solution in THF (10 ml.) was added and the reaction mixture was stirred at rt for 3 days, then was heated at 80 0C for 40 h. The resulting mixture was allowed to cool to rt, concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to obtain 3-({2-{[1-(Λ/,Λ/-dimethylglycyl)-6-methyl-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-Λ/- methyl-2-thiophenecarboxamide as a yellow solid (162 mg, 38%). 1H NMR (400 MHz, DMSOd6) δ ppm 1.86-2.01 (m, 2H), 2.12 (br s, 6H), 2.17 (s, 3H), 2.32 (s, 3H), 2.73-2.85 (m, 5H), 3.17 (br s, 2H), 3.79 (t, J=5.86 Hz, 2H), 6.49 (d, J=4.03 Hz, 1 H), 7.14 (s, 1 H), 7.24 (d, J=7.64 Hz, 2H), 7.32 (d, J=4.03 Hz, 1 H), 7.56 (d, J=5.49 Hz, 1 H), 7.67 (br s, 1 H), 7.83 (d, J=5.68 Hz, 2H), 8.00-8.24 (m, 2H), 8.76 (s, 1 H), 1 1.43 (s, 1 H); ESIMS (M+H)+ = 673.17
Step B/Example 80: 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-6-methyl-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-/\/-methyl-2- thiophenecarboxamide
A solution of 3-({2-{[1-(N,N-dimethylglycyl)-6-methyl-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-N- methyl-2-thiophenecarboxamide (160 mg, 0.238 mmol) in dioxane (20 ml.) and a 1 M aqueous KOH solution (2.378 ml_, 2.378 mmol) was heated at 80 0C for 5 h. LCMS indicated incomplete reaction, therefore more 1 M aqueous KOH solution (0.805 ml_, 0.805 mmol) was added and the reaction mixture was heated at 80 0C for 7 h. the resulting mixture was allowed to cool to rt, diluted with EtOAc (100 ml.) and a saturated NaHCOs solution (50 ml_). The organic layer was washed with a saturated NaCI solution (50 ml_), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to afford 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-6-methyl- 1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-1/-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl- 2-thiophenecarboxamide as a white solid (74 mg, 60%). 1H NMR (400 MHz, DMSO-c/6) δ ppm 1.83-1.97 (m, 2H), 2.11 (s, 6H), 2.17 (s, 3H), 2.71 (t, J=6.50 Hz, 2H), 2.78 (d, J=4.39 Hz, 3H), 3.18 (s, 2H), 3.73 (t, J=6.04 Hz, 2H), 6.23 (dd, J=3.30, 1.83 Hz, 1 H), 6.89-6.96 (m, 1 H), 7.01 (s, 1 H), 7.54 (d, J=5.31 Hz, 1 H), 7.76 (br s, 1 H), 8.10 (q, J=4.21 Hz, 1 H), 8.16 (s, 1 H), 8.29 (d, J=5.31 Hz, 1 H), 1 1.23 (s, 1 H), 11.30 (s, 1 H); ESIMS (M+H)+ = 519.08.
Example 81
3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1H-indol-6-yl]amino}-1H- pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2-furancarboxamide
Figure imgf000283_0001
Step A/Intermediate C25: 5-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 /-/-indol- 6-yl]amino}furo[3,2-c/]pyrrolo[2',3':4,5]pyrimido[1 ,6-a]pyrimidin-7(3/-/)-one
Figure imgf000283_0002
A slurry of 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4- yl}amino)-2-furancarboxylic acid (1.925 g, 4.45 mmol) in CH2Cb (100 mL) was treated with a drop of DMF, followed by oxalyl chloride (0.779 mL, 8.89 mmol). The reaction mixture was stirred at rt. The resulting mixture was divided in half and both batches were concentrated. One batch of this crude intermediate 5-chloro-3-[(4- methylphenyl)sulfonyl]furo[3,2-d]pyrrolo[2',3':4,5]pyrimido[1 ,6-a]pyrimidin-7(3H)-one hydrogen chloride was dissolved in THF (50 mL) and 1-[(dimethylamino)acetyl]-5- (methyloxy)-2,3-dihydro-1 H-indol-6-amine (555 mg, 2.225 mmol) was added. After 6 h a 2M MeNH2 solution in THF (11.13 mL, 22.25 mmol) was added. After 3 days at rt the resulting mixture was concentrated onto Celite and purified column chromatography to obtain 5-{[1 -(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}furo[3,2- c/]pyrrolo[2',3':4,5]pyrimido[1 ,6-a]pyrimidin-7(3/-/)-one as a yellow solid (25 mg). ESIMS (M+H)+ = 628.19. Step B/Example 81 : 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-/\/-methyl-2-furancarboxamide
A solution of 5-{[1-(N,N-dimethylglycyl)-5-(methyloxy)-2,3-dihydro-1 H-indol-6-yl]amino}- 3-[(4-methylphenyl)sulfonyl]furo[3,2-d]pyrrolo[2',3':4,5]pyrimido[1 ,6-a]pyrimidin-7(3H)- one (23 mg, 0.037 mmol) in a 2M MeNH2 solution in THF (10 ml_, 20.00 mmol) was stirred at rt for 20 h. The resulting mixture was concentrated. The residue was dissolved in dioxane (1.5 ml_), treated with a 2M aqueous NaOH solution (1.5 ml_, 3.00 mmol) and heated in the uW at 120 0C for 12 min. The two layers were separated, the organic layer was concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to afford 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-5- (methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/- methyl-2-furancarboxamide as a brown solid (1 1 mg). 1H NMR (400 MHz, THF-c/8) δ ppm 2.31 (s, 6H), 2.89 (d, J=4.76 Hz, 3H), 3.09 (t, J=8.24 Hz, 2H), 3.14 (s, 2H), 3.87 (s, 3H), 4.33 (t, J=8.33 Hz, 2H), 6.35 (s, 1 H), 6.81 (s, 1 H), 6.89 (s, 1 H), 7.32-7.49 (m, 3H), 7.81 (s, 1 H), 9.51 (s, 1 H), 9.79 (s, 1 H), 10.86 (s, 1 H); ESIMS (M+H)+ = 505.14.
Example 82
3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}- 1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2-furancarboxamide
Figure imgf000284_0001
Step A/Intermediate C26: 3-({2-{[1-(Λ/,Λ/-dimethylglycyl)-6-(methyloxy)-1 , 2,3,4- tetrahydro-7-quinolinyl]amino}-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4- yl}amino)-Λ/-methyl-2-furancarboxamide
Figure imgf000285_0001
A slurry of 3-({2-chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4- yl}amino)-2-furancarboxylic acid (1.925 g, 4.45 mmol) in CH2CI2 (100 mL) was treated with a drop of DMF, followed by oxalyl chloride (0.779 mL, 8.89 mmol). The reaction mixture was stirred at rt. The resulting mixture was divided in half and both batches were concentrated. One batch of this crude intermediate 5-chloro-3-[(4- methylphenyl)sulfonyl]furo[3,2-d]pyrrolo[2',3':4,5]pyrimido[1 ,6-a]pyrimidin-7(3H)-one hydrogen chloride and 1-[(dimethylamino)acetyl]-6-(methyloxy)-1 ,2,3,4-tetrahydro-7- quinolinamine (586 mg, 2.225 mmol) in 2,2,2-trifluoroethanol (50 mL) were stirred at rt. After 3 days the resulting mixture was diluted with EtOAc (200 mL) and a saturated NaHCO3 solution (100 mL). The organic layer was washed with a saturated NaCI solution (100 mL), dried (Na2SO4) and concentrated. The residue was triturated using Et2O. This crude intermediate was dissolved in THF (10 mL) and a 2M MeNH2 solution in THF (20 mL). The reaction mixture was heated at 80 0C for 6 h. The resulting mixture was allowed to cool to rt, concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to obtain 3-({2-{[1- (Λ/,Λ/-dimethylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-Λ/-methyl-2- furancarboxamide as a brown solid (88 mg). ESIMS (M+H)+ = 673.17.
Step B/Example 82: 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2-furancarboxamide
A mixture of 3-({2-{[1-(N,N-dimethylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-N- methyl-2-furancarboxamide (88 mg, 0.131 mmol) and a 2M aqueous NaOH solution (5 ml.) in dioxane (5 ml.) was heated in the MW at 120 0C for 12 min. The resulting mixture was diluted with EtOAc (50 ml.) and a saturated NaHCOs solution (50 ml_). The organic layer was washed with a saturated NaCI solution (50 ml_), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2Cl2 to afford 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-7- quinolinyl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2-furancarboxamide as a beige solid (82 mg). 1H NMR (400 MHz, DMSO-d6) δ ppm 1.79-1.97 (m, 2H), 2.12 (br s, 6H), 2.59-2.76 (m, 2H), 2.79 (d, J=4.58 Hz, 3H), 3.26 (s, 2H), 3.71 (t, J=6.04 Hz, 2H), 3.84 (s, 3H), 6.26 (dd, J=3.20, 1.56 Hz, 1 H), 6.84 (s. 1 H), 6.94-7.06 (s, 1 H), 7.63- 7.77 (m, 3H), 8.26 (q, J=4.27 Hz, 1 H), 8.36 (s, 1 H), 9.66 (s, 1 H), 1 1.40 (br s, 1 H); ESIMS (M+H)+ = 519.21.
Example 83
3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-
1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-pyridinecarboxamide
Figure imgf000286_0001
A mixture of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrido[3,2- d]pyrrolo[2',3':4,5]pyrimido[1 ,6-a]pyrimidin-7(3H)-one hydrogen chloride (300 mg, 0.649 mmol) and 1 -[(dimethylamino)acetyl]-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinamine (171 mg, 0.649 mmol) in 2,2,2-trifluoroethanol (10 ml.) was stirred at rt. After 3 days THF (10 ml.) was added, followed by a 27% aqueous NH4OH solution (30 ml_). The reaction mixture was stirred at rt. After 5 h a 2M aqueous NaOH solution (2 ml.) was added and the reaction mixture was heated at 80 0C for 6 h. The resulting mixture was allowed to cool to rt, diluted with EtOAc (100 ml.) and a saturated NaCI solution (50 ml_). The organic layer was concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3VCH2CI2 to afford 3-[(2-{[1-(Λ/,Λ/- dimethylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-1 /-/-pyrrolo[2,3- c/]pyrimidin-4-yl)amino]-2-pyridinecarboxamide as a white solid (65 mg). 1H NMR (400 MHz, DMSOd6) δ ppm 1.79-1.97 (m, 2H), 2.08 (br s, 6H), 2.71 (br s, 2H), 3.21 (s, 2H), 3.73 (t, J=5.59 Hz, 2H), 3.84 (s, 3H), 6.28-6.35 (m, 1 H), 6.85 (s, 1 H), 7.04 (s, 1 H), 7.48 (dd, J=8.42, 4.2 Hz, 1 H), 7.72 (s, 1 H), 7.80 (d, J=1.83 Hz, 1 H), 8.15-8.27 (m, 1 H), 8.38 (s, 1 H), 8.53 (d, J= 1.65 Hz, 1 H), 9.45 (d, J=8.61 Hz, 1 H), 11.44 (br s, 1 H), 12.45 (s, 1 H); ESIMS (M+H)+ = 516.38.
Example 84 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-
1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2-pyridinecarboxamide
Figure imgf000287_0001
A mixture of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrido[3,2- d]pyrrolo[2',3':4,5]pyrimido[1 ,6-a]pyrimidin-7(3H)-one hydrogen chloride (300 mg, 0.649 mmol) and 1-[(dimethylamino)acetyl]-6-(methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinamine (171 mg, 0.649 mmol) in 2,2,2-trifluoroethanol (10 ml.) was stirred at rt. After 3 days THF (10 ml.) was added, followed by a 2M MeNH2 solution in THF (20 ml_). The reaction mixture was stirred at rt. After 5 h a 2M aqueous NaOH solution (2 ml.) was added and the reaction mixture was heated at 80 0C for 6 h. The resulting mixture was allowed to cool to rt, concentrated onto Celite and purified by silica gel chromatography using 1- 10% MeOH (containing 0.2% NH3)/CH2CI2 to obtain 3-[(2-{[1-(Λ/,Λ/-dimethylglycyl)-6- (methyloxy)-1 ,2,3,4-tetrahydro-7-quinolinyl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4- yl)amino]-Λ/-methyl-2-pyridinecarboxamide as a yellow solid (197 mg). 1H NMR (400 MHz, DMSO-de) δ ppm 1.82-1.96 (m, 2H), 2.08 (br s, 6H), 2.71 (br s, 2H), 2.87 (d, J=4.94 Hz, 3H), 3.22 (s, 2H), 3.66-3.79 (m, 2H), 3.85 (s, 3H), 6.36 (dd, J=3.20, 1.74 Hz, 1 H), 6.85 (s, 1 H), 7.04 (s, 1 H), 7.48 (dd, J=8.52, 4.30 Hz, 1 H), 7.72 (s, 1 H), 8.21 (dd, J=4.39, 1.46 Hz, 1 H), 8.39 (s, 1 H), 9.18 (q, J=4.58 Hz, 1 H), 9.44 (dd, J=8.61 , 0.92 Hz, 1 H), 1 1.44 (br s, 1 H), 12.43 (s, 1 H); ESIMS (M+H)+ = 530.39.
Example 85 3-[(2-{[(2f?)-1 -(Λ/,Λ/-dimethylglycyl)-2-methyl-5-(methyloxy)-2,3-dihydro-1H-indol-6- yl]amino}-1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000288_0001
A slurry of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrogen chloride (0.600 mg, 1.284 μmol) and (2R)-1-[(dimethylamino)acetyl]-2-methyl-5-(methyloxy)-2,3-dihydro-1 H-indol- 6-amine (0.338 mg, 1.284 μmol) in 2,2,2-trifluoroethanol (10 mL) was stirred at rt for 18 h. The resulting deep green slurry was split into 2 equal batches. One of the batches was treated with a 27% aqueous NH4OH solution (10 mL). The orange slurry was heated at 80 0C for 20 h. The resulting mixture was diluted with EtOAc (50 mL) and a saturated NaHCO3 solution (50 mL). The organic layer was washed with a saturated NaCI solution (50 mL), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)ZCH2CI2. This crude intermediate was dissolved in dioxane (10 mL) and a 2M aqueous NaOH solution (2 mL) and was heated at 80 0C for 5 h. The resulting mixture was allowed to cool to rt, diluted with EtOAc (50 mL) and a saturated NaCI solution (10 mL). The organic layer was concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to afford 3- [(2-{[(2R)-1-(Λ/,Λ/-dimethylglycyl)-2-methyl-5-(methyloxy)-2,3-dihydro-1 H-indol-6- yl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide as a yellow solid (122 mg, 55%). 1H NMR (400 MHz, DMSO-d6) δ ppm 1.27 (d, J=5.13 Hz, 3H), 2.26 (s, 6H), 2.60 (d, J=15.38 Hz, 1 H), 3.08 (d, J=14.28 Hz, 1 H), 3.26-3.49 (m, 2H), 3.80 (s, 3H), 4.72-4.88 (m, 1 H), 6.24 (s, 1 H), 6.99 (s, 2H), 7.39-7.80 (m, 4H), 8.40 (d, J=5.13 Hz, 1 H), 8.59 (s, 1 H), 1 1.33 (s, 1 H), 11.43 (s, 1 H); ESIMS (M+H)+ = 521.34; Optical rotation +14.2 (1 mg/ml_ MeOH, rt).
Example 86
3-[(2-{[(2S)-1-(Λ/,Λ/-dimethylglycyl)-2-methyl-5-(methyloxy)-2,3-dihydro-1H-indol-6- yl]amino}-1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000289_0001
A slurry of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrogen chloride (0.600 mg, 1.284 μmol) and (2S)-1-[(dimethylamino)acetyl]-2-methyl-5-(methyloxy)-2,3-dihydro-1 H-indol- 6-amine (0.338 mg, 1.284 μmol) in 2,2,2-trifluoroethanol (10 mL) was stirred at rt for 18 h. The resulting deep green slurry was split into 2 equal batches. One of the batches was treated with a 27% aqueous NH4OH solution (10 mL). The resulting orange slurry was stirred at rt for 20 h. The resulting mixture was diluted with EtOAc (50 mL) and a saturated NaHCO3 solution (50 mL). The organic layer was washed with a saturated NaCI solution (50 mL), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)ZCH2CI2. This crude intermediate was taken up into dioxane (10 mL) and a 2M aqueous NaOH solution (2 mL), and was heated at 80 0C for 5 h. The resulting mixture was allowed to cool to rt, diluted with EtOAc (50 mL) and a saturated NaCI solution (10 mL). The organic layer was concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to afford 3-[(2-{[(2S)-1-(Λ/,Λ/-dimethylglycyl)-2- methyl-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4- yl)amino]-2-thiophenecarboxamide as a yellow solid (129 mg, 74%). 1H NMR (400 MHz, DMSO-de) δ ppm 1.27 (d, J=5.49 Hz, 3H), 2.26 (s, 6H), 2.60 (d, J=15.57 Hz, 1 H), 3.08 (d, J=14.28 Hz, 1 H), 3.23-3.47 (m, 2H), 3.80 (s, 3H), 4.72-4.86 (m, 1 H), 6.25 (s, 1 H), 6.99 (s, 2H), 7.38-7.78 (m, 4H), 8.40 (d, J=5.31 Hz, 1 H), 8.59 (s, 1 H), 1 1.33 (s, 1 H), 11.43 (s, 1 H); ESIMS (M+H)+ = 521.32; Optical rotation -10.6 (1 mg/ml_ MeOH, rt).
Example 87
3-[(2-{[(2f?)-1-(Λ/,Λ/-dimethylglycyl)-2-methyl-5-(methyloxy)-2,3-dihydro-1H-indol-6- yl]amino}-1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2- thiophenecarboxamide
Figure imgf000290_0001
Step A/Intermediate C27: 3-({2-{[(2R)-1-(Λ/,Λ/-dimethylglycyl)-2-methyl-5-(methyloxy)- 2,3-dihydro-1 H-indol-6-yl]amino}-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin- 4-yl}amino)-Λ/-methyl-2-thiophenecarboxamide
Figure imgf000290_0002
A slurry of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrogen chloride (0.600 mg, 1.284 μmol) and (2R)-1-[(dimethylamino)acetyl]-2-methyl-5-(methyloxy)-2,3-dihydro-1 H-indol- 6-amine (0.338 mg, 1.284 μmol) in 2,2,2-trifluoroethanol (10 mL) was stirred at rt for 18 h. The resulting deep green slurry was split into 2 equal batches. One of the batches was treated with a 2M MeNH2 solution in THF (10 mL). The orange slurry was stirred at rt for 2 days. The resulting mixture was diluted with EtOAc (50 mL) and a saturated NaCI solution (50 mL). The organic layer was washed with a saturated NaCI solution (50 mL), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)ZCH2CI2, followed by trituration using Et2O to obtain 3-({2-{[(2/?)-1- (Λ/,Λ/-dimethylglycyl)-2-methyl-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-c/]pyrimidin-4-yl}amino)-Λ/-methyl-2- thiophenecarboxamide as a yellow solid (68 mg, 16%). 1H NMR (400 MHz, DMSO-c/6) δ ppm 1.30 (s, 3H), 2.31 (s, 6H), 2.33 (s, 3H), 2.59-2.84 (m, 4H), 3.16-3.58 (m, 3H), 3.77 (s, 3H), 4.73-4.89 (m, 1 H), 6.50 (d, J=3.48 Hz, 1 H), 7.10 (s, 1 H), 7.20-7.41 (m, 3H), 7.52 (s, 1 H), 7.84-8.02 (m, 2H), 8.05-8.31 (m, 3H), 8.39 (s, 1 H), 1 1.43 (s, 1 H); ESIMS (M+H)+ = 689.26.
Step B/Example 87: 3-[(2-{[(2R)-1-(Λ/,Λ/-dimethylglycyl)-2-methyl-5-(methyloxy)-2,3- dihydro-1 /-/-indol-6-yl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2- thiophenecarboxamide
A solution of 3-({2-{[(2R)-1-(N,N-dimethylglycyl)-2-methyl-5-(methyloxy)-2,3-dihydro-1 H- indol-6-yl]amino}-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)-N- methyl-2-thiophenecarboxamide (66 mg, 0.096 mmol) in dioxane (10 mL) and a 2M aqueous NaOH solution (2 mL) was heated at 80 0C for 6 h. The resulting mixture was allowed to cool to rt, diluted with EtOAc (50 mL) and a saturated NaCI solution (10 mL). The organic layer was concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to obtain 3-[(2- {[(2R)-1-(Λ/,Λ/-dimethylglycyl)-2-methyl-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}- 1 H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2-thiophenecarboxamide as a yellow solid (48 mg, 94%). 1H NMR (400 MHz, DMSO-d6) δ ppm 1.27 (d, J=5.31 Hz, 3H), 2.26 (s, 6H), 2.61 (d, J=15.02 Hz, 1 H), 2.79 (d, J=4.58 Hz, 3H), 3.08 (d, J=14.28 Hz, 1 H), 3.22-3.47 (m, 2H), 3.80 (s, 3H), 4.72-4.86 (m, 1 H), 6.28 (s, 1 H), 6.92-7.08 (m, 2H), 7.52- 7.63 (m, 1 H), 7.66 (s, 1 H), 8.13 (s, 1 H), 8.40 (d, J=5.49 Hz, 1 H), 8.58 (s, 1 H), 1 1.33 (s, 1 H), 1 1.34 (s, 1 H). ESIMS (M+H)+ = 535.35; Optical rotation +13.7 (1 mg/mL MeOH, rt).
Example 88 3-[(2-{[(2S)-1-(Λ/,Λ/-dimethylglycyl)-2-methyl-5-(methyloxy)-2,3-dihydro-1H-indol-6- yl]amino}-1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2- thiophenecarboxamide
Figure imgf000292_0001
Step A/Intermediate C28: 3-({2-{[(2S)-1-(Λ/,Λ/-dimethylglycyl)-2-methyl-5-(methyloxy)-2,3- dihydro-1 /-/-indol-6-yl]amino}-7-[(4-methylphenyl)sulfonyl]-7/-/-pyrrolo[2,3-c/]pyrimidin-4- yl}amino)-Λ/-methyl-2-thiophenecarboxamide
Figure imgf000292_0002
A slurry of 5-chloro-3-[(4-methylphenyl)sulfonyl]pyrrolo[3,2- e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrogen chloride (0.600 mg, 1.284 μmol) and (2S)-1-[(dimethylamino)acetyl]-2-methyl-5-(methyloxy)-2,3-dihydro-1 H-indol- 6-amine (0.338 mg, 1.284 μmol) in 2,2,2-trifluoroethanol (10 mL) was stirred at rt for 18 h. The resulting deep green slurry was split into 2 equal batches. One of the batches was treated with a 2M MeNH2 solution in THF (10 mL). The orange slurry was stirred at rt for 2 days. The resulting mixture was diluted with EtOAc (50 mL) and a saturated NaCI solution (50 mL). The organic layer was washed with a saturated NaCI solution (50 mL), concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3)/CH2CI2 to afford as a yellow solid (300 mg, 70%). 1H NMR (400 MHz, DMSO-de) δ ppm 1.29 (s, 3H), 2.26 (s, 6H), 2.33 (s, 3H), 2.68 (d, J= 15.75 Hz, 1 H), 2.76 (d, J=4.58 Hz, 3H), 3.15 (d, 14.47 Hz, 1 H), 3.28-3.52 (m, 2H), 3.76 (s, 3H), 4.76- 4.87 (m, 1 H), 6.51 (d, J=3.66 Hz, 1 H), 7.09 (s, 1 H), 7.21-7.38 (m, 3H), 7.52 (br s, 1 H), 7.95 (d, J=6.59 Hz, 2H), 8.07-8.27 (m, 3H), 8.39 (s, 1 H), 1 1.43 (s, 1 H); ESIMS (M+H)+ = 689.30.
StepB/Example 88: 3-[(2-{[(2S)-1 -(Λ/,Λ/-dimethylglycyl)-2-methyl-5-(methyloxy)-2,3- dihydro-1 /-/-indol-6-yl]amino}-1 /-/-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2- thiophenecarboxamide
A solution of 3-[(2-{[(2R)-1-(N,N-dimethylglycyl)-2-methyl-5-(methyloxy)-2,3-dihydro-1 H- indol-6-yl]amino}-1 H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-N-methyl-2- thiophenecarboxamide (296 mg, 0.430 mmol) in dioxane (10 ml.) and a 2M aqueous NaOH solution (2 ml.) was heated at 80 0C for 7 h. The resulting mixture was allowed to cool to rt, diluted with EtOAc (50 ml.) and a saturated NaCI solution (10 ml_). The organic layer was concentrated onto Celite and purified by silica gel chromatography using 1-10% MeOH (containing 0.2% NH3VCH2CI2 to afford 3-[(2-{[(2S)-1-(Λ/,Λ/- dimethylglycyl)-2-methyl-5-(methyloxy)-2,3-dihydro-1 /-/-indol-6-yl]amino}-1 /-/-pyrrolo[2,3- c/]pyrimidin-4-yl)amino]-Λ/-methyl-2 -thiophenecarboxamide as a yellow solid (180 mg, 78%). 1H NMR (400 MHz, DMSO-d6) δ ppm 1.27 (d, J=5.31 Hz, 3H), 2.26 (s, 6H), 2.60 (d, J=15.20 Hz, 1 H), 2.79 (d, J=4.58 Hz, 3H), 3.08 (d, J=14.28 Hz, 1 H), 3.25-3.47 (m, 2H), 3.80 (s, 3H), 4.72-4.86 (m, 1 H), 6.28 (s, 1 H), 6.92-7.08 (m, 2H), 7.52-7.63 (m, 1 H), 7.66 (s, 1 H), 8.13 (s, 1 H), 8.40 (d, J=5.31 Hz, 1 H), 8.59 (s, 1 H), 1 1.33 (s, 1 H), 11.34 (s, 1 H); ESIMS (M+H)+ = 535.41 ; Optical rotation -8.5 (1 mg/ml_ MeOH, rt).
Example 89
3-[(2-{[5-(dimethylamino)-1-(Λ/,Λ/-dimethylglycyl)-2,3-dihydro-1H-indol-6-yl]amino}- 1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-Λ/-methyl-2 -thiophenecarboxamide
Figure imgf000294_0001
In a manner analogous to a procedure outlined previously, 3-[(2-{[5-(dimethylamino)-1- (Λ/,Λ/-dimethylglycyl)-2,3-dihydro-1/-/-indol-6-yl]amino}-1/-/-pyrrolo[2,3-c/]pyrimidin-4- yl)amino]-Λ/-methyl-2-thiophenecarboxamide was prepared from 5-chloro-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrochloride, 1-[(dimethylamino)acetyl]-N5,N5-dimethyl-2,3-dihydro-1 H-indole-5,6- diamine, and methyl amine to afford the title compound ( 0.2 g, 50% over 3 steps). ESIMS (M+H)+ = 534. 1 H NMR (400 MHz, DMSOd6) δ ppm 2.25 (s, 6 H) 2.60 (s, 6 H) 2.79 (d, J=4.41 Hz, 3 H) 3.04 - 3.12 (m, 2 H) 3.18 (s, 2 H) 4.14 - 4.21 (m, 2 H) 6.29 (br. s., 1 H) 6.99 (br. s., 1 H) 7.07 (s, 1 H) 7.62 (d, J=5.62 Hz, 1 H) 7.73 (s, 1 H) 8.09 - 8.19 (m, 1 H) 8.42 (d, J=5.22 Hz, 1 H) 8.75 (s, 1 H) 11.33(s, 1 H) 1 1.36(s, 1 H).
Example 90
3-[(2-{[5-(dimethylamino)-1-(Λ/,Λ/-dimethylglycyl)-2,3-dihydro-1H-indol-6-yl]amino}-
1H-pyrrolo[2,3-c/]pyrimidin-4-yl)amino]-2-thiophenecarboxamide
Figure imgf000294_0002
In a manner analogous to a procedure outlined previously, 3-[(2-{[5-(dimethylamino)-1- (Λ/,Λ/-dimethylglycyl)-2,3-dihydro-1/-/-indol-6-yl]amino}-1/-/-pyrrolo[2,3-c/]pyrimidin-4- yl)amino]-2-thiophenecarboxamide was prepared from 5-chloro-3-[(4- methylphenyl)sulfonyl]pyrrolo[3,2-e]thieno[3',2':4,5]pyrimido[1 ,2-c]pyrimidin-7(3H)-one hydrochloride, 1-[(dimethylamino)acetyl]-N5,N5-dimethyl-2,3-dihydro-1 H-indole-5,6- diamine, and ammonium hydroxide to afford the title compound ( 0.25 g, 63% over 2 steps). ESIMS (M+H)+ = 520. 1 H NMR (400 MHz, DMSOd6) δ ppm 2.25 (br. s., 6 H) 2.60 (s, 6 H) 3.08 (t, J=6.72 Hz, 2 H) 3.18 (br. s., 2 H) 4.17 (t, J=7.93 Hz, 2 H) 6.26 (br. s., 1 H) 6.99 (br. s., 1 H) 7.07 (br. s., 1 H) 7.5-7.64 (m, 3 H) 7.73 (br. s., 1 H) 8.42 (d, J=5.02 Hz, 1 H) 8.75 (s, 1 H) 1 1.33 (br. s., 1 H) 11.44 (br. s., 1 H).
ASSAYS
Time Resolved Fluorescence Resonance Energy Transfer (TR-FRET) assays were carried out on the exemplified compounds as follows:
The source of substrate peptide
The biotinylated substrate peptide, (sequence - Biotin-aminohexyl-
AEEEEY*MMMMAKKKK-NH2) is purchased from QCB, Inc. (Hopkinton, MA). Purity is determined by HPLC. The calculated molecular mass of the peptide is 2216 dalton. Tyrosine phosphorylation by human IGF1 R (hlGF1 R) or human IR (hlR) occurs as indicated (Y*) in the peptide sequence described. Solid peptide sample is dissolved to approximately 1 mM in DMSO, aliquoted, and stored at -200C until use. True peptide concentration is determined by amino acid analysis.
The source of enzyme: hlGFI R: GST-rTEV-IGF-1 R(957-1367) containing amino acid residues 957- 1367 of human IGF1 R (as annotated by National Center for Biotechnology Information (NCBI) accession number NP_000866) is purified from a baculovirus expression system in Sf9 cells using Glutathione Sepharose 4FF column chromatography followed by
Sephadex-200 size exclusion column chromatography. Enzyme purity of approximately 95% is achieved. Samples, in 25 mM Tris-HCI, 250 mM NaCI, 5% glycerol, 1 mM DTT, pH 7.5, are stored at -800C until use. hIR: GST-rTEV-IR(979-1382) containing amino acid residues 979-1382 of human IR (as annotated by NCBI accession number NP_000199) is expressed and purified by the same process as hlGF1 R. Enzyme purity of approximately 92% is achieved. Samples, in 25 mM Tris-HCI, 250 mM NaCI, 5% glycerol, 1 mM DTT, pH 7.5, are stored at -800C until use.
Activation of hlGF1 R and hIR by autophosphorylation: hlGFI R: Activation of GST-rTEV-IGF-1 R(957-1367) is achieved by a 4 minute incubation of hlGF1 R (2.7 μM final) with 2 mM ATP in 50 mM HEPES, 20 mM MgCL2, 0.1 mg/ml BSA, at room temperature. Autophosphorylation is stopped by addition of
EDTA (to 100 mM final). Aliquoted samples are flash frozen in liquid nitrogen and stored at -800C until use. hIR: Activation of GST-rTEV-IGF-1 R(957-1367) is achieved by a 5 minute incubation of hIR (2.7 μM final) with 2 mM ATP in 100 mM HEPES, 10 mM MgCL2, 0.1 mg/ml BSA, at room temperature. Autophosphorylation is stopped by addition of EDTA
(to 100 mM final). Aliquoted samples are flash frozen in liquid nitrogen and stored at -
800C until use.
Kinase assay of purified hlGF1 R or hIR: Assays are performed in 384-well (Greiner, Catalog No. 784076) microtiter plates. Reaction buffer (50 mM HEPES buffer, pH 7.5; 10 mM MgCL2; 3 mM DTT; 1 mM CHAPS; 0.1 mg/ml BSA) for peptide phosphorylation (10 μl volume) contained, in final concentrations, 500 nM biotinylated peptide substrate; 10 μM ATP; and purified, activated hlGFI R or hIR (0.5 nM). Compounds, titrated in DMSO, are evaluated at eleven concentrations ranging from 50 μM to 0.2 nM. Final assay concentrations of DMSO do not exceed 10%. No effect on activity relative to controls without DMSO is observed for hlGF1 R or IR at these DMSO amounts. Reactions are incubated for 1 hour at room temperature and are stopped by a 5 μl addition of EDTA (to 33 mM). A further addition of 5 μl detection reagents (for final 7 nM Streptavidin-APC (PerkinElmer #CR130-150), 1 nM Europium-labeled anti-phosphotyrosine monoclonal antibody
(PerkinElmer #AD0067), added in reaction buffer (without DTT), is required for signal generation. After 30 minutes, signal is read on PerkinElmer Viewlux microplate imager or Wallac Victor fluorometer. The data for compound concentration responses were plotted as % Inhibition, calculated with the data reduction formula 100*(1-[(U1-C2)/(C1-C2)]), versus concentration of compound, where
U is the unknown value,
C1 is the average control value obtained for DMSO only, and
C2 is the average control value obtained for reactions stopped with EDTA at t=0. Data were fitted to the curve described by: y = ((Vmax * x) / (K + x)) where
Vmax is the upper asymptote and K is the IC50. The results for each compound were recorded as plC50 calculated as follows:
PlC50 = -Log10(K).
plC50 values for the compounds of the examples (if available) were categorized by relative inhibition of IGF-1 R and IR. The results are summarized in the tables below.
Figure imgf000297_0001
Figure imgf000297_0002
Cell Proliferation Data
A. Inhibition of cell proliferation by IGF-1 R inhibitors Cell proliferation was measured by either CellTiter-Glo (measures cellular ATP level as a surrogate for total cell number) or by InCeII analyzer (counts number of nuclei as a measure of cell number).
For CellTiter-Glo assay, exponentially growing cell lines of different tumor origins, cultured in appropriate media containing 10% fetal bovine serum at 370C in a 5% CO2 incubator, were plated at low density (less than 2000cells/well) in 96-well plates. Twenty four hours post-plating, cells were treated with different concentrations of test compounds ranging from 3OuM to 1.5nM. Several wells were left untreated as a control. Seventy two hours post-treatment, cell numbers were determined using 50-10OuI per well of CellTiter-Glo (Promega #G7573). Plates were incubated at 370C for 30 minutes and the chemiluminescent signal was read on the Victor V or Envison 2100 reader. Percent inhibition of cell growth was expressed as percent proliferation relative to 100% proliferation (control). Concentration of test compound that inhibited 50% of cell growth (IC50) was determined by 4 parameter fit of data using XLfit, (value of no cell control was substracted from all samples for background).
For InCeII Analyser assay, various cell lines of different tumor origins were grown to 70-80% confluency in appropriate culture media containing 10% fetal bovine serum at 370C in a 5% CO2 incubator. On day -1 , cells were seeded at 2 densities in 384-well plates and incubated at 37°C overnight. Stock compound plates were prepared in advance which contained dimethyl sulfoxide (DMSO) alone and a 9-point half-log decreasing dose range of the compound in DMSO. The compund plates were stored at -8O0C and each plate was only thawed once and used. On day 0, the main assay plates received compound or DMSO via a sonic delivery system (ECHO). The highest final concentration of compound of the dose range in the culture plates was 10uM. These plates were cultured at 37°C for 3 days. A parallel set of cell line plates, which did not receive compound, were processed and read on day 0 to provide a T=O (time zero). On day 3, the compound treated plates were stained and fixed to measure proliferation, apoptosis, and mitotic index using an InCeIL1OOO analyser. A nuclear stain was used to identify cells in the wells. By counting the number of nuclei, the proliferation index of compound treated groups were calculated as a percentage relative to the DMSO control, which was set to 100%. The IC50 values were calculated using model 205 in ExcelFit.
IC50 (nM) values for compounds of select examples were categorized by relative inhibition of cell proliferation. The results are summarized in the tables below.
Figure imgf000299_0001
Figure imgf000299_0002
B. IGF-1 R and IR Cellular Autophosphorylation
NIH-3T3 cells overexpressing human IGF-1 R or IR were plated in 96-well plates (10,000 cells/well) in culture media containing 10% fetal bovine serum and incubated at 370C in a 5% CO2 incubator. Twenty four hours post-plating, cells were treated with different concentrations of test compounds ranging from 3OuM to 1.5nM. Two hours after compound addition, cells were stimulated with either human IGF-1 (30 ng/ml) or insulin (3 ug/ml) for 15 minutes. Cell lysates are analyzed for phosphorylated receptors using dissociation enhanced lanthanide fluor-immuno assay (DELFIA) with anti-IGF-1 R (MAB391 , R&D Systems, Minneapolis, MN) or anti-IRβ (sc-711 , Santa Cruz Biotechnology, Santa Cruz, CA) capture antibody and europium-labeled anti-pTyr antibody (Eu-NI PT66, Perkin Elmer, Waltham, MA) for detection. The fluorescence signal for cells treated with compounds was expressed as percent relative to 100% stimulation (IGF-1 or insulin stimulated signal). Concentration of test compound that inhibited 50% of ligand-induced receptor phosphorylation (IC50) was determined by 4 parameter fit of data using XLfit, (value of no cell control was substracted from all samples for background).
IC50 values for the compounds of the examples (if available) were categorized by relative inhibition of IGF-1 R and IR. The results are summarized in the tables below.
Figure imgf000299_0003
Figure imgf000300_0001
Figure imgf000300_0002
ALK Enzyme Assay: The method measures the ability of the isolated enzyme to catalyse the transfer of the gamma-phosphate from ATP onto the tyrosine residue of a biotinylated synthetic peptide. The extent of tyrosine phosphorylation was measured using an anti- phosphotyrosine antibody, and quantified by homogenous time-resolved fluorescence (HTRF) assay purchased from CisBio (62TK0PEJ). Reactions were performed in black 384-well polystyrene low volume plates in a final volume of 5 μl. Assays were performed by adding 2.5 μl of each of the following solutions, enzyme mix and substrate: The enzyme mix, final concentration in plate, contained 50 mM MOPS (pH 7.0); 10 nM supplement enzymatic buffer; 0.03 % NaN3; 0.01 % BSA, 0.1 mM orthovanadate, 5 mM MgCI2, 1 mM CHAPS, 1 mM dithiothreitol and 0.25 nM ALK. The Substrate mix, final concentration in plate, contained 50 mM MOPS (pH 7.0); 10 nM supplement enzymatic buffer; 0.03 % NaN3; 0.01 % BSA, 0.1 mM orthovanadate, 5 mM MgCI2, 1 mM CHAPS, 1 mM dithiothreitol, 15.0 μM ATP, and 0.25 uM TK substrate-biotin.
To quantify compound potencies, the enzyme mix was added to the compound plates and the plates were incubated at 2O0C for 30 minutes. The reactions were then started by adding the substrate mix. The reactions were allowed to proceed for 120 minutes at 2O0C. The reactions were then terminated by the addition of 5 μl HTRF detection mix to each well. The final concentrations of the detection reagents were: 50 mM HEPES (pH7.0); 0.1 % BSA; 0.8 M KF; 20 mM EDTA; 1 mM CHAPS; TK-antibody Europium Cryptate; and 41 nM streptavidin-XL665. Assay plates were left unsealed, stacked and incubated at 20° for 60 minutes. Quantification of activity was achieved by counting in an Envision Counter (PerkinElmer).
Compounds under analysis were dissolved in Me2SO to 1.0 mM and serially diluted 1 to 3 with Me2SO through twelve dilutions. 0.05 μl of each concentration was transferred to the corresponding well of an assay plate. This creates a final compound concentration range from 0.00017 to 10 μM.
The data for dose responses were plotted as % Inhibition calculated with the data reduction formula 100*(1-(U1-C2)/(C1-C2)) versus concentration of compound where U is the unknown value, C1 is the average control value obtained for 1% DMSO, and C2 is the average control value obtained for 0.1 M EDTA. Data were fitted with a curve described by: y = A + B - A
Figure imgf000301_0001
where A is the minimum y, B is the maximum y concentration [M], D is the slope factor, and x is the log™ of the compound. The results for each compound were recorded as plC50s, calculated as follows: plC50 = -Log10(K) plC50 values for the compounds of the examples (if available) were categorized by relative inhibition of IGF-1 R and IR. The results are summarized in the tables below.
Figure imgf000301_0002

Claims

We claim:
1. A compound of formula (I):
Figure imgf000302_0001
or a pharmaceutically acceptable salt or solvate thereof wherein:
R4a and R4b are each independently selected from H, alkylCi-c6, -OH, -alkyleneci-cβ-OH, -alkyleneCi-c6-phenyl (optionally substituted with a halo), and -alkyleneCi-c6-C(O)NH2; one or two of X1, X2, and X3 are independently selected from N, O, and S, while the remaining one or two of X1, X2, and X3 are C; m is 0 or 1 ;
R1 is selected from H, halo, alkylCi-c6, -alkyleneci-cs-haloi-s, and -O-alkylCi-c6when X1 is C, otherwise R1 is absent;
R2 is selected from H, halo, alkylCi-c6, -alkyleneci-cs-haloi-s, and -O-alkylCi-c6when X2 is C, otherwise R2 is absent;
R3 is selected from H, halo, alkylCi-c6, -alkyleneci-cs-haloi-s, and -O-alkyld-cewhen X3 is C, otherwise R3 is absent;
R7 is selected from alkylCi-c6, -O-alkylCi-c6, halo, -NR22R23, and -O-alkyleneCi-c6-halθi-3; R8 is selected from H, halo, and alkyld-cβ; one of R9 and R10 is selected from -alkyleneCi-c6-SO2-alkylci-c6,
-N-alkyleneCo-c6-C(0)-alkyleneCo-c6-NR22R23, -O-alkyleneco-ceCoptionally substituted with -OH)-NR22R23,
Figure imgf000303_0001
(R14)i-2 , and (R14)i-2; and the other of R9 and R10 is selected from H, alkylCi-c6, -O-alkylCi-c6, and halo;
wherein Het1 and Het2 are each independently a five or six membered heterocyclic ring having an N atom and optionally one or two additional heteroatoms selected from N and O, and each R14 is independently selected from H, OH, halo, alkylCi-c6, -O-alkylCi-c6, -cyclopropyl, -C(O)-alkylCi-c6, SO2-alkylCi-c6, -(CH2)i-4-halo ,and -(CH2)1-4-SO2-alkylci-c6; -C(O )-alkyleneC0-c6-N R22R23, -NR22R23; or
R9 and R10, together with the atoms to which they are attached form a five, six, or seven- membered heterocyclic ring containing one or two N atom and the remainder C atoms, wherein at least one N atom is substituted with R15, and the C atoms of the heterocyclic ring are optionally substituted with (R19)i-2; wherein R15 is selected from H, -alkylCi-c4, -alkyleneCi-c4-halo, -C(O)-alkyleneC0-c6-N R22R23, -C(O)-alkylci-c6, -alkyleneCi-c4-N R22R23, -alkyleneCi-c4-C(O)-NR22R23, -C(O)-alkyleneCi-c4-O-alkylCi-c6, -C(O)-pyrrolidine, and -C(O)-pyrrolidine-alkylci-c6; and, each R19 is independently selected from H and alkylCi-c6; each R22 is selected from H, alkylCi-c6, -0-alkylCi-c6, -alkyleneci-cδ-O-alkylci-ce; and, each R23 is selected from H and alkylCi-c6; or
R22 and R23 combine to form a four, five, or six membered, heterocyclic ring containing the N atom to which they are attached and optionally an additional heteroatom selected from N and O, wherein the ring is optionally substituted with -OH or -alkyld-ce.
2. The compound of claim 1 , wherein R7 is -O-alkylci-cβ-
3. The compound of claim 1 or 2, wherein R7 is -O-methyl.
The compound of claim 1 having a formula (Ia)
Figure imgf000304_0001
The compound of claim 1 having a formula (Ib)
Figure imgf000304_0002
(Ib).
6. The compound of claim 1 having a formula (Ic)
Figure imgf000305_0001
7. The compound of claim 1 having a formula (Id)
Figure imgf000305_0002
8. The compound of any of claims 1-7, wherein R9 and R10, together with the atoms to which they are attached form a five, six, or seven-membered heterocyclic ring containing one or two N atom and the remainder C atoms, wherein at least one N atom is substituted with R15, and the C atoms of the heterocyclic ring are optionally substituted with one or more groups selected from (R19)i-2.
9. The compound of any of claims 1-8, wherein R9 and R10, together with the atoms to which they are attached form a five or six-membered heterocyclic ring selected from
Figure imgf000306_0001
10. The compound of any of claims 1-9, wherein R4a and R4b, are H.
11. A pharmaceutical composition comprising a compound according to any of claims 1 through 10 and a pharmaceutically acceptable carrier, diluent or excipient.
12. The pharmaceutical composition according to claim 11 further comprising a chemotherapeutic agent.
13. A method for treating a susceptible neoplasm in an mammal in need thereof, said method comprising administering to the mammal a therapeutically effective amount of a compound according to any of claims 1 through 10.
14. The method according to claim 13, wherein said susceptible neoplasm is selected from breast cancer, sarcomas, lung cancer, prostate cancer, colorectal cancer, renal cancer, pancreatic cancer, hematologic cancers, neuroblastomas, gliomas, head and neck cancer, thyroid cancer, hepatocarcinoma, ovarian cancer, vulval cancer, cervical cancer, endometrial cancer, testicular cancer, bladder cancer, esophageal cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, gastrointestinal stromal tumor and skin cancer.
15. A compound according to any of claims 1 through 10 for use in the treatment of a condition mediated by IGF-1 R in a mammal.
16. The compound according to any of claims 1 through 10 for use in the treatment of a susceptible neoplasm.
17. The compound according to claim 16, wherein the susceptible neoplasm is selected from breast cancer, sarcomas, lung cancer (including non-small cell lung carcinoma), prostate cancer, colorectal cancer, renal cancer, pancreatic cancer, hematologic cancers (including multiple myeloma), neuroblastomas, gliomas, head and neck cancer, thyroid cancer, hepatocarcinoma, ovarian cancer, vulval cancer, cervical cancer, endometrial cancer, testicular cancer, bladder cancer, esophageal cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, gastrointestinal stromal tumor and skin cancer (including melanoma).
18. The use of a compound according to any of claims 1 through 10 for the preparation of a medicament for the treatment of a condition mediated by IGF-1 R in a mammal.
19. The use of a compound according to any of claims 1 through 10 for the preparation of a medicament for the treatment of a susceptible neoplasm in a mammal.
20. The use according to claim 19, wherein the susceptible neoplasm is selected from breast cancer, sarcomas, lung cancer (including non-small cell lung carcinoma), prostate cancer, colorectal cancer, renal cancer, pancreatic cancer, hematologic cancers (including multiple myeloma), neuroblastomas, gliomas, head and neck cancer, thyroid cancer, hepatocarcinoma, ovarian cancer, vulval cancer, cervical cancer, endometrial cancer, testicular cancer, bladder cancer, esophageal cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, gastrointestinal stromal tumor and skin cancer (including melanoma).
21. A pharmaceutical composition for use in the treatment of the susceptible neoplasm selected from breast cancer, sarcomas, lung cancer (including non-small cell lung carcinoma), prostate cancer, colorectal cancer, renal cancer, pancreatic cancer, hematologic cancers (including multiple myeloma), neuroblastomas, gliomas, head and neck cancer, thyroid cancer, hepatocarcinoma, ovarian cancer, vulval cancer, cervical cancer, endometrial cancer, testicular cancer, bladder cancer, esophageal cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, gastrointestinal stromal tumor and skin cancer (including melanoma), wherein the composition comprises a compound of any of claims 1 through 10.
PCT/US2009/060830 2008-10-16 2009-10-15 Pyrrolopyrimidine compounds WO2010045451A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10583108P 2008-10-16 2008-10-16
US61/105,831 2008-10-16

Publications (1)

Publication Number Publication Date
WO2010045451A1 true WO2010045451A1 (en) 2010-04-22

Family

ID=42106890

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/060830 WO2010045451A1 (en) 2008-10-16 2009-10-15 Pyrrolopyrimidine compounds

Country Status (1)

Country Link
WO (1) WO2010045451A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012045195A1 (en) * 2010-10-09 2012-04-12 Abbott Laboratories Pyrrolopyrimidines as fak and alk inhibiters for treatment of cancers and other diseases
JP2013501713A (en) * 2009-08-07 2013-01-17 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Novel azaheterocyclic compounds
US8685988B2 (en) 2012-08-06 2014-04-01 Acea Biosciences, Inc. EGFR modulators and uses thereof
WO2014080640A1 (en) 2012-11-26 2014-05-30 国立大学法人東北大学 Erythropoietin expression promoter
CN105461616A (en) * 2015-02-27 2016-04-06 上海麦步医药科技有限公司 New synthesis technology of ceritinib intermediate
US9464089B2 (en) 2012-01-13 2016-10-11 Acea Biosciences Inc. Heterocyclic compounds and uses thereof
US9586965B2 (en) 2012-01-13 2017-03-07 Acea Biosciences Inc. Pyrrolo[2,3-d]pyrimidine compounds as inhibitors of protein kinases
US20180244676A1 (en) * 2015-02-13 2018-08-30 Dana-Farber Cancer Institute, Inc. Lrrk2 inhibitors and methods of making and using the same
US10533011B2 (en) 2015-10-09 2020-01-14 ACEA Therapeutics, Inc. Pharmaceutical salts, physical forms, and compositions of pyrrolopyrimidine kinase inhibitors, and methods of making same
US10562918B2 (en) 2013-07-11 2020-02-18 ACEA Therapeutics, Inc. Heterocyclic compounds and uses thereof
US10596174B2 (en) 2012-01-13 2020-03-24 ACEA Therapeutics, Inc. Pyrrolopyrimidine compounds as inhibitors of protein kinases
US11498922B2 (en) 2017-04-07 2022-11-15 ACEA Therapeutics, Inc. Pharmaceutical composition comprising N-(3-((2-((3-fluoro-4-(4-methylpiperazin-1-yl phenyl)amino)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)oxy)phenylacrylamide

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060211678A1 (en) * 2004-08-02 2006-09-21 Saleh Ahmed Aryl-amino substituted pyrrolopyrimidine multi-kinase inhibiting compounds
WO2007140222A2 (en) * 2006-05-26 2007-12-06 Novartis Ag Pyrrolopyrimidine compounds and their uses

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060211678A1 (en) * 2004-08-02 2006-09-21 Saleh Ahmed Aryl-amino substituted pyrrolopyrimidine multi-kinase inhibiting compounds
WO2007140222A2 (en) * 2006-05-26 2007-12-06 Novartis Ag Pyrrolopyrimidine compounds and their uses

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2013501713A (en) * 2009-08-07 2013-01-17 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Novel azaheterocyclic compounds
WO2012045195A1 (en) * 2010-10-09 2012-04-12 Abbott Laboratories Pyrrolopyrimidines as fak and alk inhibiters for treatment of cancers and other diseases
US9920074B2 (en) 2012-01-13 2018-03-20 Acea Biosciences Inc. Heterocyclic compounds and uses thereof
US9034885B2 (en) 2012-01-13 2015-05-19 Acea Biosciences Inc. EGFR modulators and uses thereof
US11612602B2 (en) 2012-01-13 2023-03-28 ACEA Therapeutics, Inc. Heterocyclic compounds and uses as anticancer agents
US9464089B2 (en) 2012-01-13 2016-10-11 Acea Biosciences Inc. Heterocyclic compounds and uses thereof
US9586965B2 (en) 2012-01-13 2017-03-07 Acea Biosciences Inc. Pyrrolo[2,3-d]pyrimidine compounds as inhibitors of protein kinases
US9763949B2 (en) 2012-01-13 2017-09-19 Acea Biosciences Inc. EGFR modulators and uses thereof
US10799504B2 (en) 2012-01-13 2020-10-13 ACEA Therapeutics, Inc. Heterocyclic compounds and uses as anticancer agents
US10596174B2 (en) 2012-01-13 2020-03-24 ACEA Therapeutics, Inc. Pyrrolopyrimidine compounds as inhibitors of protein kinases
US8685988B2 (en) 2012-08-06 2014-04-01 Acea Biosciences, Inc. EGFR modulators and uses thereof
US10449196B2 (en) 2012-08-06 2019-10-22 ACEA Therapeutics, Inc. EGFR modulators and uses thereof
US11007197B2 (en) 2012-08-06 2021-05-18 ACEA Therapeutics, Inc. EGFR modulators and uses thereof
WO2014080640A1 (en) 2012-11-26 2014-05-30 国立大学法人東北大学 Erythropoietin expression promoter
US10562918B2 (en) 2013-07-11 2020-02-18 ACEA Therapeutics, Inc. Heterocyclic compounds and uses thereof
US20180244676A1 (en) * 2015-02-13 2018-08-30 Dana-Farber Cancer Institute, Inc. Lrrk2 inhibitors and methods of making and using the same
US10913744B2 (en) * 2015-02-13 2021-02-09 Dana-Farber Cancer Institute, Inc. LRRK2 inhibitors and methods of making and using the same
CN105461616A (en) * 2015-02-27 2016-04-06 上海麦步医药科技有限公司 New synthesis technology of ceritinib intermediate
US10533011B2 (en) 2015-10-09 2020-01-14 ACEA Therapeutics, Inc. Pharmaceutical salts, physical forms, and compositions of pyrrolopyrimidine kinase inhibitors, and methods of making same
US11498922B2 (en) 2017-04-07 2022-11-15 ACEA Therapeutics, Inc. Pharmaceutical composition comprising N-(3-((2-((3-fluoro-4-(4-methylpiperazin-1-yl phenyl)amino)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)oxy)phenylacrylamide

Similar Documents

Publication Publication Date Title
WO2010045451A1 (en) Pyrrolopyrimidine compounds
EP1828185B1 (en) 2-pyrimidinyl pyrazolopyridine erbb kinase inhibitors
EP2162454B1 (en) Imidazopyridine kinase inhibitors
US7981903B2 (en) 2-[2-{phenylamino}-1H-pyrrolo[2,3-D]pyrimidin-4-yl)amino] benzamide derivatives as IGF-1R inhibitors for the treatment of cancer
EP1971606B1 (en) 2-pyrimidinyl pyrazolopyridine erbb kinase inhibitors
ES2325440T3 (en) PIRIMIDINE COMPOUNDS.
EP3060563B1 (en) Ring-fused bicyclic pyridyl derivatives as fgfr4 inhibitors
JP4705695B2 (en) Pyrrolo [2,3-D] pyrimidine derivatives as protein kinase B inhibitors
ES2649144T3 (en) Heteroaromatic compounds and their use as dopamine D1 ligands
WO2012098068A1 (en) Pyrazolo pyrimidines as dyrk1a and dyrk1b inhibitors
CA2933480A1 (en) Inhibitors of lysine specific demethylase-1
JP2002504552A (en) 6,9-disubstituted 2- [trans- (4-aminocyclohexyl) amino] purine
KR20090104030A (en) Substituted piperidines containing a heteroarylamide or heteroarylphenyl moiety
EP3313852B1 (en) Substituted pyrazolo/imidazolo bicyclic compounds as pde2 inhibitors
US7812022B2 (en) 2-pyrimidinyl pyrazolopyridine ErbB kinase inhibitors
CA2716202A1 (en) Pyrrolo[2,3-d]pyrimidin-2-yl-amine derivatives as pkc-theta inhibitors

Legal Events

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

Ref document number: 09821249

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: 09821249

Country of ref document: EP

Kind code of ref document: A1