YKL-40 INHIBITORS AND THEIR THERAPEUTIC APPLICATIONS

Abstract
This invention is related to novel inhibitors of chitinase-like protein YKL-40 and their therapeutic applications. Such YKL-40 inhibitors are of use as therapeutic agents for the treatment a variety of diseases and disorders, including cancers, inflammation, and pulmonary diseases.
Description
FIELD OF THE INVENTION

This invention is related to novel inhibitors of chitinase-like protein YKL-40, and their therapeutic applications.


BACKGROUND OF THE INVENTION

The glycosyl hydrolase family 18 (GH 18) is an ancient gene family that is widely expressed in species as diverse as plants, insects and mammals. GH 18 protein family in humans, consists of two enzymatically active chitinases, CHIT1 and AMCase, and the non-active chitinase-like proteins (CLPs): YKL-40, YKL-39, oviductin and stabilin-interacting protein that lack enzymatic activity due to amino acid substitutions in the active site. Although chitinases and CLPs are evolutionary highly conserved, their physiological role in mammals has not been fully elucidated. In most mammals, e.g., rodents, chitinases protect against chitin-containing organisms which are either inhaled or ingested. Mammals are not known to synthesize chitin or metabolize it as a nutrient, yet the human genome encodes eight GH18 family members (Funkhouser et al., Chitinase family GH18: evolutionary insights from the genomic history of a diverse protein family. BMC Evol Biol. June 26:7-96, 2007). In humans, chitinases and CLPs are thought to have evolved to perform different functions, often associated with inflammatory and fibrotic pathologies as well as tissue remodeling and cancer progression (Lee et al., Role of chitin and chitinase/chitinase-like proteins in inflammation, tissue remodeling, and injury. Annu Rev Physiol. 73:479-501, 2011).


Chitinase-3-like protein 1 (CHI3L1), also known as YKL-40 is a secreted glycoprotein that was first identified in culture media of a human osteosarcoma cell line MG63 (Johansen et al., Identification of proteins secreted by human osteoblastic cells in culture. Journal of Bone & Mineral Research 7:501-512, 1992). YKL-40 protein is approximately 40 kDa in size that in humans is encoded by the CHI3L1 gene, located on chromosome 1931-1932 and consists of 10 exons and spans about 8 kilobases of genomic DNA (Rehli et al., Molecular characterization of the gene for human cartilage gp-39 (CHI3L1), a member of the chitinase protein family and marker for late stages of macrophage differentiation. Genomics. 43(2):221-5, 1997). The name YKL-40 derives from the three N-terminal amino acids present on the secreted form and its molecular mass in kilodaltons. YKL-40 lacks chitinase activity due to mutations of one of the three amino acids (Asp, Glu and Asp) essential for the chitinase-like catalytic activity, namely Glu->Leu, which completely deprives chitinase activity (Renkema et al., Chitotriosidase, a chitinase, and the 39-kDa human cartilage glycoprotein, a chitin-binding lectin, are homologues of family 18 glycosyl hydrolases secreted by human macrophages. Eur J Biochem. 251: 504-509, 1998). YKL-40 has been shown to bind chitin oligomers, chitin-like oligosaccharides, hyalurorian, heparin as well as heparan sulfate. However, the biological role of YKL-40 still remains unclear. YKL-40 is not even known to have a specific receptor, although it has been proposed to act through IL13Rα2 (Chuanet at al., Chitinase 3-like 1 Regulates Cellular and Tissue Responses via IL-13 Receptor α2. Cell Rep. 4(4): 830-841, 2013) and Syndecan-1 (Shao et al., YKL-40, a secreted glycoprotein, promotes tumor angiogenesis, Oncogene. 28:4456-4468, 2009), CD44 (Geng et al. Chitinase 3-like 1-CD44 interaction promotes metastasis and epithelial-to-mesenchymal transition through p-catenin/Erk/Akt signaling in gastric cancer, J Exp Clin Cancer Res 37:208, 2018, RAGE (Low et al., Chitinase 3-like 1 induces survival and proliferation of intestinal epithelial cells during chronic inflammation and colitis-associated cancer by regulating S100A9. Oncotarget; 6:36535-50 2015) receptors and other proteins as lectin—Galectin3 (Bouvet et al., Peripheral blood mononuclear cell response to YKL-40 and Galectin-3 in cystic fibrosis. Cytokine. 146:155635, 2021) via respectively chitin-binding and heparin-binding domains but its detailed biological functions are still poorly understood.


Despite the fact that the exact function of YKL-40 is still unknown, the pattern of YKL-40 expression is associated with pathogenic processes related to inflammation (including CNS), extracellular tissue remodeling, fibrosis oncogenesis. YKL-40 is expressed and secreted by various cell-types including macrophages, chondrocytes, fibroblast-like synovial cells, vascular smooth muscle cells, hepatic stellate cells as well as cancer cells. In accordance to these observations, elevated levels of YKL-40 protein have been detected in a wide range of diseases, comprising of infections, arthritis, inflammatory bowel disease, chronic obstructive lung disease, idiopathic pulmonary fibrosis, asthma, nonalcoholic steatohepatitis, hepatitis, diabetes, atherosclerosis, giant cell arteritis (Lee et al., Chitin, chitinases and chitinase-like proteins in allergic inflammation and tissue remodeling. Yonsei Med J 50:22-30, 2009; Lee et al., Role of chitin and chitinase/chitinase-like proteins in inflammation, tissue remodeling and injury. Annu Rev Physiol 73:479-501, 2011; Lee et al., Role of breast regression protein-39/YKL-40 in asthma and allergic responses. Allergy Asthma Immunol Res 2:20-7, 2010; 7. Lee et al. Role of breast regression protein 39 (BRP-39)/chitinase 3-like-1 in Th2 and IL-13-induced tissue responses and apoptosis. J Exp Med 206:1149-66, 2009; Dela Cruz et al., Chitinase 3-like-1 (Chi311) Regulation of Streptococcus pneumoniae Lung Infection. Cell Host Microbe 12:34-4, 2012; Johansen et al., A new biochemical marker for joint injury. Analysis of YKL-40 in serum and synovial fluid. Br J Rheumatol 32:949-55, 1993), neurodegenerative diseases like Alzheimer disease (Muszynski et al., YKL-40 as a Potential Biomarker and a Possible Target in Therapeutic Strategies of Alzheimer's Disease. Curr Neuropharmacol. 15(6):906-917, 2017), cancer and many others.


Cancer is a group of diseases caused by abnormal and uncontrolled cell proliferation with the potential to invade or spread to other organs of the body. YKL-40 is produced and secreted by several types of human cancer cells and tumor-associated macrophages as well as cancer-associated fibroblasts. It has been postulated that it may play a role in cell proliferation, differentiation, cell survival, invasiveness, metastasis, in angiogenesis, inflammatory response to cancer and tissue remodeling. YKL-40 serum protein level is considered as prognostic biomarker in a variety of different cancer types (breast, colon, lung, kidney, ovarian, prostate, uterine, osteosarcoma, oligodendroglioma, glioblastoma and germ cell tumors), elevated serum YKL-40 level has been correlated with poor prognosis and shorter survival of patients with cancer (Johansen et al., Is YKL-40 a new therapeutic target in cancer? Expert Opin Ther Targets. 11(2):219-34, 2017). Studies in animal models have demonstrated that YKL-40 overexpression enhances breast (MDA-MB-231 cells) and colon (HCT116 cells) tumor growth and angiogenesis in vivo, on the other hand, U87 glioblastoma cancer cells expressing YKL-40 siRNA abrogated tumor angiogenesis and tumor growth in vivo (Shao et al., YKL-40, a secreted glycoprotein, promotes tumor angiogenesis. Oncogene 28(50): 4456-4468, 2009). It has been also reported that YKL-40 neutralizing antibodies are effective in blocking tumor angiogenesis and tumor progression (Faibish et al., A YKL-40-neutralizing antibody blocks tumor angiogenesis and progression: a potential therapeutic agent in cancers. Mol Cancer Ther. 10(5):742-51, 2011). Also several small molecules targeting YKL-40 were shown to suppress tumor growth and metastasis in lung, osteosarcoma, melanoma and breast cancer models (Park, et al. G721-0282 inhibits cell growth and induces apoptosis in human osteosarcoma through down-regulation of the STAT3 pathway. International journal of biological sciences 16, 2, 330-341, 2020; Lee et al., A small molecule targeting CHI3L1 inhibits lung metastasis by blocking IL-13Rα2-mediated JNK-AP-1 signals. Mol Oncol. 16(2):508-526, 2022).


Chitinase and chitinase-like proteins are over expressed in many cancers including brain tumors such as glioblastoma (Francescone et al. J Biol Chem 2011; 286:15332-43; Ku et al. Int J Cancer 2011; 128:1316-26) or astrocytoma (Zhang et al. Cancer. 2010; 116:2688), breast cancer (Johansen et al. Breast Cancer Res Treat 2003; 80:15-21), colon cancer (Nutt et al., 2005, Pelloski et al., 2005; Fijneman et al. Clin Cancer Res. 2012; 18:2613; Chen et al. Am J Pathol. 2011; 179:1494), primary and metastatic lung cancer (Wang et al. Tumour Biol 2015; 36:901-7; Johansen et al. Lung Cancer 2004; 46:333-40), mesothelioma (Corradi et al. Anticancer Res. 2013 December; 33(12):5517), osteosarcoma, malignant melanoma (Ma et al. Cancer Res 2015; 75:487-96), ovarian cancer (Hogdall et al. BMC Cancer 2009; 9:8; Dupont et al. J Clin Oncol. 2004; 22:3330), cervical cancer (Ngernyuang et al. Int J Biochem Cell Biol 2014; 51:45-52), prostate cancer (Jeet et al. Endocr Relat Cancer. 2014; 21:723), liver cancer (Pan et al. J Cancer Res Clin Oncol 2013; 139:1043-54), gastric cancer (Li et al. Chin Med J 2012; 125:1777), metastatic renal cancer (Zhangg et al. Tumour Biol 2014; 35:12131-7), hematologic malignancies such as leukemia or lymphoma (Mactier et al. J Proteome Res. 2011; 10:1030; Marchesi et al. Vet Pathol. 2006; 43:773-6; Marchesi et al. J Vet Med A Physiol Pathol Clin Med. 2003; 50:103) and other types of cancers with inflammatory background (Quershi et al. Genes Cancer. 2011; 2:74; Eurich et al. World J Gastroenterol. 2009; 15:5249; Roslind and Johansen, Methods of Mol Biol. 2009; 511:159). In fact, higher plasma levels indicate poor prognosis and increased metastatic potential for wide range of cancers (Johansen et al., Cancer Epidemiol. Biomarkers Prev., 15(2):194-202, 2006). Inhibition of chitinase and chitinase-like protein biological function with one or more compounds described in this invention is anticipated to have therapeutic utility in subjects with cancer.


Interstitial lung diseases (ILDs) is a group of over 300 lung disorders which affect lung interstitium: the most common are sarcoidosis and idiopathic pulmonary fibrosis (IPF). These diseases, many with unknown etiology, are characterized by the alveolar damage which often leads to a chronic inflammation and fibrosis resulting in diminished lung functions.


Sarcoidosis is a multiorgan systemic disease characterized by a formation of granulomas that can develop in various organs, with most patients developing pulmonary presentation. Spontaneous remission occurs in a majority of cases, but up to one-third of patients develop a chronic, progressive or relapsing disease with a concomitant interstitial fibrosis and decline in lung functions (Valeyre et al., Sarcoidosis. Lancet 383(9923):1155-67, 2014). The clinical studies demonstrated highly elevated YKL-40 serum levels in sarcoidosis patients compared to controls (Long et al., Serum YKL-40 as predictor of outcome in hypersensitivity pneumonitis. Eur Respir J. 23:49-51, 2017) and YKL-40 serum protein level has been postulated as new sarcoidosis marker (Johansen et al., Increased serum YKL-40 in patients with pulmonary sarcoidosis-a potential marker of disease activity? Respir Med. 99(4):396-402, 2005).


IPF is a progressive fibroproliferative disorder with no curative therapies with a median survival of only 3-5 years following diagnosis. IPF is a devastating disease characterized by excessive matrix deposition that disrupts the normal architecture of the lung parenchyma and impairs lung functions. The key pathological features of IPF include fibroblastic foci, areas of epithelial cysts associated with the honeycombing appearance of the lung, and mild lymphoplasmacytic interstitial inflammation that is associated with areas of type II cell hyperplasia (Richeldi et al., Idiopathic pulmonary fibrosis. Lancet 389(10082):1941-1952, 2017). A series of clinical studies have demonstrated an increased YKL-40 protein levels in serum and bronchoalveolar lavage fluid (BALF) of patients with idiopathic pulmonary fibrosis (Furuhashi et al., Increased expression of YKL-40, a chitinase-like protein, in serum and lung of patients with idiopathic pulmonary fibrosis. Respir Med. 104(8):1204-10, 2010; Vij et al., Peripheral blood biomarkers in idiopathic pulmonary fibrosis. Transl Res. 159(4):218-27, 2012) suggesting that YKL-40 protein might be involved in remodeling and tissue damage seen in lungs from IPF patients. Furthermore, elevated YKL-40 serum and BALF level are considered as predictors of survival in idiopathic pulmonary fibrosis (Korthagen et al., Serum and BALF YKL-40 levels are predictors of survival in idiopathic pulmonary fibrosis. Respir Med. 2011 January; 105(1):106-13).


Obstructive lung diseases (among them asthma and chronic obstructive lung disease COPD) are chronic inflammatory disorders that involve the small airways and cause airflow limitation. Asthma is characterized by recurrent episodes of reversible airway obstruction and airway hyper responsiveness. Typical clinical manifestations include shortness of breath, wheezing, coughing and chest tightness that can become life threatening or fatal. While existing therapies focus on reducing the symptomatic bronchospasm and pulmonary inflammation, there is a growing recognition of the role of long-term airway remodeling in the accelerated lung deterioration in asthmatics. (Papi et al. Asthma. Lancet 391(10122):783-800, 2018). COPD is a disease characterized by a progressive, irreversible limitation of airflow due to emphysema and remodeling of small airways. Currently, no drugs reducing COPD progression are available with smoking cessation the only intervention demonstrated to slow the rate of decline in lung function. (Rabe et al. Chronic obstructive pulmonary disease. Lancet 389(10082):1931-1940, 2017). Multiple studies have demonstrated that YKL-40 may be implicated in bronchial inflammation and remodeling in COPD and its elevated level may be considered as a useful biomarker for COPD diagnosis and monitoring (Xiang et al., The YKL-40 protein is a potential biomarker for COPD: a meta-analysis and systematic review. Int J Chron Obstruct Pulmon Dis. 13: 409-418, 2018). Another meta-analysis suggested that YKL-40 protein may play an important role in the pathogenesis of asthma and its serum levels may correlate with exacerbation attacks and severity as well (Xiang et al., The serum YKL-40 is a useful biomarker for asthma. European Respiratory Journal 50: PA3566, 2017).


There is a need for novel active compounds interfering with interactions of YKL-40 and other biomolecules such as chitin oligomers and heparins. Such active compounds could act as YKL-40 inhibitors, and be of use as therapeutic agents for the treatment a variety of diseases and disorders, including cancers, inflammation, and pulmonary diseases. Alternative way of targeting YKL-40 is to use bispecific small molecule degraders that bind strongly to YKL-40 and simultaneously to one of proteins from protein degradation machinery. The mechanism of action of such degraders is to bring into close proximity these two proteins and trigger degradation of YKL-40.


SUMMARY OF THE INVENTION

In one aspect, this invention concerns a novel compound represented by structural formula (I),




embedded image


wherein:

    • W represents —C(—R1f)(—R1g)—;
    • X represents —C(—R1h)(—R1j)—;
    • Y represents —C(—R1k)(—R1m)—, —N(—R1k)—, or —O—;
    • R1b represents —H;
    • R1c represents —H;
    • R1d represents 4-R10-benzyl;
    • R1e represents —H;
    • R1f represents —H;
    • R1g represents —H;
    • R1h is selected from the group consisting of C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, hydroxy-C1-C6 alkyl, azido-C1-C6 alkyl, hydroxycarbonyl-C1-C3 alkoxy-C1-C6 alkyl, C3-C6 alkynyloxy-C1-C6 alkyl, halo-C1-C6 alkyl, C1-C3 alkylthio-C1-C6 alkyl, C1-C3 alkylsulfinyl-C1-C6 alkyl, C1-C3 alkylsulfonyl-C1-C6 alkyl, hydroxycarbonyl-C1-C3 alkylsulfonyl-C1-C6 alkyl, hydroxycarbonyl-C1-C6 alkyl, C1-C3 alkylcarbonyl-C1-C6 alkyl, C1-C3 alkylaminocarbonyl-C1-C6 alkyl, di(C1-C3 alkyl)aminocarbonyl-C1-C6 alkyl, C1-C3 alkylsulfonylamino-C1-C6 alkyl, halo-C1-C3 alkylsulfonylamino-C1-C6 alkyl, 5- or 6-membered monocyclic heteroaryl-carbonylamino-C1-C6 alkyl, 5- or 6-membered monocyclic heteroaryl-C1-C3 alkoxy-C1-C6 alkyl, 5- or 6-membered monocyclic heteroaryl-C1-C6 alkyl, hydroxycarbonyl, C1-C3 alkoxycarbonyl, aminocarbonyl, C1-C3 alkylaminocarbonyl, di(C1-C3 alkyl)aminocarbonyl, C3-C6 alkynylaminocarbonyl, hydroxycarbonyl-C1-C3 alkyl(C1-C3 alkyl)aminocarbonyl, (biotinyl-PEG11)aminocarbonyl, (biotinyl-PEG11)aminocarbonyl-C1-C3 alkylsulfonyl-C1-C6 alkyl, hydroxylaminocarbonyl, N2-acetylhydrazinecarbonyl, (5- or 6-membered monocyclic heteroaryl)-C1-C3 alkylaminocarbonyl, (5- or 6-membered monocyclic heteroaryl)aminocarbonyl, C1-C3 alkylsulfonyl, 5- or 6-membered monocyclic heteroaryl, (3-azabicyclo[3.1.0]hexan-3-yl)-C1-C3 alkyl, (4-, 5-, or 6-membered heterocyclyl)amino-C1-C3 alkyl, hydroxycarbonyl-C1-C6 alkyl-(5- or 6-membered monocyclic heteroaryl), R20, and 5-(2-(2,6-dioxopiperidin-3-yl)-3-oxoisoindolin-4-yl)pentyl, where any available position of any heteroaryl ring in R1h can be substituted with a substituent selected from the group consisting of C1-C6 alkyl, C3-C6 alkynyl, halo, trifluoromethyl, 2,2,2-trifluoroethyl, biotinyl-PEG11, and any available position of any carbon chain in R1h excluding heteroaryl rings can be substituted with a substituent selected from halogen, amino, hydroxy, and oxo;
    • R1j represents —H;
    • R1k represents —H;
    • R1m represents —H;
    • or R1k and R1h taken along with nitrogen and carbon atoms bearing them, respectively, represent a 6-membered heterocyclic ring of structural formula (II):




embedded image




    • R3a represents —H;

    • R3b represents —H;

    • R3c represents —H;

    • R3d represents —H;

    • R3e represents —H;

    • R3f is selected from the group consisting of 1-methyl-1H-imidazol-4-yl, 1,2-dimethyl-1H-imidazol-4-yl, 5-methyloxazol-2-yl, 4,5-dimethyloxazol-2-yl, 1H-pyrazol-1-yl, 1-methyl-1H-pyrazol-3-yl, 4-methyl-1H-pyrazol-1-yl, 4-fluoro-1H-pyrazol-1-yl, 1-isopropyl-1H-pyrazol-3-yl, 5-methyl-1H-pyrazol-3-yl, 1,5-dimethyl-1H-pyrazol-3-yl, 1-ethyl-5-methyl-1H-pyrazol-3-yl, 1,5-dimethyl-1H-pyrazol-3-yl, 1,5-dimethyl-1H-pyrazol-4-yl, 1-methyl-5-(trifluoromethyl)-1H-pyrazol-3-yl, 1-methyl-2-cyano-1H-pyrrol-4-yl, 1H-tetrazol-5-yl, 2-methyl-2H-tetrazol-5-yl, 5-methyl-1,3,4-thiadiazol-2-yl, 5-methylthiazol-2-yl, 4,5-dimethylthiophen-2-yl, 1H-1,2,4-triazol-1-yl, 1-methyl-1H-1,2,3-triazol-4-yl, 1-methyl-1H-1,2,4-triazol-3-yl, 2-methyl-2H-1,2,3-triazol-4-yl, 1,5-dimethyl-1H-1,2,4-triazol-3-yl, 5-hydroxy-1-methyl-1H-1,2,4-triazol-3-yl, and 5-methoxy-1-methyl-1H-1,2,4-triazol-3-yl;

    • R3g represents —H;

    • R3h represents —H;

    • R3j represents —H;

    • R3k represents —H;

    • R3m represents —H;

    • or R3d and R3g taken along with the single bond linking two carbon atoms bearing them represent a double bond between these two carbon atoms;

    • R4b represents —H;

    • R4c represents —H;

    • R4f represents —H;

    • R4g represents —H;

    • R4h represents —H;

    • R4j represents —H;

    • R10 represents halogen or trifluoromethyl;

    • and R20 represents







embedded image


or a tautomer, stereoisomer, N-oxide, methiodide, pharmaceutically acceptable salt, solvate, or polymorph thereof.


Another aspect of the invention is directed to a pharmaceutical composition comprising a therapeutically effective amount of at least one compound according to the invention and a pharmaceutically acceptable carrier therefor.


Yet another aspect of the invention concerns a method for inhibiting the chitinase-like protein YKL-40 in a cell or a tissue, comprising contacting the cell or the tissue with at least one compound according to the invention.


Yet another aspect of the invention concerns a method for degrading the chitinase-like protein YKL-40 in a cell or a tissue, comprising contacting the cell or the tissue with at least one compound according to the invention.


The invention provides also a method for the treatment or prevention of a disease, disorder, or condition associated with aberrant expression or activity of the chitinase-like protein YKL-40, comprising administering to a subject in need thereof a therapeutically effective amount of at least one compound according to the invention, or a pharmaceutical composition according to the invention.







DETAILED DESCRIPTION OF THE INVENTION
Definitions

The articles “a” and “an” are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element.


The terms “substituent,” “group,” and “radical” are used interchangeably herein and denote a portion of a molecule in question which is bound to the rest of the molecule by a covalent bond (or bonds, as it results from the previous paragraph).


The terms used herein may be preceded and/or followed by a single dash “—”, or a double dash “═”, to indicate the bond order of the bond between the named substituent and its parent moiety; a single dash indicates a single bond and a double dash indicates a double bond. In the absence of a single or double dash, it is understood that a single bond is formed between the substituent and its parent moiety; further, substituents are intended to be read “from left to right,” unless a dash indicates otherwise. For example, C1-C6 alkylcarboxy and —OC(O)—C1-C6 alkyl indicate the same functionality; similarly, arylalkyl and -alkylaryl indicate the same functionality.


The term “single bond” as used herein, denotes a single covalent bond between two atoms, such as C—C, C—H, or C—O.


The term “alkyl” as used herein is a term of art and refers to saturated aliphatic groups, including straight-chain alkyl groups and branched-chain alkyl groups. The subscripts following C indicate the number (or range of numbers) of carbon atoms in the straight-chain or branched-chain alkyl. From the practical reasons, the number of carbon atoms in a straight-chain alkyl is limited to the range of 1-8, inclusive, and the number of carbon atoms in a branched-chain alkyl is in the range of 3-8, inclusive. If there is no subscript specified defining the number of carbon atoms in an alkyl group, then this number is not greater than 8. Representative examples of C1-C6 alkyl include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, isopentyl, neopentyl, and n-hexyl. Examples of C1-C3 alkyl include methyl, ethyl, n-propyl, and isopropyl. Alkyl may represent a group, as already defined, or a portion of a larger moiety, such as C1-C3 alkoxy-C1-C3 alkyl. A C1-C3 alkoxy-C1-C3 alkyl is bound to the rest of the molecule through the C1-C3 alkyl moiety.


If the name of particular alkyl group appears in the name of a chemical compound, the prefixes such as iso-, sec-, tert-, or neo-, indicate an appropriately branched alkyl, as understood by the person skilled in the art; the prefix n- or lack of any prefix means that the alkyl group in question is a straight-chain alkyl.


The term “cycloalkyl” means a radical derived from mono- or bicyclic or bridged saturated or partially saturated carbocyclic rings, each having from 3 to 12 carbon atoms. Certain cycloalkyls have from 3-8, or from 3-6 carbon atoms in their ring structure. Certain cycloalkyls have from 5-12 carbon atoms in their ring structure, and may have 6-10 carbons in the ring structure. Preferably, cycloalkyl is C3-C7 cycloalkyl, which represents a monocyclic saturated carbocyclic ring, having from 3 to 7 carbon atoms. Examples of monocyclic cycloalkyls include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, and cyclooctyl. Bicyclic cycloalkyl ring systems include bridged monocyclic rings and fused bicyclic rings. Bridged monocyclic rings contain a monocyclic cycloalkyl ring where two non-adjacent carbon atoms of the monocyclic ring are linked by an alkylene bridge of between one and three additional carbon atoms (i.e., a bridging group of the form —(CH2)w—, where w is 1, 2, or 3). Representative examples of bicyclic ring systems include, but are not limited to, bicyclo[3.1.1]heptane, bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane, bicyclo[3.2.2]nonane, bicyclo[3.3.1]nonane, and bicyclo[4.2.1]nonane. Fused bicyclic cycloalkyl ring systems contain a monocyclic cycloalkyl ring fused to either a phenyl, a monocyclic cycloalkyl, a monocyclic cycloalkenyl, a monocyclic heterocyclyl, or a monocyclic heteroaryl. The bridged or fused bicyclic cycloalkyl is attached to the parent molecular moiety through any carbon atom contained within the monocyclic cycloalkyl ring. Cycloalkyl groups are optionally substituted. In certain embodiments, the fused bicyclic cycloalkyl is a 5 or 6 membered monocyclic cycloalkyl ring fused to either a phenyl ring, a 5 or 6 membered monocyclic cycloalkyl, a 5 or 6 membered monocyclic cycloalkenyl, a 5 or 6 membered monocyclic heterocyclyl, or a 5 or 6 membered monocyclic heteroaryl, wherein the fused bicyclic cycloalkyl is optionally substituted.


The term “cycloalkylalkyl” as used herein refers to an alkyl group substituted with one or more cycloalkyl groups. An example of cycloalkylalkyl is cyclohexylmethyl group.


The terms “heterocyclyl” and “heterocycloalkyl” as used herein refers to a radical of a non-aromatic ring system, including, but not limited to, monocyclic, bicyclic, and tricyclic rings, which can be completely saturated or which can contain one or more units of unsaturation, for the avoidance of doubt, the degree of unsaturation does not result in an aromatic ring system, and having 3 to 14, or 3 to 12 atoms other than hydrogen, including at least one heteroatom, such as nitrogen, oxygen, or sulfur. More preferred heterocycloalkyl groups have from 5-10 ring members where from 1-4 of the ring members are hetero atoms selected from the group consisting of O, N, and S, the remaining ring atoms being C. For purposes of exemplification, which should not be construed as limiting the scope of this invention, the following are examples of heterocycloalkyl: aziridinyl, azirinyl, oxiranyl, thiiranyl, thiirenyl, dioxiranyl, diazirinyl, diazepanyl, 1,3-dioxanyl, 1,3-dioxolanyl, 1,3-dithiolanyl, 1,3-dithianyl, imidazolidinyl, isothiazolinyl, isothiazolidinyl, isoxazolinyl, isoxazolidinyl, azetyl, oxetanyl, oxetyl, thietanyl, thietyl, diazetidinyl, dioxetanyl, dioxetenyl, dithietanyl, dithietyl, dioxalanyl, oxazolyl, thiazolyl, triazinyl, isothiazolyl, isoxazolyl, azepines, azetidinyl, morpholinyl, oxadiazolinyl, oxadiazolidinyl, oxazolinyl, oxazolidinyl, oxopiperidinyl, oxopyrrolidinyl, piperazinyl, piperidinyl, pyranyl, pyrazolinyl, pyrazolidinyl, pyrrolinyl, pyrrolidinyl, quinuclidinyl, thiomorpholinyl, tetrahydropyranyl, tetrahydrofuranyl, tetrahydrothienyl, thiadiazolinyl, thiadiazolidinyl, thiazolinyl, thiazolidinyl, thiomorpholinyl, 1,1-dioxidothiomorpholinyl (thiomorpholine sulfone), thiopyranyl, and trithianyl. A heterocycloalkyl group is optionally substituted by one or more substituents as described below.


As used herein, the terms “heterocyclylene” and “heterocycloalkylene” refers to a bivalent heterocyclyl (heterocycloalkyl) group, i.e., a cyclic alkylene group, having from 3-10 members and from 1-4 hetero atoms selected from S, O, and N. An example is piperidine-2,3-dicarboxylic acid, i.e., in that compound, the piperidine ring is a heterocyclylene group.


The term “heteroatom” is art-recognized, and includes an atom of any element other than carbon or hydrogen. Illustrative heteroatoms include boron, nitrogen, oxygen, phosphorus, sulfur and selenium, and alternatively oxygen, nitrogen or sulfur.


The term “heterocycloalkylalkyl” as used herein refers to an alkyl group substituted with one or more heterocycloalkyl (i.e., heterocyclyl) groups.


The term “alkenyl” as used herein means a straight or branched chain hydrocarbon radical containing from 2 to 10 carbons and containing at least one carbon-carbon double bond formed by the removal of two hydrogens. Preferably, alkenyl contains from 2 to 6 carbons. Representative examples of alkenyl include, but are not limited to, ethenyl, 2-propenyl, 2-methyl-2-propenyl, 3-butenyl, 4-pentenyl, and 5-hexenyl. The unsaturated bond(s) of the alkenyl group can be located anywhere in the moiety and can have either the (Z) or the (E) configuration about the double bond(s). The molecules differing only in their configuration about the double bond are called geometrical isomers.


The term “alkynyl” as used herein means a straight or branched chain hydrocarbon radical containing from 2 to 10 carbon atoms and containing at least one carbon-carbon triple bond. “C3-C6 alkynyl” means alkynyl containing from 3 to 6 carbon atoms. Representative examples of alkynyl include, but are not limited, to acetylenyl (i.e., ethynyl), 1-propynyl, 2-propynyl, 3-butynyl, 2-pentynyl, and 1-butynyl. This group can form a portion of another substituent, such as C3-C6 alkynyloxy-C1-C6 alkyl.


The term “alkylene” is art-recognized, and as used herein pertains to a diradical obtained by removing two hydrogen atoms of an alkyl group, as defined above. In one embodiment an alkylene refers to a disubstituted alkane, i.e., an alkane substituted at two positions with substituents such as halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, carboxamido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or heteroaromatic moieties, fluoroalkyl (such as trifluromethyl), cyano, or the like. That is, in one embodiment, a “substituted alkyl” is an “alkylene”.


The term “amino” is a term of art and as used herein refers to both unsubstituted and substituted amines, e.g., a moiety that may be represented by the general formulas:




embedded image


wherein Ra, Rb, and Rc each independently represent a hydrogen, an alkyl, an alkenyl, —(CH2)x—Rd, or Ra and Rb, taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure; Rd represents an aryl, a cycloalkyl, a cycloalkenyl, a heterocyclyl or a polycyclyl; and x is zero or an integer in the range of 1 to 8. In certain embodiments, only one of Ra or Rb may be a carbonyl, e.g., Ra, Rb, and the nitrogen together do not form an imide. In other embodiments, Ra and Rb (and optionally Rc) each independently represent a hydrogen, an alkyl, an alkenyl, or —(CH2)x—Rd. In certain embodiments, the term “amino” refers to —NH2.


The term “aminocarbonyl” refers to H2NC(═O)—, and one or both hydrogen atoms attached to the N atom can be substituted with the substituents Ra, or Ra and Rb, as defined above. Additionally, Ra can also represent C3-C6 alkynyl, hydroxyl, or (biotynyl-PEGn), wherein n ranges from 9 to 15.


The term “carboxamido”, as used herein, means —NHC(═O)—, wherein the carboxamido group is bound to the parent molecular moiety through the nitrogen. Examples of carboxamido include alkylcarboxamido such as CH3C(═O)N(H)— and CH3CH2C(═O)N(H)—.


The term “acyl” is a term of art and as used herein refers to any group or radical of the form RCO— where R is any organic group, e.g., alkyl, aryl, heteroaryl, aralkyl, and heteroaralkyl. Representative acyl groups include acetyl, benzoyl, and malonyl.


The term “N2-acetylhydrazinecarbonyl” as used herein refers to a group of the formula: Ac-NH—NH—C(═O)— that occurs, e.g., in the molecular formula of Example 23 below, where Ac represents acetyl.


The term “aminoalkyl” as used herein refers to an alkyl group substituted with one or more one amino groups. In one embodiment, the term “aminoalkyl” refers to an aminomethyl group.


The term “aminoacyl” is a term of art and as used herein refers to an acyl group substituted with one or more amino groups.


The term “aminothionyl” as used herein refers to an analog of an aminoacyl in which the O of RC(═O)— has been replaced by sulfur, hence is of the form RC(═S)—.


The term “azide” or “azido”, as used herein, means an —N3 group.


The term “carbonyl” as used herein refers to —C(═O)—.


The term “thiocarbonyl” as used herein refers to —C(═S)—.


The term “alkylthio” as used herein refers to alkyl-S—. Representative examples of C1-C6 alkylthio include methylthio, ethylthio, n-propylthio, and tert-butylthio. Representative examples of C1-C3 alkylthio include methylthio, ethylthio, and n-propylthio.


The term “alkylsulfinyl” as used herein refers to alkyl-S(═O)—. Here, the sulfur atom may have three different substituents and a lone pair of electrons, thus making sulfur a center of chirality and giving rise to a pair of optically stable epimers. Representative examples of C1-C3 alkylsulfinyl include methylsulfinyl, ethylsulfinyl, and n-propylsulfinyl, in the form of either one or both of their epimers.


The term “alkylsulfonyl” as used herein refers to alkyl-S(═O)2—. Representative examples of C1-C3 alkylsulfonyl include methylsulfonyl (also termed methanesulfonyl), ethylsulfonyl, and n-propylsulfonyl.


The term “mercaptoalkyl” as used herein refers to an alkyl group substituted by an —SH moiety. Representative examples of C1-C6 mercaptoalkyl include mercaptomethyl, mercaptoethyl, and mercapto-n-propyl.


The term “carboxy” as used herein refers to a —C(═O)—O— moiety, forming a portion of another substituents, such as alkylaminocarboxymethyl, or a portion of carboxylic acid esters. There is an exception to this convention in the case of functionalized cellulose derivatives, such as carboxymethyl cellulose, wherein “carboxymethyl” denotes HO2C—CH2—.


The terms “carboxyl”, and “hydroxycarbonyl”, as used herein, refer to a —CO2H group. This group can form a portion of another substituent, such as hydroxycarbonylmethyl, i.e., HO2C—CH2—.


The term “aryl” is a term of art and as used herein refers to include monocyclic, bicyclic, and polycyclic aromatic hydrocarbon groups, for example, benzene, naphthalene, 1,2,3,4-tetrahydronaphthalene, indene, 2,3-dihydroindene, and pyrene. The aromatic ring may be substituted at one or more ring positions with one or more substituents, such as halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, (cycloalkyl)alkoxyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, carboxamido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, aminosulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, heterocyclylalkyl, aromatic or heteroaromatic moieties, aminoalkyl, haloalkyl, fluoroalkyl (such as trifluoromethyl), haloalkoxyl, cyano, or the like. The term “aryl” also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are “fused rings”) wherein at least one of the rings is an aromatic hydrocarbon, e.g., the other cyclic rings may be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocycloalkyls. Representative examples of the polycyclic aryl ring systems include, but are not limited to, azulenyl, naphthyl, dihydroinden-1-yl, dihydroinden-2-yl, dihydroinden-3-yl, dihydroinden-4-yl, 2,3-dihydroindol-4-yl, 2,3-dihydroindol-5-yl, 2,3-dihydroindol-6-yl, 2,3-dihydroindol-7-yl, inden-1-yl, inden-2-yl, inden-3-yl, inden-4-yl, dihydronaphthalen-2-yl, dihydronaphthalen-3-yl, dihydronaphthalen-4-yl, dihydronaphthalen-1-yl, 5,6,7,8-tetrahydronaphthalen-1-yl, 5,6,7,8-tetrahydronaphthalen-2-yl, 2,3-dihydrobenzofuran-4-yl, 2,3-dihydrobenzofuran-5-yl, 2,3-dihydrobenzofuran-6-yl, 2,3-dihydrobenzofuran-7-yl, benzo[d][1,3]dioxol-4-yl, benzo[d][1,3]dioxol-5-yl, 2H-chromen-2-on-5-yl, 2H-chromen-2-on-6-yl, 2H-chromen-2-on-7-yl, 2H-chromen-2-on-8-yl, isoindoline-1,3-dion-4-yl, isoindoline-1,3-dion-5-yl, inden-1-on-4-yl, inden-1-on-5-yl, inden-1-on-6-yl, inden-1-on-6-yl, 2,3-dihydrobenzo[b][1,4]dioxan-5-yl, 2,3-dihydrobenzo[b][1,4]dioxan-6-yl, 2H-benzo[b][1,4]oxazin3(4H)-on-5-yl, 2H-benzo[b][1,4]oxazin3(4H)-on-6-yl, 2H-benzo[b][1,4]oxazin3(4H)-on-7-yl, 2H-benzo[b][1,4]oxazin3(4H)-on-8-yl, benzo[d]oxazin-2(3H)-on-5-yl, benzo[d]oxazin-2(3H)-on-6-yl, benzo[d]oxazin-2(3H)-on-7-yl, benzo[d]oxazin-2(3H)-on-8-yl, quinazolin-4(3H)-on-5-yl, quinazolin-4(3H)-on-6-yl, quinazolin-4(3H)-on-7-yl, quinazolin-4(3H)-on-8-yl, quinoxalin-2(1H)-on-5-yl, quinoxalin-2(1H)-on-6-yl, quinoxalin-2(1H)-on-7-yl, quinoxalin-2(1H)-on-8-yl, benzo[d]thiazol-2(3H)-on-4-yl, benzo[d]thiazol-2(3H)-on-5-yl, benzo[d]thiazol-2(3H)-on-6-yl, and, benzo[d]thiazol-2(3H)-on-7-yl. In certain embodiments, the bicyclic aryl is (i) naphthyl, or (ii) a phenyl ring fused to either a 5 or 6 membered monocyclic cycloalkyl, a 5 or 6 membered monocyclic cycloalkenyl, or a 5 or 6 membered monocyclic heterocyclyl, wherein the fused cycloalkyl, cycloalkenyl, and heterocyclyl groups are optionally substituted. In certain embodiments, the term “aryl” refers to C6-C10 aryl. In certain embodiments, the term “aryl” refers to a phenyl group or a naphthyl group.


The term “heteroaryl” is a term of art and as used herein refers to a monocyclic or bicyclic aromatic group having 3 to 14, 5 to 14, 3 to 12, or 3 to 10 total atoms (other than hydrogen) including one or more heteroatoms such as nitrogen, oxygen, or sulfur in the ring structure. More preferred heteroaryl groups have from 5-10 ring members where from 1-4 of the ring members are heteroatoms selected from the group consisting of O, N, and S. Exemplary heteroaryl groups include, for example, azaindolyl, benzo(b)thienyl, benzimidazolyl, benzofuranyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, benzotriazolyl, benzoxadiazolyl, furanyl, imidazolyl, imidazopyridinyl, indolyl, indolinyl, indazolyl, isoindolinyl, isoxazolyl, isothiazolyl, isoquinolinyl, oxadiazolyl, oxazolyl, purinyl, pyranyl, pyrazinyl, pyrazolyl, pyridinyl, pyrimidinyl, pyrrolyl, pyrrolo[2,3-d]pyrimidinyl, pyrazolo[3,4-d]pyrimidinyl, quinolinyl, quinazolinyl, triazolyl, thiazolyl, thiophenyl, tetrahydroindolyl, tetrazolyl, thiadiazolyl, thienyl, thiomorpholinyl, triazolyl or tropanyl, and the like.


Exemplary “5- or 6-membered monocyclic heteroaryl” groups include, for example, pyrrolyl, furyl, thiophenyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, dithiazolyl, oxadiazolyl, tetrazolyl, pyridyl, pyranyl, diazinyl, oxazinyl, thiazinyl, triazinyl, tetrazinyl, where the pattern of heteroatoms within the heteroaryl ring is any allowable (such as in 1,2,3-triazine, 1,2,4-triazine, or 1,3,5-triazine), and the attachment point of the heteroaryl group to the rest of the molecule is at any allowable carbon atom being the member of said heteroaryl group (such as in 2-thiophenyl or 3-thiophenyl).


Any heteroaryl can be optionally substituted at one or more ring positions with one or more substituents such as halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, carboxamido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or heteroaromatic moieties, fluoroalkyl (such as trifluromethyl), cyano, or the like. The term “heteroaryl” also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are “fused rings”) wherein at least one of the rings is an aromatic group having one or more heteroatoms in the ring structure, e.g., the other cyclic rings may be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls. Representative examples of bicyclic heteroaryl include, but are not limited to, benzimidazolyl, benzofuranyl, benzothienyl, benzoxadiazolyl, benzoxathiadiazolyl, benzothiazolyl, cinnolinyl, 5,6-dihydroquinolin-2-yl, 5,6-dihydroisoquinolin-1-yl, furopyridinyl, indazolyl, indolyl, isoquinolinyl, naphthyridinyl, quinolinyl, purinyl, 5,6,7,8-tetrahydroquinolin-2-yl, 5,6,7,8-tetrahydroquinolin-3-yl, 5,6,7,8-tetrahydroquinolin-4-yl, 5,6,7,8-tetrahydroisoquinolin-1-yl, thienopyridinyl, 4,5,6,7-tetrahydro-benzo[c][1,2,5]oxadiazolyl, and 6,7-dihydrobenzo[c][1,2,5]oxadiazol-4(5H)-onyl. Any heteroaryl or bicyclic heteroaryl can be optionally substituted as detailed below.


The term “aralkyl”, or “arylalkyl”, for example “aryl-C1-C6 alkyl”, is a term of art and as used herein refers to an alkyl group, for example a C1-C6 alkyl group, substituted with an aryl group, wherein the moiety is appended to the parent molecule through the alkyl group. An example of aralkyl is the benzyl group, i.e., the phenyl-methyl-group.


The term “arylene” is art-recognized, and as used herein pertains to a diradical obtained by removing two hydrogen atoms of an aryl group, as defined above. An exemplary arylene group is 1,4-phenylene. Another exemplary arylene group is benzo, or 1,2-phenylene, of the following structural formula:




embedded image


The term “heteroaralkyl”, “heteroarylalkyl”, for example, “heteroaryl-C1-C6 alkyl” is a term of art and as used herein refers to an alkyl group, for example a C1-C6 alkyl group, substituted with a heteroaryl group, appended to the parent molecular moiety through the alkyl group.


The term “alkoxy” or “alkoxyl” as used herein means an alkyl group, as defined herein, appended to the parent molecular moiety through an oxygen atom. Preferably, the alkoxy group is C1-C6 alkoxy. Representative examples include, but are not limited to, methoxy, ethoxy, propoxy, 2-propoxy, butoxy, tert-butoxy, pentyloxy, and hexyloxy. Representative examples of C1-C3 alkoxy include methoxy, ethoxy, and propoxy.


The term “alkoxycarbonyl” means an alkoxy group, as defined herein, appended to the parent molecular moiety through a carbonyl group, represented by —C(═O)—, as defined herein. Representative examples of alkoxycarbonyl include, but are not limited to, methoxycarbonyl, ethoxycarbonyl, and tert-butoxycarbonyl. Alkoxycarbonyl can form a portion of another moiety, e.g., methoxycarbonylmethyl.


The term “alkylcarbonyl”, as used herein, means an alkyl group, as defined herein, appended to the parent molecular moiety through a carbonyl group, as defined herein. Representative examples of alkylcarbonyl include, but are not limited to, acetyl, 1-oxopropyl, 2,2-dimethyl-1-oxopropyl, 1-oxobutyl, and 1-oxopentyl.


The term “arylcarbonyl”, as used herein, means an aryl group, as defined herein, appended to the parent molecular moiety through a carbonyl group, as defined herein. Representative examples of arylcarbonyl include, but are not limited to, benzoyl and (2-naphthyl)carbonyl.


The term “alkylcarbonyloxy” and “arylcarbonyloxy”, as used herein, means an alkylcarbonyl or arylcarbonyl group, as defined herein, appended to the parent molecular moiety through an oxygen atom. Representative examples of alkylcarbonyloxy include, but are not limited to, acetyloxy, ethylcarbonyloxy, and tert-butylcarbonyloxy. Representative examples of arylcarbonyloxy include, but are not limited to phenylcarbonyloxy.


The term “alkenoxy” or “alkenoxyl” means an alkenyl group, as defined herein, appended to the parent molecular moiety through an oxygen atom. Representative examples of alkenoxy include, but are not limited to, 2-propen-1-oxy (i.e., CH2═CH—CH2—O—) and vinyloxy (i.e., CH2═CH—O—).


The term “aryloxy” as used herein means an aryl group, as defined herein, appended to the parent molecular moiety through an oxygen atom.


The term “heteroaryloxy” as used herein means a heteroaryl group, as defined herein, appended to the parent molecular moiety through an oxygen atom.


The terms “cyano” and “nitrile” are terms of art and as used herein refers to —CN.


The term “nitro”, as used herein, means —NO2.


The term “halo” or “halogen” is a term of art and as used herein refers to —F, —Cl, —Br, or —I.


The term “haloalkyl” as used herein refers to an alkyl group, as defined herein, wherein one or more or all of the hydrogens are replaced with halogen atoms. The term “haloalkoxy” refers to an alkoxy group, as defined herein, wherein one or more or all of the hydrogens are replaced with halogen atoms. An exemplary C1-C6 haloalkyl group is trifluoromethyl.


The term “hydroxy” or “hydroxyl” is a term of art and as used herein refers to —OH.


The term “hydroxyalkyl”, as used herein, means at least one hydroxy group, as defined herein, is appended to the parent molecular moiety through an alkyl group, as defined herein. Representative examples of C1-C6 hydroxyalkyl include, but are not limited to, hydroxymethyl, 2-hydroxyethyl, 3-hydroxypropyl, and 2,3-dihydroxypentyl.


The term “sulfonyl”, as used herein, refers to the group —S(═O)2— that may form a portion of larger moieties, such as methanesulfonyl or p-toluenesulfonyl.


The term “silyl”, as used herein, includes hydrocarbyl derivatives of the silyl (H3Si—) group (i.e., (hydrocarbyl)3Si—), wherein a hydrocarbon radicals are univalent groups formed by removing a hydrogen atom from a hydrocarbon, e.g., ethyl, phenyl. The hydrocarbon radicals can be combinations of differing groups which can be varied in order to provide a number of silyl groups, such as trimethylsilyl (TMS), tert-butyldiphenylsilyl (TBDPS), tert-butyldimethylsilyl (TBS/TBDMS), triisopropylsilyl (TIPS), and [2-(trimethylsilyl)ethoxy]methyl (SEM).


The term “silyloxy”, as used herein, means a silyl group, as defined herein, is appended to the parent molecule through an oxygen atom.


The term “PEG”, is a term of art and, as used herein, refers to a poly(ethylene glycol) group. The number appearing after PEG denotes the number of repeating ethylene glycol units. PEG may appear as a part of a substituent attached to the molecule in question. For example, a full structural formula of the biotin-attaching substituent, such as:




embedded image


may be drawn as:




embedded image


or even written as biotinyl-PEG11-for brevity.


The term “asialofetuin”, as used herein, means a glycoprotein with three asparagine-linked triantennary complex carbohydrate chains and terminal N-acetylgalactosamine residues. Asialofetuin is known to bind selectively to ASGP-R (asialoglycoprotein receptor) and some galectins.


The term “small molecule protein degrader”, as used herein, means a bifunctional molecule comprised of three components: i) a ligand for a target protein of interest, ii) a ligand that binds to protein involved in protein degradation system (i.e., an E3 ubiquitin ligase ligand or ligand of asialoglycoprotein receptors), and iii) a linker. Formation of a complex between the target protein and the protein involved in proteasome machinery, aided by a small molecule degrader, induces degradation of the target protein and down regulates its level.


Certain compounds contained in compositions of the present invention may exist in particular geometric or stereoisomeric forms. In addition, compounds of the present invention may also be optically active. The present invention contemplates all such compounds, including cis- and trans-isomers, (R)- and (S)-enantiomers, diastereomers, (D)-isomers, (L)-isomers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention. Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are intended to be included in this invention.


The term “racemic mixture” refers to a mixture containing equal proportions of the first enantiomer of the molecule and of the second enantiomer of this molecule, wherein the second enantiomer is the mirror image of the first one.


The term “scalemic mixture” refers to any non-racemic mixture of stereoisomers of the molecule.


If, for instance, a particular enantiomer of compound of the present invention is desired, it may be prepared by asymmetric synthesis, or by derivation with a chiral auxiliary, where the resulting diastereomeric mixture is separated and the auxiliary group cleaved to provide the pure desired enantiomers. Alternatively, where the molecule contains a basic functional group, such as amino, or an acidic functional group, such as carboxyl, diastereomeric salts are formed with an appropriate optically-active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic means well known in the art, and subsequent recovery of the pure enantiomers.


Organic compounds frequently occur in more than one crystalline form, that can differ in their physical and biological properties, such as melting point, stability, solubility, bioavailability. Such crystalline forms are termed polymorphs. All polymorphs of the inventive compounds of formula (I) and of their salts are intended to be within the scope of this invention.


Since many chemical elements can occur as isotopes, their abundance in the molecule of the inventive compound of formula (I) may be identical as in the nature or altered. Some isotopes exhibit different spectral or biological properties, and this phenomenon may be used for analysis of distribution and metabolism of drugs in the body of the recipient. All forms of the compounds of formula (I), both having a natural or unnatural abundance of isotopes of any of their constituent elements are intended to be within the scope of this invention.


It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, fragmentation, decomposition, cyclization, elimination, or other reaction.


The term “substituted” is also contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds. Illustrative substituents include, for example, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, (cycloalkyl)alkoxyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, carboxamido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, aminosulfonyl, sulfonamido, ketone, aldehyde, ester, heterocycloalkyl, heterocycloalkylalkyl, (heterocycloalkyl)alkoxyl, aromatic or heteroaromatic moieties, aminoalkyl, haloalkyl, fluoroalkyl (such as trifluoromethyl), haloalkoxyl, cyano, or other substituents described above. The permissible substituents may be one or more and the same or different for appropriate organic compounds. For purposes of this invention, the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. This invention is not intended to be limited in any manner by the permissible substituents of organic compounds.


The phrase “protecting group,” as used herein, means temporary substituents which protect a potentially reactive functional group from undesired chemical transformations. Examples of such protecting groups include esters of carboxylic acids, silyl ethers of alcohols, and acetals and ketals of aldehydes and ketones, respectively. The field of protecting group chemistry has been reviewed (Greene, T. W.; Wuts, P. G. M. Protective Groups in Organic Synthesis, 2nd ed.; Wiley: New York, 1991). Protected forms of the inventive compounds are included within the scope of this invention.


A “saturated” or “fully saturated” compound means that the referenced chemical structure does not contain any multiple carbon-carbon bonds. For example, a saturated cycloalkyl group as defined herein includes cyclohexyl, cyclopropyl, and the like.


A “unsaturated” or “partially saturated” compound means that the referenced chemical structure may contain one or more multiple carbon-carbon bonds, but is not aromatic. For example, a unsaturated cycloalkyl group as defined herein includes cyclohexenyl, cyclopentenyl, cyclohexadienyl, and the like.


For purposes of the invention, the chemical elements are identified in accordance with the Periodic Chart of the Elements, IUPAC version, The Merck Index, Twelfth Edition, 1996, inside cover.


Other chemistry terms herein are used according to conventional usage in the art, as exemplified by The McGraw-Hill Dictionary of Chemical Terms (ed. Parker, S., 1985), McGraw-Hill, San Francisco). Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention pertains.


It will be apparent to one skilled in the art that certain compounds of this disclosure may exist in tautomeric forms, all such tautomeric forms of the compounds being within the scope of the disclosure.


Unless otherwise stated, structures depicted herein include all stereochemical configurations consistent with the depicted structure. For example, (i) a structure with a single stereocenter encompasses both the R and S configurations at the stereocenter and mixtures thereof, including racemic mixtures, and (ii) in a structure with two or more stereocenters, any wedged and dashed bonds show relative stereochemistry unless otherwise noted, such that the structure encompasses the individual enantiomers of the depicted compound and mixtures thereof, including racemic mixtures. Therefore, single stereochemical isomers as well as enantiomeric and diastereomeric mixtures of the present compounds are within the scope of the disclosure. Both the R and the S stereochemical isomers, as well as all mixtures thereof, are included within the scope of the disclosure. Similarly, indications of stereochemistry in chemical structures with two or more chiral centers convey relative stereochemistry unless otherwise defined.


The chemical structures of examples that are a mixture of diastereoisomers or a single diastereoisomer but with unknown relative configuration are drawn and named without specifying defined stereochemical configuration.


The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.


The term “pharmaceutically acceptable salt” as used herein includes salts derived from inorganic or organic acids including, for example, hydrochloric, hydrobromic, sulfuric, nitric, perchloric, phosphoric, formic, acetic, lactic, maleic, fumaric, succinic, tartaric, glycolic, salicylic, citric, methanesulfonic, benzenesulfonic, benzoic, malonic, trifluoroacetic, trichloroacetic, naphthalene-2-sulfonic, and other acids. Pharmaceutically acceptable salt forms can include forms wherein the ratio of molecules comprising the salt is not 1:1. For example, the salt may comprise more than one inorganic or organic acid molecule per molecule of base, such as two hydrochloric acid molecules per molecule of compound of Formula I. As another example, the salt may comprise less than one inorganic or organic acid molecule per molecule of base, such as two molecules of compound of Formula I per molecule of tartaric acid.


As used herein, a protic solvent is a solvent that has a hydrogen atom bound to an oxygen (as in a hydroxyl group) or a nitrogen (as in an amine group). In general terms, any solvent that contains labile H+ is called a protic solvent. The molecules of such solvents readily donate protons (H+) to reagents. In contrast, an aprotic solvent is a solvent that does not have a hydrogen atom bound to an oxygen (as in a hydroxyl group) or a nitrogen (as in an amine group), and it cannot donate hydrogen.


As used herein, a polar protic solvent is a protic solvent that will dissolve many salts. In general, these solvents have high dielectric constants and high polarity. Non-limiting examples of polar protic solvents include acetic acid, ammonia, ethanol, formic acid, isopropanol, methanol, n-butanol, nitromethane, n-propanol, tert-butanol, and water.


As used herein, a polar aprotic solvent is a solvent that will dissolve many salts, but lacks an acidic hydrogen; these solvents generally have intermediate to high dielectric constants and polarity. Non-limiting examples of polar aprotic solvents include acetone, acetonitrile, dichloromethane (DCM), dimethyl sulfoxide (DMSO), ethyl acetate, hexamethylphosphoric triamide (HMPT), N,N-dimethylformamide (DMF), and tetrahydrofuran (THF).


As used herein, a nonpolar aprotic solvent is a solvent that will dissolve many salts, but lacks an acidic hydrogen; these solvents generally have low dielectric constants and polarity. Non-limiting examples of nonpolar aprotic solvents include benzene, chloroform, cyclohexane, diethyl ether, hexane, pentane, and toluene.


Many organic compounds form stable crystals that incorporate solvent molecules into the crystalline structure, without chemical alteration of the organic molecule. Such solvated compounds are called solvates in general, and they are called hydrates when the solvent is water.


A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the therapeutically effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the pharmaceutical composition or compound at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. By “therapeutically effective amount” is meant the concentration of a compound that is sufficient to elicit the desired therapeutic effect. It is generally understood that the effective amount of the compound will vary according to the weight, sex, age, and medical history of the subject. Other factors which influence the effective amount may include, but are not limited to, the severity of the patient's condition, the disorder being treated, the stability of the compound, the mode of administration, the bioavailability of the particular compound, and, if desired, another type of therapeutic agent being administered with the compound of the invention. A larger total dose can be delivered by multiple administrations of the agent. Methods to determine efficacy and dosage are known to those skilled in the art (Isselbacher et al. (1996) Harrison's Principles of Internal Medicine 13 ed., 1814-1882, herein incorporated by reference).


“Modulating” or “modulate” refers to the treating, prevention, suppression, enhancement or induction of a function, condition or disorder.


The term “treating” includes prophylactic and/or therapeutic treatments. The term “prophylactic or therapeutic” treatment is art-recognized and includes administration to the host of one or more of the subject compositions. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic (i.e., it protects the host against developing the unwanted condition), whereas if it is administered after manifestation of the unwanted condition, the treatment is therapeutic, (i.e., it is intended to diminish, ameliorate, or stabilize the existing unwanted condition or side effects thereof). In certain embodiments, the methods of the invention are for therapeutically treating.


As used herein, “subject” refers to a warm blooded animal such as a mammal, preferably a human, or a human child, which is afflicted with, or has the potential to be afflicted with one or more diseases and disorders described herein.


“EC50” refers to a dosage, concentration or amount of a particular test compound that elicits a dose-dependent response at 50% of maximal expression of a particular response that is induced, provoked or potentiated by the particular test compound.


“IC50” refers to an amount, concentration or dosage of a particular test compound that achieves a 50% inhibition of a maximal response in an assay that measures such response.


“Kd”, expressed in molar units (M) refers to the concentration of analyzed compound/probe at which 50% of ligand binding sites of protein are occupied at equilibrium, or the concentration at which the number of ligand molecules bound to target protein equals the number of ligand molecules that is not bound.


Compounds of the Invention


In one aspect, this invention concerns a novel compound represented by structural formula (I),




embedded image


wherein:

    • W represents —C(—R1f)(—R1g)—;
    • X represents —C(—R1h)(—R1j)—;
    • Y represents —C(—R1k)(—R1m)—, —N(—R1k)—, or —O—;
    • R1b represents —H;
    • R1c represents —H;
    • R1d represents 4-R10-benzyl;
    • R1e represents —H;
    • R1f represents —H;
    • R1g represents —H;
    • R1h is selected from the group consisting of C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, hydroxy-C1-C6 alkyl, azido-C1-C6 alkyl, hydroxycarbonyl-C1-C3 alkoxy-C1-C6 alkyl, C3-C6 alkynyloxy-C1-C6 alkyl, halo-C1-C6 alkyl, C1-C3 alkylthio-C1-C6 alkyl, C1-C3 alkylsulfinyl-C1-C6 alkyl, C1-C3 alkylsulfonyl-C1-C6 alkyl, hydroxycarbonyl-C1-C3 alkylsulfonyl-C1-C6 alkyl, hydroxycarbonyl-C1-C6 alkyl, C1-C3 alkylcarbonyl-C1-C6 alkyl, C1-C3 alkylaminocarbonyl-C1-C6 alkyl, di(C1-C3 alkyl)aminocarbonyl-C1-C6 alkyl, C1-C3 alkylsulfonylamino-C1-C6 alkyl, halo-C1-C3 alkylsulfonylamino-C1-C6 alkyl, 5- or 6-membered monocyclic heteroaryl-carbonylamino-C1-C6 alkyl, 5- or 6-membered monocyclic heteroaryl-C1-C3 alkoxy-C1-C6 alkyl, 5- or 6-membered monocyclic heteroaryl-C1-C6 alkyl, hydroxycarbonyl, C1-C3 alkoxycarbonyl, aminocarbonyl, C1-C3 alkylaminocarbonyl, di(C1-C3 alkyl)aminocarbonyl, C3-C6 alkynylaminocarbonyl, hydroxycarbonyl-C1-C3 alkyl(C1-C3 alkyl)aminocarbonyl, (biotinyl-PEG11)aminocarbonyl, (biotinyl-PEG11)aminocarbonyl-C1-C3 alkylsulfonyl-C1-C6 alkyl, hydroxylaminocarbonyl, N2-acetylhydrazinecarbonyl, (5- or 6-membered monocyclic heteroaryl)-C1-C3 alkylaminocarbonyl, (5- or 6-membered monocyclic heteroaryl)aminocarbonyl, C1-C3 alkylsulfonyl, 5- or 6-membered monocyclic heteroaryl, (3-azabicyclo[3.1.0]hexan-3-yl)-C1-C3 alkyl, (4-, 5-, or 6-membered heterocyclyl)amino-C1-C3 alkyl, hydroxycarbonyl-C1-C6 alkyl-(5- or 6-membered monocyclic heteroaryl), R20, and 5-(2-(2,6-dioxopiperidin-3-yl)-3-oxoisoindolin-4-yl)pentyl, where any available position of any heteroaryl ring in R1h can be substituted with a substituent selected from the group consisting of C1-C6 alkyl, C3-C6 alkynyl, halo, trifluoromethyl, 2,2,2-trifluoroethyl, biotinyl-PEG11, and any available position of any carbon chain in R1h excluding heteroaryl rings can be substituted with a substituent selected from halogen, amino, hydroxy, and oxo;
    • R1j represents —H;
    • R1k represents —H;
    • R1m represents —H;
    • or R1k and R1h taken along with nitrogen and carbon atoms bearing them, respectively, represent a 6-membered heterocyclic ring of structural formula (II):




embedded image




    • R3a represents —H;

    • R3b represents —H;

    • R3c represents —H;

    • R3d represents —H;

    • R3e represents —H;

    • R3f is selected from the group consisting of 1-methyl-1H-imidazol-4-yl, 1,2-dimethyl-1H-imidazol-4-yl, 5-methyloxazol-2-yl, 4,5-dimethyloxazol-2-yl, 1H-pyrazol-1-yl, 1-methyl-1H-pyrazol-3-yl, 4-methyl-1H-pyrazol-1-yl, 4-fluoro-1H-pyrazol-1-yl, 1-isopropyl-1H-pyrazol-3-yl, 5-methyl-1H-pyrazol-3-yl, 1,5-dimethyl-1H-pyrazol-3-yl, 1-ethyl-5-methyl-1H-pyrazol-3-yl, 1,5-dimethyl-1H-pyrazol-3-yl, 1,5-dimethyl-1H-pyrazol-4-yl, 1-methyl-5-(trifluoromethyl)-1H-pyrazol-3-yl, 1-methyl-2-cyano-1H-pyrrol-4-yl, 1H-tetrazol-5-yl, 2-methyl-2H-tetrazol-5-yl, 5-methyl-1,3,4-thiadiazol-2-yl, 5-methylthiazol-2-yl, 4,5-dimethylthiophen-2-yl, 1H-1,2,4-triazol-1-yl, 1-methyl-1H-1,2,3-triazol-4-yl, 1-methyl-1H-1,2,4-triazol-3-yl, 2-methyl-2H-1,2,3-triazol-4-yl, 1,5-dimethyl-1H-1,2,4-triazol-3-yl, 5-hydroxy-1-methyl-1H-1,2,4-triazol-3-yl, and 5-methoxy-1-methyl-1H-1,2,4-triazol-3-yl;

    • R3g represents —H;

    • R3h represents —H;

    • R3j represents —H;

    • R3k represents —H;

    • R3m represents —H;

    • or R3d and R3g taken along with the single bond linking two carbon atoms bearing them represent a double bond between these two carbon atoms;

    • R4b represents —H;

    • R4c represents —H;

    • R4f represents —H;

    • R4g represents —H;

    • R4h represents —H;

    • R4j represents —H;

    • R10 represents halogen or trifluoromethyl;

    • and R20 represents







embedded image




    • or a tautomer, stereoisomer, N-oxide, methiodide, pharmaceutically acceptable salt, solvate, or polymorph thereof.





The R20 substituent is an asialoglycoprotein receptor ligand containing a linker with amide and triazole groups.


Preferably, this invention concerns a novel compound of structural formula (I),




embedded image


wherein:

    • W represents —C(—R1f)(—R1g)—;
    • X represents —C(—R1h)(—R1j)—;
    • Y represents —C(—R1k)(—R1m)—, —N(—R1k)—, or —O—;
    • R1b represents —H;
    • R1c represents —H;
    • R1d represents 4-R10-benzyl;
    • R1e represents —H;
    • R1f represents —H;
    • R1g represents —H;
    • R1h is selected from the group consisting of C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, hydroxy-C1-C6 alkyl, azido-C1-C6 alkyl, hydroxycarbonyl-C1-C3 alkoxy-C1-C6 alkyl, C3-C6 alkynyloxy-C1-C6 alkyl, halo-C1-C6 alkyl, C1-C3 alkylthio-C1-C6 alkyl, C1-C3 alkylsulfinyl-C1-C6 alkyl, C1-C3 alkylsulfonyl-C1-C6 alkyl, hydroxycarbonyl-C1-C3 alkylsulfonyl-C1-C6 alkyl, hydroxycarbonyl-C1-C6 alkyl, C1-C3 alkylcarbonyl-C1-C6 alkyl, C1-C3 alkylaminocarbonyl-C1-C6 alkyl, di(C1-C3 alkyl)aminocarbonyl-C1-C6 alkyl, C1-C3 alkylsulfonylamino-C1-C6 alkyl, halo-C1-C3 alkylsulfonylamino-C1-C6 alkyl, 5- or 6-membered monocyclic heteroaryl-carbonylamino-C1-C6 alkyl, 5- or 6-membered monocyclic heteroaryl-C1-C3 alkoxy-C1-C6 alkyl, 5- or 6-membered monocyclic heteroaryl-C1-C6 alkyl, hydroxycarbonyl, C1-C3 alkoxycarbonyl, aminocarbonyl, C1-C3 alkylaminocarbonyl, di(C1-C3 alkyl)aminocarbonyl, C3-C6 alkynylaminocarbonyl, hydroxycarbonyl-C1-C3 alkyl(C1-C3 alkyl)aminocarbonyl, (biotinyl-PEG11)aminocarbonyl, hydroxylaminocarbonyl, N2-acetylhydrazinecarbonyl, (5- or 6-membered monocyclic heteroaryl)-C1-C3 alkylaminocarbonyl, (5- or 6-membered monocyclic heteroaryl)aminocarbonyl, C1-C3 alkylsulfonyl, 5- or 6-membered monocyclic heteroaryl, (3-azabicyclo[3.1.0]hexan-3-yl)-C1-C3 alkyl, (4-, 5-, or 6-membered heterocyclyl)amino-C1-C3 alkyl, and hydroxycarbonyl-C1-C6 alkyl-(5- or 6-membered monocyclic heteroaryl),
    • where any available position of any heteroaryl ring in R1h can be substituted with a substituent selected from the group consisting of C1-C6 alkyl, C3-C6 alkynyl, halo, trifluoromethyl, 2,2,2-trifluoroethyl, biotinyl-PEG11, and any available position of any carbon chain in R1h excluding heteroaryl rings can be substituted with a substituent selected from halogen, amino, hydroxy, and oxo;
    • R1j represents —H;
    • R1k represents —H;
    • R1m represents —H;
    • or R1k and R1h taken along with nitrogen and carbon atoms bearing them, respectively, represent a 6-membered heterocyclic ring of structural formula (II):




embedded image




    • R3a represents —H;

    • R3b represents —H;

    • R3c represents —H;

    • R3d represents —H;

    • R3e represents —H;

    • R3f is selected from the group consisting of 5-methyloxazol-2-yl, 1,5-dimethyl-1H-pyrazol-3-yl, 1,5-dimethyl-1H-1,2,4-triazol-3-yl, 4,5-dimethyloxazol-2-yl, 5-fluoro-1-methyl-1H-pyrazol-3-yl, 4,5-dimethylthiazol-2-yl, and 5-methylthiazol-2-yl;

    • R3g represents —H;

    • R3h represents —H;

    • R3j represents —H;

    • R3k represents —H;

    • R3m represents —H;

    • or R3d and R3g taken along with the single bond linking two carbon atoms bearing them represent a double bond between these two carbon atoms;

    • R4b represents —H;

    • R4c represents —H;

    • R4f represents —H;

    • R4g represents —H;

    • R4h represents —H;

    • R4j represents —H;

    • and R10 represents halogen or trifluoromethyl;

    • or a tautomer, stereoisomer, N-oxide, methiodide, pharmaceutically acceptable salt, solvate, or polymorph thereof.





The above-defined general formula (I) covers certain compounds of the invention, which can be described in more detail as follows.


In some embodiments, the compound of the invention is of structural formula (Ia)




embedded image


wherein:

    • Y represents —CH2—, —N(—R1k)—, or —O—;
    • R1h is selected from the group consisting of methyl, ethynyl, 2-hydroxyprop-2-yl, azidomethyl, hydroxymethyl, hydroxycarbonylmethoxymethyl, (prop-2-yn-1-yloxy)methyl, fluoromethyl, methylthiomethyl, methylsulfinylmethyl, methylsulfonylmethyl, ethylsulfonylmethyl, 2-(hydroxycarbonyl)ethylsulfonylmethyl, hydroxycarbonylmethyl, methoxycarbonylmethyl, ethylaminocarbonylmethyl, methylsulfonylaminomethyl, trifluoromethylsulfonylaminomethyl, (1H-pyrazol-3-yl)carbonylaminomethyl, 1-(trifluoromethyl)-1H-1,2,3-triazol-4-yl, (1-(biotinyl-PEG11)-1H-1,2,3-triazol-4-yl)methoxymethyl, (2H-1,2,3-triazol-4-yl)carbonylaminomethyl, (1H-1,2,3-triazol-5-yl)carbonylaminomethyl, (1-methyl-1H-imidazol-4-yl)carbonylaminomethyl, (1H-imidazol-4-yl)carbonylaminomethyl, (1,5-dimethyl-1H-pyrazol-3-yl)carbonylaminomethyl, (2,5-dioxopyrrolidin-1-yl)methyl, (2,4-dioxoimidazolidin-3-yl)methyl, (1-methyl-2,4-dioxoimidazolidin-3-yl)methyl, hydroxycarbonyl, methoxycarbonyl, aminocarbonyl, methylaminocarbonyl, dimethylaminocarbonyl, ethylaminocarbonyl, isopropylaminocarbonyl, (prop-2-yn-1-yl)aminocarbonyl, hydroxycarbonylmethyl(methyl)aminocarbonyl, (biotinyl-PEG11)aminocarbonyl, hydroxylaminocarbonyl, N2-acetylhydrazinecarbonyl, (1-(biotinyl-PEG11)-1H-1,2,3-triazol-4-yl)methylaminocarbonyl, (1-methyl-1H-imidazol-4-yl)aminocarbonyl, (1,5-dimethyl-1H-pyrazol-3-yl)aminocarbonyl, (2-(3-(but-3-yn-1-yl)-3H-diazirin-3-yl)ethyl)aminocarbonyl, methylsulfonyl, 3-methylisoxazol-5-yl, 1-(2,2,2-trifluoroethyl)-1H-1,2,3-triazol-4-yl, 5-methyl-1,3,4-oxadiazol-2-yl, (6,6-dimethyl-2,4-dioxo-3-azabicyclo[3.1.0]hexan-3-yl)methyl, (1,2-dioxo-3-hydroxy-cyclobut-3-en-4-yl)aminomethyl, (1,2-dioxo-3-amino-cyclobut-3-en-4-yl)aminomethyl, 1-(biotinyl-PEG11)-1H-1,2,3-triazol-4-yl, 1-(hydroxycarbonylmethyl)-1H-1,2,3-triazol-4-yl, 1-(2-(hydroxycarbonyl)ethyl)-1H-1,2,3-triazol-4-yl, 1-(3-(hydroxycarbonyl)prop-1-yl)-1H-1,2,3-triazol-4-yl, 1-(4-(hydroxycarbonyl)but-1-yl)-1H-1,2,3-triazol-4-yl, and 5-methyl-1H-pyrazol-3-yl;
    • or R1k and R1h taken along with nitrogen and carbon atoms bearing them, respectively, represent a 6-membered heterocyclic ring of structural formula (IIa):




embedded image




    • R3d represents H;

    • R3g represents H;

    • or R3d and R3g taken along with the single bond linking two carbon atoms bearing them represent a double bond between these two carbon atoms;

    • R3f is selected from the group consisting of 5-methyloxazol-2-yl, 1,5-dimethyl-1H-pyrazol-3-yl, 1,5-dimethyl-1H-1,2,4-triazol-3-yl, 4,5-dimethyloxazol-2-yl, 5-fluoro-1-methyl-1H-pyrazol-3-yl, 4,5-dimethylthiazol-2-yl, and 5-methylthiazol-2-yl;

    • and R10 represents chloro, bromo, or trifluoromethyl;

    • or a tautomer, stereoisomer, pharmaceutically acceptable salt, solvate, or polymorph thereof.





In a preferred embodiment, the compound according to the invention is as defined above, and:

    • X represents —CH(—R1h)—;
    • Y represents —CH2—; and
    • R1h represents methanesulfonyl.


In a more preferred embodiment, the compound according to the invention is as defined above, and:

    • X represents —CH(—R1h)—;
    • Y represents —N(—R1k)—.
    • and R1k and R1h taken along with nitrogen and carbon atoms bearing them, respectively, represent a 6-membered heterocyclic ring of structural formula (IIb):




embedded image


In another embodiment, the compound according to the invention is represented by structural formula (I) or (Ia), and Y represents —O—.


More preferably, the compound of the invention is as defined above, and R1d and R1h are in cis relationship.


Preferably, the compound of the invention is as defined above, and the absolute stereochemistry at the carbon atom bearing R1d is S.


In some embodiments, the compound of the invention is of structural formula (Ib)




embedded image


wherein:

    • R1h is selected from the group consisting of methyl, ethynyl, 2-hydroxyprop-2-yl, azidomethyl, hydroxymethyl, hydroxycarbonylmethoxymethyl, (prop-2-yn-1-yloxy)methyl, fluoromethyl, methylthiomethyl, methylsulfinylmethyl, methylsulfonylmethyl, ethylsulfonylmethyl, 2-(hydroxycarbonyl)ethylsulfonylmethyl, hydroxycarbonylmethyl, methoxycarbonylmethyl, ethylaminocarbonylmethyl, methylsulfonylaminomethyl, trifluoromethylsulfonylaminomethyl, (1H-pyrazol-3-yl)carbonylaminomethyl, 1-(trifluoromethyl)-1H-1,2,3-triazol-4-yl, (1-(biotinyl-PEG11)-1H-1,2,3-triazol-4-yl)methoxymethyl, (2H-1,2,3-triazol-4-yl)carbonylaminomethyl, (1H-1,2,3-triazol-5-yl)carbonylaminomethyl, (1-methyl-1H-imidazol-4-yl)carbonylaminomethyl, (1H-imidazol-4-yl)carbonylaminomethyl, (1,5-dimethyl-1H-pyrazol-3-yl)carbonylaminomethyl, (2,5-dioxopyrrolidin-1-yl)methyl, (2,4-dioxoimidazolidin-3-yl)methyl, (1-methyl-2,4-dioxoimidazolidin-3-yl)methyl, hydroxycarbonyl, methoxycarbonyl, aminocarbonyl, methylaminocarbonyl, dimethylaminocarbonyl, ethylaminocarbonyl, isopropylaminocarbonyl, (prop-2-yn-1-yl)aminocarbonyl, (biotinyl-PEG11)aminocarbonyl, hydroxylaminocarbonyl, N2-acetylhydrazinecarbonyl, (1-(biotinyl-PEG11)-1H-1,2,3-triazol-4-yl)methylaminocarbonyl, (1-methyl-1H-imidazol-4-yl)aminocarbonyl, (1,5-dimethyl-1H-pyrazol-3-yl)aminocarbonyl, (2-(3-(but-3-yn-1-yl)-3H-diazirin-3-yl)ethyl)aminocarbonyl, 3-methylisoxazol-5-yl, 1-(2,2,2-trifluoroethyl)-1H-1,2,3-triazol-4-yl, 5-methyl-1,3,4-oxadiazol-2-yl, (6,6-dimethyl-2,4-dioxo-3-azabicyclo[3.1.0]hexan-3-yl)methyl, (1,2-dioxo-3-hydroxy-cyclobut-3-en-4-yl)aminomethyl, (1,2-dioxo-3-amino-cyclobut-3-en-4-yl)aminomethyl, 1-(biotinyl-PEG11)-1H-1,2,3-triazol-4-yl, 1-(hydroxycarbonylmethyl)-1H-1,2,3-triazol-4-yl, 1-(2-(hydroxycarbonyl)ethyl)-1H-1,2,3-triazol-4-yl, 1-(3-(hydroxycarbonyl)prop-1-yl)-1H-1,2,3-triazol-4-yl, 1-(4-(hydroxycarbonyl)but-1-yl)-1H-1,2,3-triazol-4-yl, and 5-methyl-1H-pyrazol-3-yl; and
    • R3f is selected from the group consisting of 5-methyloxazol-2-yl, 1,5-dimethyl-1H-pyrazol-3-yl, 1,5-dimethyl-1H-1,2,4-triazol-3-yl, 4,5-dimethyloxazol-2-yl, 5-fluoro-1-methyl-1H-pyrazol-3-yl, 4,5-dimethylthiazol-2-yl, and 5-methylthiazol-2-yl;
    • or a tautomer, stereoisomer, pharmaceutically acceptable salt, solvate, or polymorph thereof.


More preferably, the compound of the invention is as defined above, and R10 is bromo.


Alternatively, the compound of the invention is as defined above, and R10 is chloro.


In some embodiments, R3f is 5-methyloxazol-2-yl.


In some embodiments, R3f is 1,5-dimethyl-1H-pyrazol-3-yl.


In some embodiments, R3f is 1,5-dimethyl-1H-1,2,4-triazol-3-yl.


In some embodiments, R3f is 4,5-dimethyloxazol-2-yl.


In some embodiments, R3f is 5-fluoro-1-methyl-1H-pyrazol-3-yl.


In some embodiments, R3f is 4,5-dimethylthiazol-2-yl.


In some embodiments, R3f is 5-methylthiazol-2-yl.


More preferably, the compound of the invention is as defined above, and:

    • R3a represents H;
    • R3g represents H; and
    • the R3f substituent and the heterocyclic ring attached by its nitrogen ring member atom to the cyclohexane ring in 1,4 substitution pattern are in trans relationship.


Preferably, the invention concerns the compound of structural formula (I), that is selected from the group consisting of the following compounds:

  • (2S,5S)-5-(4-chlorobenzyl)-2-methyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine;
  • (2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-methylmorpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-((1r,4S)-4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(3-methylisoxazol-5-yl)morpholine;
  • (7S,9aR)-7-(4-chlorobenzyl)-8-(4-(5-methyloxazol-2-yl)cyclohexyl)octahydropyrazino[2,1-c][1,4]oxazine;
  • (7S,9aR)-7-(4-chlorobenzyl)-8-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)octahydropyrazino[2,1-c][1,4]oxazine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • methyl (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylate;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(1-(2,2,2-trifluoroethyl)-1H-1,2,3-triazol-4-yl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-(1-(2,2,2-trifluoroethyl)-1H-1,2,3-triazol-4-yl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)morpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(fluoromethyl)morpholine;
  • ((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methanol;
  • (2R,5S)-5-(4-chlorobenzyl)-N-ethyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N,N-dimethylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-N,N-dimethyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(5-methyl-1,3,4-oxadiazol-2-yl)morpholine;
  • (2R,5S)—N′-acetyl-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carbohydrazide;
  • (2R,5S)-5-(4-chlorobenzyl)-2-(5-methyl-1,3,4-oxadiazol-2-yl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine;
  • (2R,5S)—N′-acetyl-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carbohydrazide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1,5-dimethyl-1H-pyrazole-3-carboxamide;
  • (2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-ethynylmorpholine;
  • N-(35-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxapentatriacontyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-isopropylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-methylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-N,N-dimethylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-N-isopropylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-N-methyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-fluoro-1-methyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)-4-(4-(5-methylthiazol-2-yl)cyclohexyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((prop-2-yn-1-yloxy)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;
  • N-(35-(4-((((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methoxy)methyl)-1H-1,2,3-triazol-1-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxapentatriacontyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide;
  • (2R,5S)-2-(azidomethyl)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-(prop-2-yn-1-yl)morpholine-2-carboxamide;
  • N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1H-pyrazole-3-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-((1-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)-1H-1,2,3-triazol-4-yl)methyl)morpholine-2-carboxamide;
  • N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-2H-1,2,3-triazole-4-carboxamide;
  • 3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)imidazolidine-2,4-dione;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;
  • N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1H-imidazole-4-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohex-3-en-1-yl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohex-3-en-1-yl)-N-ethylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)morpholine-2-carboxamide;
  • (2R,5S)-2-(azidomethyl)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-((1-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)-1H-1,2,3-triazol-4-yl)methyl)morpholine-2-carboxamide;
  • N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methyl)-1H-1,2,3-triazole-5-carboxamide;
  • N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1-methyl-1H-imidazole-4-carboxamide;
  • methyl 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)acetate;
  • 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)acetic acid;
  • 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)-N-ethylacetamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • N-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)methanesulfonamide;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • N-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1,1,1-trifluoromethanesulfonamide;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;
  • (2R,5S)—N-(2-(3-(but-3-yn-1-yl)-3H-diazirin-3-yl)ethyl)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)morpholine-2-carboxamide;
  • 2-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methoxy)acetic acid;
  • 2-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)propan-2-ol;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(5-methyl-1H-pyrazol-3-yl)morpholine;
  • (2R,5R)-2-(4-chlorobenzyl)-1-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-5-(methylsulfonyl)piperidine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamide;
  • 2-(4-((2R,5R)-2-(4-chlorobenzyl)-5-(methylsulfonyl)piperidin-1-yl)cyclohexyl)-5-methyloxazole;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfinyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylthio)methyl)morpholine;
  • 1-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)pyrrolidine-2,5-dione;
  • (2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-2-ethynylmorpholine;
  • 2-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)acetic acid;
  • 3-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)propanoic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohex-3-en-1-yl)-2-((methylsulfonyl)methyl)morpholine;
  • N-(35-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxapentatriacontyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide;
  • 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)acetic acid;
  • 3-((((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methyl)sulfonyl)propanoic acid;
  • 3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2,4-dione;
  • 3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2,4-dione;
  • 3-((((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)amino)-4-hydroxycyclobut-3-ene-1,2-dione;
  • 3-amino-4-((((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)amino)cyclobut-3-ene-1,2-dione;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-2-((ethylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((ethylsulfonyl)methyl)morpholine;
  • 3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1-methylimidazolidine-2,4-dione;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-(1-methyl-1H-imidazol-4-yl)morpholine-2-carboxamide;
  • 4-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)butanoic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-hydroxymorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-N-(1,5-dimethyl-1H-pyrazol-3-yl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carboxamide;
  • 5-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)pentanoic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-N-(1,5-dimethyl-1H-pyrazol-3-yl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamide;
  • N-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carbonyl)-N-methylglycine;
  • (2R,5S)-4-(4-(1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)-5-(4-(trifluoromethyl)benzyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-imidazol-4-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-4-(4-(1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-5-(4-(trifluoromethyl)benzyl)morpholine-2-carboxylic acid;
  • (2S,5S)-5-(4-chlorobenzyl)-2-methyl-4-(4-(5-methyl-1H-pyrazol-3-yl)cyclohexyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-1,2,3-triazol-4-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • methyl (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-1,2,3-triazol-4-yl)cyclohexyl)morpholine-2-carboxylate;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-1,2,3-triazol-4-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-4-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(2-methyl-2H-1,2,3-triazol-4-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(2-methyl-2H-tetrazol-5-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-N,N-dimethyl-4-(4-(5-methyl-1,3,4-thiadiazol-2-yl)cyclohexyl)morpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyl-1,3,4-thiadiazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methoxy-1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • 3-(4-((2R,5S)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)morpholino)cyclohexyl)-1-methyl-1H-1,2,4-triazol-5-ol;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-isopropyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methoxy-1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-N,N-dimethylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-hydroxy-1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-N,N-dimethylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,2-dimethyl-1H-imidazol-4-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-5-(trifluoromethyl)-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiophen-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • 4-(4-((2R,5S)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)morpholino)cyclohexyl)-1-methyl-1H-pyrrole-2-carbonitrile;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-cyano-1-methyl-1H-pyrrol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-ethyl-5-methyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-ethyl-5-methyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-N-ethyl-4-(4-(1-ethyl-5-methyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4-methyl-1H-pyrazol-1-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4-methyl-1H-pyrazol-1-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • N-(39-((((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methyl)sulfonyl)-37-oxo-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azanonatriacontyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-(1H-tetrazol-5-yl)morpholine-2-carboxamide;
  • (2R,5S)-4-(4-(1H-tetrazol-5-yl)cyclohexyl)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)-morpholine;
  • (2R,5S)-4-(4-(1H-1,2,4-triazol-1-yl)cyclohexyl)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)-morpholine;
  • (2R,5S)-4-(4-(1H-pyrazol-1-yl)cyclohexyl)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)-morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4-fluoro-1H-pyrazol-1-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-fluoro-1-methyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • N1-(1,31-bis(((2R,3R,4R,5R,6R)-3-acetamido-4,5-dihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl)oxy)-16-((3-((3-(5-(((2R,3R,4R,5R,6R)-3-acetamido-4,5-dihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl)oxy)pentanamido)propyl)amino)-3-oxopropoxy)methyl)-5,11,21,27-tetraoxo-14,18-dioxa-6,10,22,26-tetraazahentriacontan-16-yl)-N2-(3-(4-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamido)methyl)-1H-1,2,3-triazol-1-yl)propyl)dodecanediamide; and
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-(5-(2-(2,6-dioxopiperidin-3-yl)-3-oxoisoindolin-4-yl)pentyl)morpholine-2-carboxamide;


    or is a tautomer, stereoisomer, pharmaceutically acceptable salt, solvate, or polymorph thereof.


More preferably, the invention concerns the compound of structural formula (I), that is selected from the group consisting of the following compounds:

  • (2S,5S)-5-(4-chlorobenzyl)-2-methyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine;
  • (2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-methylmorpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-((1r,4S)-4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(3-methylisoxazol-5-yl)morpholine;
  • (7S,9aR)-7-(4-chlorobenzyl)-8-(4-(5-methyloxazol-2-yl)cyclohexyl)octahydropyrazino[2,1-c][1,4]oxazine;
  • (7S,9aR)-7-(4-chlorobenzyl)-8-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)octahydropyrazino[2,1-c][1,4]oxazine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • methyl (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylate;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(1-(2,2,2-trifluoroethyl)-1H-1,2,3-triazol-4-yl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-(1-(2,2,2-trifluoroethyl)-1H-1,2,3-triazol-4-yl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)morpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(fluoromethyl)morpholine;
  • ((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methanol;
  • (2R,5S)-5-(4-chlorobenzyl)-N-ethyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N,N-dimethylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-N,N-dimethyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(5-methyl-1,3,4-oxadiazol-2-yl)morpholine;
  • (2R,5S)—N′-acetyl-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carbohydrazide;
  • (2R,5S)-5-(4-chlorobenzyl)-2-(5-methyl-1,3,4-oxadiazol-2-yl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine;
  • (2R,5S)—N′-acetyl-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carbohydrazide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1,5-dimethyl-1H-pyrazole-3-carboxamide;
  • (2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-ethynylmorpholine;
  • N-(35-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxapentatriacontyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-isopropylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-methylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-N,N-dimethylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-N-isopropylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-N-methyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-fluoro-1-methyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)-4-(4-(5-methylthiazol-2-yl)cyclohexyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((prop-2-yn-1-yloxy)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;
  • N-(35-(4-((((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methoxy)methyl)-1H-1,2,3-triazol-1-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxapentatriacontyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide;
  • (2R,5S)-2-(azidomethyl)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-(prop-2-yn-1-yl)morpholine-2-carboxamide;
  • N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1H-pyrazole-3-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-((1-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)-1H-1,2,3-triazol-4-yl)methyl)morpholine-2-carboxamide;
  • N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-2H-1,2,3-triazole-4-carboxamide;
  • 3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)imidazolidine-2,4-dione;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;
  • N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1H-imidazole-4-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohex-3-en-1-yl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohex-3-en-1-yl)-N-ethylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)morpholine-2-carboxamide;
  • (2R,5S)-2-(azidomethyl)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-((1-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)-1H-1,2,3-triazol-4-yl)methyl)morpholine-2-carboxamide;
  • N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methyl)-1H-1,2,3-triazole-5-carboxamide;
  • N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1-methyl-1H-imidazole-4-carboxamide;
  • methyl 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)acetate;
  • 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)acetic acid;
  • 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)-N-ethylacetamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • N-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)methanesulfonamide;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • N-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1,1,1-trifluoromethanesulfonamide;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;
  • (2R,5S)—N-(2-(3-(but-3-yn-1-yl)-3H-diazirin-3-yl)ethyl)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)morpholine-2-carboxamide;
  • 2-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methoxy)acetic acid;
  • 2-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)propan-2-ol;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(5-methyl-1H-pyrazol-3-yl)morpholine;
  • (2R,5R)-2-(4-chlorobenzyl)-1-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-5-(methylsulfonyl)piperidine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamide;
  • 2-(4-((2R,5R)-2-(4-chlorobenzyl)-5-(methylsulfonyl)piperidin-1-yl)cyclohexyl)-5-methyloxazole;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfinyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylthio)methyl)morpholine;
  • 1-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)pyrrolidine-2,5-dione;
  • (2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-2-ethynylmorpholine;
  • 2-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)acetic acid;
  • 3-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)propanoic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohex-3-en-1-yl)-2-((methylsulfonyl)methyl)morpholine;
  • N-(35-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxapentatriacontyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide;
  • 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)acetic acid;
  • 3-((((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methyl)sulfonyl)propanoic acid;
  • 3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2,4-dione;
  • 3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2,4-dione;
  • 3-((((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)amino)-4-hydroxycyclobut-3-ene-1,2-dione;
  • 3-amino-4-((((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)amino)cyclobut-3-ene-1,2-dione;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-2-((ethylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((ethylsulfonyl)methyl)morpholine;
  • 3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1-methylimidazolidine-2,4-dione;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-(1-methyl-1H-imidazol-4-yl)morpholine-2-carboxamide;
  • 4-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)butanoic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-hydroxymorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-N-(1,5-dimethyl-1H-pyrazol-3-yl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carboxamide;
  • 5-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)pentanoic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-N-(1,5-dimethyl-1H-pyrazol-3-yl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamide; and
  • N-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carbonyl)-N-methylglycine;
  • or is a tautomer, stereoisomer, pharmaceutically acceptable salt, solvate, or polymorph thereof.


Still more preferably, the inventive compound as defined above is selected from the group consisting of the following compounds:

  • (2S,5S)-5-(4-chlorobenzyl)-2-methyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine;
  • (2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-methylmorpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-((1r,4S)-4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(3-methylisoxazol-5-yl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(fluoromethyl)morpholine;
  • ((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methanol;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N,N-dimethylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-methylmorpholine-2-carboxamide;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-fluoro-1-methyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohex-3-en-1-yl)-2-((methylsulfonyl)methyl)morpholine;
  • 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)acetic acid;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • N-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)methanesulfonamide;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;
  • (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;
  • (2R,5R)-2-(4-chlorobenzyl)-1-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-5-(methylsulfonyl)piperidine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamide;
  • 2-(4-((2R,5R)-2-(4-chlorobenzyl)-5-(methylsulfonyl)piperidin-1-yl)cyclohexyl)-5-methyloxazole;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfinyl)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylthio)methyl)morpholine;
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohex-3-en-1-yl)-2-((methylsulfonyl)methyl)morpholine;
  • 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)acetic acid; and
  • (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((ethylsulfonyl)methyl)morpholine;


or is a tautomer, stereoisomer, pharmaceutically acceptable salt, solvate, or polymorph thereof.


Pharmaceutical Compositions of the Invention


Another aspect of the invention provides a pharmaceutical composition comprising a therapeutically effective amount of at least one compound according to the invention, and a pharmaceutically acceptable carrier therefor.


The exact nature of the carrier, or, for example excipient or diluent, will depend upon the desired use for the composition, and may be suitable or acceptable for veterinary use and/or suitable or acceptable for human use. The composition may optionally include one or more additional compounds, including one or more additional therapeutic agents.


Compounds of the invention can be combined with other therapeutic agents. The compound of the invention and other therapeutic agent may be administered simultaneously or sequentially. When the other therapeutic agents are administered simultaneously, they can be administered in the same or separate formulations, but they are administered substantially at the same time. The other therapeutic agents are administered sequentially with one another and with compound of the invention, when the administration of the other therapeutic agents and the compound of the invention is temporally separated. The separation in time between the administration of these compounds may be a matter of minutes or it may be longer.


Examples of other therapeutic agents that may be administered with the compounds of the invention include steroids, membrane stabilizers, 5LO inhibitors, leukotriene synthesis and receptor inhibitors, inhibitors of IgE isotype switching or IgE synthesis, inhibitors of IgG isotype switching or IgG synthesis, β-agonists, tryptase inhibitors, acetylsalicylic acid, COX inhibitors, methotrexate, anti-TNF drugs, rituxin, PD4 inhibitors, p38 inhibitors, PDE4 inhibitors, and antihistamines.


Thus, another embodiment of the invention provides a pharmaceutical composition as defined above, that further comprises a therapeutically effective amount of a therapeutic agent selected from the group consisting of steroids, membrane stabilizers, 5LO inhibitors, leukotriene synthesis and receptor inhibitors, inhibitors of IgE isotype switching or IgE synthesis, IgG isotype switching or IgG synthesis, β-agonists, tryptase inhibitors, acetylsalicylic acid, COX inhibitors, methotrexate, anti-TNF drugs, rituxin and other B-cell targeting agents, THF-targeting agents, PD4 inhibitors, p38 inhibitors, PDE4 inhibitors, and antihistamines.


As stated above, an “effective amount” refers to any amount that is sufficient to achieve a desired biological effect. Combined with the teachings provided herein, by choosing among the various active compounds and weighing factors such as potency, relative bioavailability, patient body weight, severity of adverse side-effects and preferred mode of administration, an effective prophylactic or therapeutic treatment regimen can be planned which does not cause substantial unwanted toxicity and yet is effective to treat the particular subject. The effective amount for any particular application can vary depending on such factors as the disease or condition being treated, the particular compound of the invention being administered, the size of the subject, or the severity of the disease or condition. One of ordinary skill in the art can empirically determine the effective amount of a particular compound of the invention and/or other therapeutic agent without necessitating undue experimentation. It is preferred generally that a maximum dose be used, that is, the highest safe dose according to some medical judgment. Multiple doses per day may be contemplated to achieve appropriate systemic levels of compounds. Appropriate systemic levels can be determined by, for example, measurement of the patient's peak or sustained plasma level of the drug. “Dose” and “dosage” are used interchangeably herein.


Generally, daily oral doses of active compounds will be, for human subjects, from about 0.0001 milligrams/kg per day, 0.001 milligrams/kg per day, or 0.01 milligrams/kg per day to about 100 milligrams/kg per day or 1000 milligrams/kg per day. It is expected that oral doses in the range of 0.5 to 50 milligrams/kg, in one or several administrations per day, will yield the desired results. Dosage may be adjusted appropriately to achieve desired drug levels sufficient to achieve or maintain a desired therapeutic effect, local or systemic, depending upon the mode of administration. For example, it is expected that intravenous administration would be from one order to several orders of magnitude lower dose per day. In the event that the response in a subject is insufficient at such doses, even higher doses (or effective higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits. Multiple doses per day are contemplated to achieve appropriate systemic levels of compounds. The compounds may be administered once per week, several times per week (e.g., every other day), once per day or multiple times per day, depending upon, among other things, the mode of administration, the specific indication being treated and the judgment of the prescribing physician.


In one embodiment, intravenous administration of a compound of the invention may typically be from 0.1 mg/kg/day to 20 mg/kg/day.


Determination of an effective dosage of a compound for a particular use and mode of administration is well within the capabilities of those skilled in the art. Effective dosages may be estimated initially from in vitro activity and metabolism assays. For example, an initial dosage of compound for use in animals may be formulated to achieve a circulating blood or serum concentration of the metabolite active compound that is at or above an IC50 of the particular compound as measured in as in vitro assay. Calculating dosages to achieve such circulating blood or serum concentrations taking into account the bioavailability of the particular compound via the desired route of administration is well within the capabilities of skilled artisans. Initial dosages of compound can also be estimated from in vivo data, such as animal models. For any compound described herein the therapeutically effective amount can be initially determined from animal models. A therapeutically effective dose can also be determined from human data for compounds of the invention which have been tested in humans and for compounds which are known to exhibit similar pharmacological activities, such as other related active agents. Higher doses may be required for parenteral administration. The applied dose can be adjusted based on the relative bioavailability and potency of the administered compound. Adjusting the dose to achieve maximal efficacy based on the methods described above and other methods as are well-known in the art is well within the capabilities of the ordinarily skilled artisan.


The formulations of the invention can be administered in pharmaceutically acceptable solutions, which may routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers, adjuvants, and optionally other therapeutic ingredients.


Pharmaceutical compositions comprising the compound of the invention may be manufactured by means of conventional mixing, dissolving, granulating, dragée-making, levigating, emulsifying, encapsulating, entrapping or lyophilization processes. The compositions may be formulated in conventional manner using one or more physiologically acceptable carriers, diluents, excipients or auxiliaries which facilitate processing of the compounds into preparations which can be used pharmaceutically.


For use in therapy, an effective amount of the compound of the invention can be administered to a subject by any mode that delivers the compound of the invention to the desired surface. Administering the pharmaceutical composition of the present invention may be accomplished by any means known to the skilled artisan. Routes of administration include but are not limited to oral, buccal, nasal, rectal, vaginal, ocular, topical, intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, intrathecal, direct injection (for example, into an abscess), mucosal, inhalation, and insufflation.


For oral administration, the compounds (i.e., compounds of the invention, and other therapeutic agents) can be formulated readily by combining the active compound(s) with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragées, lozenges, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a subject to be treated. Pharmaceutical preparations for oral use can be obtained as solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragée cores. Suitable excipients are, in particular, binding agents, fillers, lubricants, disintegrants, and wetting agents. Suitable fillers include sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropyl methyl cellulose, sodium carboxymethyl cellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. Optionally the oral formulations may also be formulated in saline or buffers, e.g., EDTA for neutralizing internal acid conditions or may be administered without any carriers.


Also specifically contemplated are oral dosage forms of the above component or components. The component or components may be chemically modified so that oral delivery of the derivative is efficacious. Generally, the chemical modification contemplated is the attachment of at least one moiety to the component molecule itself, where said moiety permits (a) inhibition of acid hydrolysis; and (b) uptake into the blood stream from the stomach or intestine. Also desired is the increase in overall stability of the component or components and increase in circulation time in the body. Examples of such moieties include: polyethylene glycol, copolymers of ethylene glycol and propylene glycol, carboxymethyl cellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone and polyproline. Abuchowski and Davis, “Soluble Polymer-Enzyme Adducts”, In: Enzymes as Drugs, Hocenberg and Roberts, eds., Wiley-Interscience, New York, N.Y., pp. 367-383 (1981); Newmark et al., J Appl Biochem 4:185-9 (1982). Other polymers that could be used are poly-1,3-dioxolane and poly-1,3,6-tioxocane. Preferred for pharmaceutical usage, as indicated above, are polyethylene glycol moieties.


For the component (or derivative) the location of release may be the stomach, the small intestine (the duodenum, the jejunum, or the ileum), or the large intestine. One skilled in the art has available formulations which will not dissolve in the stomach, yet will release the material in the duodenum or elsewhere in the intestine. Preferably, the release will avoid the deleterious effects of the stomach environment, either by protection of the compound of the invention (or derivative) or by release of the biologically active material beyond the stomach environment, such as in the intestine.


To ensure full gastric resistance a coating impermeable to at least pH 5.0 is essential. Examples of the more common inert ingredients that are used as enteric coatings are cellulose acetate trimellitate (CAT), hydroxypropyl methyl cellulose phthalate (HPMCP), HPMCP 50, HPMCP 55, polyvinyl acetate phthalate (PVAP), Eudragit L30D, Aquateric, cellulose acetate phthalate (CAP), Eudragit L, Eudragit S, and shellac. These coatings may be used as mixed films.


A coating or mixture of coatings can also be used on tablets, which are not intended for protection against the stomach. This can include sugar coatings, or coatings which make the tablet easier to swallow. Capsules may consist of a hard shell (such as gelatin) for delivery of dry therapeutic (e.g., powder); for liquid forms, a soft gelatin shell may be used. The shell material of cachets could be thick starch or other edible paper. For pills, lozenges, molded tablets or tablet triturates, moist massing techniques can be used.


The therapeutic can be included in the formulation as fine multi-particulates in the form of granules or pellets of particle size about 1 mm. The formulation of the material for capsule administration could also be as a powder, lightly compressed plugs or even as tablets. The therapeutic could be prepared by compression.


Colorants and flavoring agents may all be included. For example, the compound of the invention (or derivative) may be formulated (such as by liposome or microsphere encapsulation) and then further contained within an edible product, such as a refrigerated beverage containing colorants and flavoring agents.


One may dilute or increase the volume of the therapeutic with an inert material. These diluents could include carbohydrates, especially mannitol, α-lactose, anhydrous lactose, cellulose, sucrose, modified dextrans and starch. Certain inorganic salts may be also be used as fillers including calcium triphosphate, magnesium carbonate and sodium chloride. Some commercially available diluents are Fast-Flo, Emdex, STA-Rx 1500, Emcompress and Avicell.


Disintegrants may be included in the formulation of the therapeutic into a solid dosage form. Materials used as disintegrates include but are not limited to starch, including the commercial disintegrant based on starch, Explotab. Sodium starch glycolate, Amberlite, sodium carboxymethyl cellulose, ultramylopectin, sodium alginate, gelatin, orange peel, acid carboxymethyl cellulose, natural sponge and bentonite may all be used. Another form of the disintegrants are the insoluble cationic exchange resins. Powdered gums may be used as disintegrants and as binders and these can include powdered gums such as agar, Karaya or tragacanth. Alginic acid and its sodium salt are also useful as disintegrants.


Binders may be used to hold the therapeutic agent together to form a hard tablet and include materials from natural products such as acacia, tragacanth, starch and gelatin. Others include methyl cellulose (MC), ethyl cellulose (EC) and carboxymethyl cellulose (CMC). Polyvinyl pyrrolidone (PVP) and hydroxypropyl methyl cellulose (HPMC) could both be used in alcoholic solutions to granulate the therapeutic.


An anti-frictional agent may be included in the formulation of the therapeutic to prevent sticking during the formulation process. Lubricants may be used as a layer between the therapeutic and the die wall, and these can include but are not limited to; stearic acid including its magnesium and calcium salts, polytetrafluoroethylene (PTFE), liquid paraffin, vegetable oils and waxes. Soluble lubricants may also be used such as sodium lauryl sulfate, magnesium lauryl sulfate, polyethylene glycol of various molecular weights, Carbowax 4000 and 6000.


Glidants that might improve the flow properties of the drug during formulation and to aid rearrangement during compression might be added. The glidants may include starch, talc, pyrogenic silica and hydrated silicoaluminate.


To aid dissolution of the therapeutic into the aqueous environment a surfactant might be added as a wetting agent. Surfactants may include anionic detergents such as sodium lauryl sulfate, dioctyl sodium sulfosuccinate and dioctyl sodium sulfonate. Cationic detergents which can be used and can include benzalkonium chloride and benzethonium chloride. Potential non-ionic detergents that could be included in the formulation as surfactants include lauromacrogol 400, polyoxyl 40 stearate, polyoxyethylene hydrogenated castor oil 10, 50 and 60, glycerol monostearate, polysorbate 40, 60, 65 and 80, sucrose fatty acid ester, methyl cellulose and carboxymethyl cellulose. These surfactants could be present in the formulation of the compound of the invention or derivative either alone or as a mixture in different ratios.


Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. Microspheres formulated for oral administration may also be used. Such microspheres have been well defined in the art. All formulations for oral administration should be in dosages suitable for such administration.


Liquid preparations for oral administration may take the form of, for example, elixirs, solutions, syrups or suspensions, or they may be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters, ethyl alcohol, or fractionated vegetable oils); and preservatives (e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid). The preparations may also contain buffer salts, preservatives, flavoring, coloring and sweetening agents as appropriate.


The pharmaceutical compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the compound(s). The pack may, for example, comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration.


For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.


For topical administration, the compound may be formulated as solutions, gels, ointments, creams, suspensions, etc. as are well-known in the art. Systemic formulations include those designed for administration by injection, e.g., subcutaneous, intravenous, intramuscular, intrathecal or intraperitoneal injection, as well as those designed for transdermal, transmucosal oral or pulmonary administration.


For administration by inhalation, the compounds for use according to the present invention may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichloro-fluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.


Also contemplated herein is pulmonary delivery of the compounds of the invention (or derivatives thereof). The compound of the invention (or derivative) is delivered to the lungs of a mammal while inhaling and traverses across the lung epithelial lining to the blood stream. Other reports of inhaled molecules include Adjei et al., Pharm Res 7:565-569 (1990); Adjei et al., Int J Pharmaceutics 63:135-144 (1990) (leuprolide acetate); Braquet et al., J Cardiovasc Pharmacol 13(suppl. 5):143-146 (1989) (endothelin-1); Hubbard et al., Annal Int Med 3:206-212 (1989) (al-antitrypsin); Smith et al., 1989, J Clin Invest 84:1145-1146 (a-1-proteinase); Oswein et al., 1990, “Aerosolization of Proteins”, Proceedings of Symposium on Respiratory Drug Delivery II, Keystone, Colorado, March, (recombinant human growth hormone); Debs et al., 1988, J Immunol 140:3482-3488 (interferon-gamma and tumor necrosis factor alpha) and Platz et al., U.S. Pat. No. 5,284,656 (granulocyte colony stimulating factor). A method and composition for pulmonary delivery of drugs for systemic effect is described in U.S. Pat. No. 5,451,569, issued Sep. 19, 1995 to Wong et al.


Contemplated for use in the practice of this invention are a wide range of mechanical devices designed for pulmonary delivery of therapeutic products, including but not limited to nebulizers, metered dose inhalers, and powder inhalers, all of which are familiar to those skilled in the art.


Some specific examples of commercially available devices suitable for the practice of this invention are the Ultravent nebulizer, manufactured by Mallinckrodt, Inc., St. Louis, Mo.; the Acorn II nebulizer, manufactured by Marquest Medical Products, Englewood, Colo.; the Ventolin metered dose inhaler, manufactured by Glaxo Inc., Research Triangle Park, North Carolina; the Spinhaler powder inhaler, manufactured by Fisons Corp., Bedford, Mass.; and the Respimat Soft Mist Inhaler, manufactured by Boehringer Ingelheim, Germany. Other hand-driven or human-powered inhaler devices are also applicable.


All such devices require the use of formulations suitable for the dispensing of compound of the invention (or derivative). Typically, each formulation is specific to the type of device employed and may involve the use of an appropriate propellant material, in addition to the usual diluents, adjuvants and/or carriers useful in therapy. Also, the use of liposomes, microcapsules or microspheres, inclusion complexes, or other types of carriers is contemplated. Chemically modified compound of the invention may also be prepared in different formulations depending on the type of chemical modification or the type of device employed.


Formulations suitable for use with a nebulizer, either jet, ultrasonic, or soft mist type, will typically comprise compound of the invention (or derivative) dissolved in water at a concentration of about 0.1 to 25 mg of biologically active compound of the invention per mL of solution. The formulation may also include a buffer and a simple sugar (e.g., for compound of the invention stabilization and regulation of osmotic pressure). The nebulizer formulation may also contain a surfactant, to reduce or prevent surface induced aggregation of the compound of the invention caused by atomization of the solution in forming the aerosol.


Formulations for use with a metered-dose inhaler device will generally comprise a finely divided powder containing the compound of the invention (or derivative) suspended in a propellant with the aid of a surfactant. The propellant may be any conventional material employed for this purpose, such as a chlorofluorocarbon, a hydrochlorofluorocarbon, a hydrofluorocarbon, or a hydrocarbon, including trichlorofluoromethane, dichlorodifluoromethane, dichlorotetrafluoroethanol, and 1,1,1,2-tetrafluoroethane, or combinations thereof. Suitable surfactants include sorbitan trioleate and soya lecithin. Oleic acid may also be useful as a surfactant.


Formulations for dispensing from a powder inhaler device will comprise a finely divided dry powder containing compound of the invention (or derivative) and may also include a bulking agent, such as lactose, sorbitol, sucrose, or mannitol in amounts which facilitate dispersal of the powder from the device, e.g., 50 to 90% by weight of the formulation. The compound of the invention (or derivative) should advantageously be prepared in particulate form with an average particle size of less than 10 micrometers (μm), most preferably 0.5 to 5 μm, for most effective delivery to the deep lung.


Nasal delivery of a pharmaceutical composition of the present invention is also contemplated. Nasal delivery allows the passage of a pharmaceutical composition of the present invention to the blood stream directly after administering the therapeutic product to the nose, without the necessity for deposition of the product in the lung. Formulations for nasal delivery include those with dextran or cyclodextran.


For nasal administration, a useful device is a small, hard bottle to which a metered dose sprayer is attached. In one embodiment, the metered dose is delivered by drawing the pharmaceutical composition of the present invention solution into a chamber of defined volume, which chamber has an aperture dimensioned to aerosolize and aerosol formulation by forming a spray when a liquid in the chamber is compressed. The chamber is compressed to administer the pharmaceutical composition of the present invention. In a specific embodiment, the chamber is a piston arrangement. Such devices are commercially available.


Alternatively, a plastic squeeze bottle with an aperture or opening dimensioned to aerosolize an aerosol formulation by forming a spray when squeezed is used. The opening is usually found in the top of the bottle, and the top is generally tapered to partially fit in the nasal passages for efficient administration of the aerosol formulation. Preferably, the nasal inhaler will provide a metered amount of the aerosol formulation, for administration of a measured dose of the drug.


The compounds, when it is desirable to deliver them systemically, may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as sterile suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.


Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.


Alternatively, the active compounds may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, buffer, dextrose solution, before use. To this end, the active compound may be dried by any art-known technique, such as lyophilization, and reconstituted prior to use.


The compounds may also be formulated in rectal or vaginal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.


For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are known in the art.


For ocular administration, the compound(s) may be formulated as a solution, emulsion, suspension, etc. suitable for administration to the eye. A variety of vehicles suitable for administering compounds to the eye are known in the art.


In addition to the formulations described above, for prolonged delivery, the compounds may also be formulated as a depot preparation for administration by, for example, implantation or intramuscular injection. Such long acting formulations may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt. Alternatively, transdermal delivery systems manufactured as an adhesive disc or patch which slowly releases the compound for percutaneous absorption may be used. To this end, permeation enhancers may be used to facilitate transdermal penetration of the compound.


The pharmaceutical compositions also may comprise suitable solid or gel phase carriers or excipients. Examples of such carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.


Suitable liquid or solid pharmaceutical preparation forms are, for example, aqueous or saline solutions for inhalation, microencapsulated, encochleated, coated onto microscopic gold particles, contained in liposomes, nebulized, aerosols, pellets for implantation into the skin, or dried onto a sharp object to be scratched into the skin. The pharmaceutical compositions also include granules, powders, tablets, coated tablets, (micro)capsules, suppositories, syrups, emulsions, suspensions, creams, drops or preparations with protracted release of active compounds, in whose preparation excipients and additives and/or auxiliaries such as disintegrants, binders, coating agents, swelling agents, lubricants, flavorings, sweeteners or solubilizers are customarily used as described above. The pharmaceutical compositions are suitable for use in a variety of drug delivery systems. For a brief review of methods for drug delivery, see Langer R, Science 249:1527-33 (1990), which is incorporated herein by reference.


The compounds of the invention and optionally other therapeutics may be administered per se (neat) or in the form of a pharmaceutically acceptable salt. When used in medicine the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically acceptable salts thereof. Such salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, maleic, acetic, salicylic, p-toluene sulphonic, tartaric, citric, methane sulphonic, formic, malonic, succinic, naphthalene-2-sulphonic, and benzene sulphonic. Also, such salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts of the carboxylic acid group. Typically, such salts are more soluble in aqueous solutions than the corresponding free acids and bases, but salts having lower solubility than the corresponding free acids and bases may also be formed.


The compounds may alternatively be formulated in the pharmaceutical composition per se, or in the form of a hydrate, solvate, or N-oxide.


Suitable buffering agents include: acetic acid and a salt (1-2% w/v); citric acid and a salt (1-3% w/v); boric acid and a salt (0.5-2.5% w/v); and phosphoric acid and a salt (0.8-2% w/v). Suitable preservatives include benzalkonium chloride (0.003-0.03% w/v); chlorobutanol (0.3-0.9% w/v); parabens (0.01-0.25% w/v) and thimerosal (0.004-0.02% w/v).


Pharmaceutical compositions of the invention contain an effective amount of a compound of the invention and optionally therapeutic agents included in a pharmaceutically acceptable carrier. The term “pharmaceutically acceptable carrier” means one or more compatible solid or liquid filler, diluents or encapsulating substances which are suitable for administration to a human or other vertebrate animal. The term “carrier” denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application. The components of the pharmaceutical compositions also are capable of being commingled with the compounds of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficiency.


The therapeutic agent(s), including specifically but not limited to the compound of the invention, may be provided in particles. Particles as used herein means nanoparticles or microparticles (or in some instances larger particles) which can consist in whole or in part of the compound of the invention or the other therapeutic agent(s) as described herein. The particles may contain the therapeutic agent(s) in a core surrounded by a coating, including, but not limited to, an enteric coating. The therapeutic agent(s) also may be dispersed throughout the particles. The therapeutic agent(s) also may be adsorbed into the particles. The particles may be of any order release kinetics, including zero-order release, first-order release, second-order release, delayed release, sustained release, immediate release, and any combination thereof, etc. The particle may include, in addition to the therapeutic agent(s), any of those materials routinely used in the art of pharmacy and medicine, including, but not limited to, erodible, nonerodible, biodegradable, or nonbiodegradable material or combinations thereof. The particles may be microcapsules which contain the compound of the invention in a solution or in a semi-solid state. The particles may be of virtually any shape.


Both non-biodegradable and biodegradable polymeric materials can be used in the manufacture of particles for delivering the therapeutic agent(s). Such polymers may be natural or synthetic polymers. The polymer is selected based on the period of time over which release is desired. Bioadhesive polymers of particular interest include bioerodible hydrogels described in Sawhney H S et al. (1993) Macromolecules 26:581-7, the teachings of which are incorporated herein. These include polyhyaluronic acids, casein, gelatin, glutin, polyanhydrides, polyacrylic acid, alginate, chitosan, poly(methyl methacrylates), poly (ethyl methacrylates), poly(butylmethacrylate), poly(isobutyl methacrylate), poly(hexylmethacrylate), poly(isodecyl methacrylate), poly(lauryl methacrylate), poly(phenyl methacrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate), and poly(octadecyl acrylate).


The therapeutic agent(s) may be contained in controlled release systems. The term “controlled release” is intended to refer to any drug-containing formulation in which the manner and profile of drug release from the formulation are controlled. This refers to immediate as well as non-immediate release formulations, with non-immediate release formulations including but not limited to sustained release and delayed release formulations. The term “sustained release” (also referred to as “extended release”) is used in its conventional sense to refer to a drug formulation that provides for gradual release of a drug over an extended period of time, and that preferably, although not necessarily, results in substantially constant blood levels of a drug over an extended time period. The term “delayed release” is used in its conventional sense to refer to a drug formulation in which there is a time delay between administration of the formulation and the release of the drug there from. “Delayed release” may or may not involve gradual release of drug over an extended period of time, and thus may or may not be “sustained release.”


Use of a long-term sustained release implant may be particularly suitable for treatment of chronic conditions. “Long-term” release, as used herein, means that the implant is constructed and arranged to deliver therapeutic levels of the active ingredient for at least 7 days, and preferably 30-60 days. Long-term sustained release implants are well-known to those of ordinary skill in the art and include some of the release systems described above.


It will be understood by one of ordinary skill in the relevant arts that other suitable modifications and adaptations to the compositions and methods described herein are readily apparent from the description of the invention contained herein in view of information known to the ordinarily skilled artisan, and may be made without departing from the scope of the invention or any embodiment thereof.


Methods and Uses


As shown herein, the compounds of the invention are useful for inhibiting binding activity of the chitinase-like protein YKL-40.


Accordingly, the invention provides methods for inhibiting the chitinase-like protein YKL-40 in a cell or a tissue, comprising contacting the cell or the tissue with at least one compound according to the invention, or with a pharmaceutical composition according to the invention.


In other aspects, the invention provides methods for the treatment or prevention of a disease, disorder, or condition associated with aberrant expression or activity of the chitinase-like protein YKL-40, comprising administering to a subject in need thereof a therapeutically effective amount of at least one compound according to the invention, or of a pharmaceutical composition according to the invention.


Preferably, the disease, disorder, or condition is selected from the group consisting of cancer, allergic diseases, acute and chronic inflammatory diseases, autoimmune diseases, dental diseases, neurologic diseases, metabolic diseases, liver diseases, kidney diseases, polycystic ovary syndrome, endometriosis, asthma, and fibrotic disorders.


In one particular aspect, the disease is cancer selected from the group consisting of glioblastoma, breast cancer, colon cancer, kidney cancer, uterine cancer, primary and metastatic lung cancer, mesothelioma, osteosarcoma, malignant melanoma, ovarian cancer, cervical cancer, prostate cancer, liver cancer, gastric cancer, metastatic renal cancer, leukemia, lymphoma, oligodendroglioma, glioblastoma, and germ cell tumors.


In another particular aspect, the disease, disorder, or condition is an allergic disease selected from the group consisting of asthma, allergic rhinitis, seasonal allergic rhinitis, chronic rhinosinusitis with or without nasal polyps, conjunctivitis, keratoconjunctivitis, seasonal allergic conjunctivitis, dry eye syndrome, eosinophilic esophagitis, celiac disease, food allergy, irritable bowel syndrome, irritable bowel disease, atopic eczema, atopic dermatitis, allergic contact dermatitis, eosinophilic otitis media, eosinophilic pneumonia, and IgG4 mediated diseases.


In a further particular aspect, disease, disorder, or condition is an acute or chronic inflammatory disease selected from the group consisting of fungal diseases, parasitic infections, celiac disease, microscopic colitis, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis, interstitial lung diseases, cystic fibrosis, Hermansky-Pudlak syndrome, and Alzheimer's disease.


In another aspect, the disease, disorder, or condition is an autoimmune disorder selected from the group consisting of inflammatory bowel disease, ulcerative colitis, Crohn's disease, rheumatoid arthritis, osteoarthritis, psoriasis, scleroderma, multiple sclerosis, Sjögren's syndrome, atherosclerosis, and sarcoidosis.


In one particular aspect, the disease, disorder, or condition is periodontitis.


In another aspect, the disease, disorder, or condition is a metabolic disease selected from the group consisting of insulin-dependent diabetes mellitus and non-insulin-dependent diabetes mellitus.


In a further aspect, the disease, disorder, or condition is a liver disease selected from the group consisting of non-alcoholic fatty liver disease, non-alcoholic steatohepatitis, hepatitis-C virus-induced fibrosis and cirrhosis, and alcoholic fibrosis.


In another aspect, the disease is a kidney disease selected from the group consisting of nephropathy (e.g., diabetic nephropathy), focal segmental glomerulosclerosis, tubulointerstitial fibrosis, postransplant fibrosis, and retroperitoneal fibrosis (Ormond's disease).


In one particular aspect, the disease, disorder, or condition is a fibrotic disorder.


Preferably, the fibrotic disorder is idiopathic pulmonary fibrosis (IPF).


In another aspect, the disease is a storage disease selected from the group consisting of Gaucher disease, Fabry disease, lysosomal storage disorders, Niemann-Pick disease, nephropathic cystinosis, and X-linked globotriaosylceramidosis.


Moreover, the invention provides methods of treating diseases caused by infectious agents, such as fungi, worms, and parasites, the method comprising administering to a subject in need of such treatment an effective amount of one or more compounds of the invention.


In one embodiment, the invention provides methods of treating allergies, comprising administering to a subject in need of such treatment an effective amount of one or more compounds of the invention. In certain embodiments, such allergies are caused by any of a variety of antigens including biological sources such as dust mites, mold, cockroaches and other insects, dander from pets or other mammals, pollens, spores, mold, other fungal sources, and other plant antigens, or non-biological sources such as chemicals (e.g., isocyanates).


In other embodiments, the invention provides a method of screening for therapeutic agents useful for treating asthma in a mammal, comprising: (a) contacting the chitinase-like protein YKL-40 with a compound (e.g., a compound of the invention) and a substrate of said chitinase-like protein; and (b) determining if the compound inhibits the binding activity of the chitinase-like protein; wherein if the compound inhibits the binding activity of the chitinase-like protein, then the compound is a therapeutic agent useful for treating asthma.


In other aspects, the invention provides methods for monitoring the efficacy of a treatment for asthma, comprising (a) administering a compound of the invention to a mammal, and (b) monitoring the expression of the chitinase-like protein YKL-40 in the mammal after administration of the compound, wherein a decrease in the expression of the chitinase-like protein YKL-40 indicates that the compound is useful in treating asthma, allergic diseases such as hay fever, allergic rhinitis, atopic dermatitis or other Th-2 mediated or associated diseases.


In other aspects, the invention provides methods for monitoring the efficacy of a treatment for asthma and other allergic diseases, comprising (a) administering a compound of the invention to a mammal, and (b) monitoring the expression of inflammatory mediators such as IL-13, IL-5, IL-4, eotaxin, or IgE or inflammatory cells such as eosinophils, neutrophils, or lymphocytes in broncho-alveolar washings, sputum, or tissues obtained from the mammal after administration of the compound; wherein a decrease in expression indicates that the compound is useful in treating asthma or allergic diseases such as hay fever, allergic rhinitis, atopic dermatitis or other Th-2 mediated or associated diseases.


In another aspect, the invention provides methods for assessing the efficacy of an agent for treating asthma in a subject, comprising the steps of:

    • a) detecting in a subject sample collected at a first point in time the expression level of the chitinase-like protein YKL-40;
    • b) repeating step a) at one or more subsequent points in time after administration of the agent; and
    • c) comparing expression level of the chitinase-like protein YKL-40 detected in step a) with the expression level(s) detected in step(s) b),


      wherein a higher expression level of the chitinase-like protein YKL-40 at the first point in time relative to at least one subsequent point in time indicates that the agent is efficacious in treating asthma.


In certain embodiments, an agent identified by such a method is efficacious in treating asthma, hay fever, allergic rhinitis, atopic dermatitis, allergic reactions, or a disorder associated with Th-2.


Alternatively, the efficacy of an agent for treating asthma or an allergic reaction can be assessed via measuring the expression level of an inflammatory mediator such as IL-13, IL-5, IL-4, eotaxin, IgE, or measuring the amount of inflammatory cells such as eosinophils, neutrophils, or lymphocytes in broncho-alveolar washings, sputum, or tissues obtained from a mammal. In certain such embodiments, the expression level can be measured prior to and after administration of an agent. When the expression level of the inflammatory mediator or the level of inflammatory cells decreases after administration of an agent, such an agent is efficacious in treating asthma, hay fever, allergic rhinitis, atopic dermatitis, allergic reactions, or a disorder associated with Th-2.


Another aspect of the invention provides methods of identifying an agent for treating asthma, comprising:

    • a) contacting a sample comprising the chitinase-like protein YKL-40 with the agent; and
    • b) determining the ability of the agent to inhibit binding activity of the chitinase-like protein YKL-40, wherein decreased activity of the chitinase-like protein YKL-40 identifies an agent for treating asthma.


In certain embodiments, the binding activity of the chitinase-like protein YKL-40 is assessed by MicroScale Thermophoresis direct binding assay and AlphaScreen indirect binding assays using fluorescently labelled YKL-40 protein and biotinylated small molecule compound binding YKL-40, respectively.


Another aspect of the invention provides a method of identifying compounds that interfere with the chitinase-like protein YKL-40 and asialofetuin interaction by:

    • (1) in a binding competition assay, subjecting (i) a compound according to the invention, and (ii) biotinylated asialofetuin to YKL-40 or derivative of YKL-40 comprising a purification tag; and
    • (2) assessing the resulting AlphaScreen luminescence signal; wherein an emission signal decline from the high level at lowest inhibitor concentration to the lower level (diminished by the value of at least 3× standard deviation of the noise displayed at plateau values at highest concentrations of inhibitor in the response plots for IC50 determinations) at highest inhibitor concentration indicates that the compound interferes with the chitinase-like protein YKL-40 and asialofetuin interaction.


Therapeutic Applications


The inventive compounds are useful for inhibiting the biological activity of the chitinase-like protein YKL-40. The expression of chitinase-like protein YKL-40 has been linked to interstitial lung diseases (ILDs) including idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD).


In some embodiments, the invention provides the inventive compounds for the treatment or prevention of a disease or condition associated with expression or biological activity of YKL-40 in a subject in need thereof. For instance, the disease, disorder, or condition is selected from the group consisting of cancer, allergic diseases, acute and chronic inflammatory diseases, autoimmune diseases, dental diseases, neurologic diseases, metabolic diseases, liver diseases, polycystic ovary syndrome, endometriosis, asthma, and fibrotic disorders.


According to certain embodiments, the invention provides the inventive compounds for the treatment of allergic diseases, such as asthma, allergic rhinitis, seasonal allergic rhinitis, chronic rhinosinusitis with or without nasal polyps, conjunctivitis, keratoconjunctivitis, seasonal allergic conjunctivitis, dry eye syndrome, eosinophilic esophagitis, celiac disease, food allergies, irritable bowel syndrome, irritable bowel disease, atopic eczema, atopic dermatitis, allergic contact dermatitis, eosinophilic otitis media, eosinophilic pneumonia, and IgG4 mediated disease.


In certain embodiments, the reaction caused by an allergen is allergic rhinitis or atopic dermatitis.


In certain embodiments, the reaction caused by an allergen is characterized by the occurrence of one or more symptoms, which can include red eyes, itchiness, runny nose, eczema, impaired hearing, hives, an asthma attack, increased mucus production in the lungs, coughing, wheezing, and shortness of breath.


Exemplary acute and chronic inflammatory disorders that can be treated using the inventive compounds include fungal diseases, parasitic infection, celiac disease, microscopic colitis, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis, interstitial lung diseases, cystic fibrosis (CF), Hermansky-Pudlak and Alzheimer's disease (AD).


In certain embodiments, the disease or condition to be treated is an autoimmune disorder selected from the group consisting of inflammatory bowel disease, ulcerative colitis (UC), Crohn's disease (CD), rheumatoid arthritis (RA), osteoarthritis, psoriasis, scleroderma, multiple sclerosis (MS), Sjögren's syndrome, atherosclerosis, and sarcoidosis.


The inventive compounds are also useful for treating dental diseases such as periodontitis.


The compounds of the invention are also useful for treating metabolic diseases such as insulin-dependent diabetes mellitus (IDDM) and non-insulin-dependent diabetes mellitus (NIDDM).


In certain embodiments, the invention provides the inventive compounds for the treatment of a liver disease selected from group consisting of non-alcoholic fatty liver disease, non-alcoholic steatohepatitis, hepatitis-C virus-induced fibrosis and cirrhosis, and alcoholic fibrosis.


In some embodiments, the invention provides the inventive compounds for the treatment of cancer, wherein the cancer is selected from the group consisting of glioblastoma, breast cancer, colon cancer, kidney cancer, uterine cancer, primary and metastatic lung cancer, mesothelioma, osteosarcoma, malignant melanoma, ovarian cancer, cervical cancer, prostate cancer, liver cancer, gastric cancer, metastatic renal cancer, leukemia, lymphoma, oligodendroglioma, glioblastoma and germ cell tumors.


In certain embodiments, the disease or condition to be treated is a kidney disease selected from the group consisting of nephropathy (e.g., diabetic nephropathy), focal segmental glomerulosclerosis, tubulointerstitial fibrosis, postransplant fibrosis, and retroperitoneal fibrosis (Ormond's disease).


In certain embodiments, the disease or condition to be treated is a storage disease selected from the group consisting of Gaucher disease, Fabry disease, lysosomal storage disorders, Niemann-Pick disease, nephropathic cystinosis, and X-linked globotriaosylceramidosis.


In some embodiments, the subject receiving treatment is a mammal. For instance, the methods and uses described herein are suitable for medical use in humans. Alternatively, the methods and uses are also suitable in a veterinary context, wherein the invention may be administered to warm-blooded animals, birds and reptiles. Warm-blooded animals include, for example, all non-human primates (e.g., chimpanzee and ape), ruminants (e.g., cow, sheep and goat), porcines (e.g., pig), equines (e.g., horse, mule and donkey), camelines (e.g., camel and dromedary), canines (e.g., dog), felines (e.g., cat), leporine (e.g., rabbit), murines (e.g., mouse and rat) cavines (e.g., guinea pig), gerbiline (e.g., gerbil), cricetine (e.g., hamster), mustelines (e.g., ferret and weasel) and chinchilines (e.g., chinchilla). Birds include animals of the avian class, for example, all phasianines (e.g., chicken and quail), anserines (e.g., goose), anatines (e.g., ducks), meleagridines (e.g., turkey), daruduelines (e.g., canary), psittacines (e.g., parrot, macaw, parakeet and lovebird), cacatuines (e.g., cockatoo) and columbines (e.g., pigeon and turtle dove).


In certain embodiments, the inventive compound or pharmaceutical composition thereof is preferably administered to domesticated companion animals and to productive and breeding animals.


In certain embodiments, the treatment further comprises administering a second therapeutic agent. Exemplary second therapeutic agents include steroids, membrane stabilizers, 5LO inhibitors, leukotriene synthesis and receptor inhibitors, inhibitors of IgE isotype switching or IgE synthesis, inhibitors of IgG isotype switching or IgG synthesis, β-agonists, tryptase inhibitors, acetylsalicylic acid, COX inhibitors, methotrexate, anti-TNF drugs, rituxin and other B-cell targeting agents, THF-targeting agents, PD4 inhibitors, p38 inhibitors, PDE4 inhibitors, and antihistamines.


EXAMPLES

The present invention is further illustrated by the following examples, which in no way should be construed as limiting the scope of the claimed invention.


Materials and Methods of Preparation and Characterization

The compounds of the present disclosure may be prepared by use of known chemical reactions and procedures. Representative methods for synthesizing compounds of the disclosure are presented below. It is understood that the nature of the substituents required for the desired target compound often determines the preferred method of synthesis. All variable groups of these methods are as described in the generic description if they are not specifically defined below. Substituents carry the same meaning as defined above, unless otherwise noted.


Those having skill in the art will recognize that the starting materials and reaction conditions may be varied, the sequence of the reactions altered, and additional steps employed to produce compounds encompassed by the present disclosure, as demonstrated by the following examples. Many general references providing commonly known chemical synthetic schemes and conditions useful for synthesizing the disclosed compounds are available (see, e.g., Smith and March, March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, Fifth Edition, Wiley-Interscience, 2001; or Vogel, A Textbook of Practical Organic Chemistry, Including Qualitative Organic Analysis, Fourth Edition, New York: Longman, 1978).


The reactions are performed in a solvent appropriate to the reagents and materials employed and suitable for the transformations being effected. It will be understood by those skilled in the art of organic synthesis that the functionality present on the molecule should be consistent with the transformations proposed. This will sometimes require a judgment to modify the order of the synthetic steps or to select one particular process scheme over another in order to obtain a desired compound of the disclosure.


In some cases, protection of certain reactive functionalities may be necessary to achieve some of the above transformations. In general, the need for such protecting groups as well as the conditions necessary to attach and remove such groups will be apparent to those skilled in the art of organic synthesis. An authoritative account describing the many alternatives to the trained practitioner are J. F. W. McOmie, “Protective Groups in Organic Chemistry,” Plenum Press, London and New York 1973, in T. W. Greene and P. G. M. Wuts, “Protective Groups in Organic Synthesis,” Third edition, Wiley, New York 1999, in “The Peptides;” Volume 3 (editors: E. Gross and J. Meienhofer), Academic Press, London and New York 1981, in “Methoden der organischen Chemie,” Houben-Weyl, 4th edition, Vol. 15/1, Georg Thieme Verlag, Stuttgart 1974, in H.-D. Jakubke and H. Jescheit, “Aminosauren, Peptide, Proteine,” Verlag Chemie, Weinheim, Deerfield Beach, and Basel 1982, and/or in Jochen Lehmann, “Chemie der Kohlenhydrate: Monosaccharide und Derivate,” Georg Thieme Verlag, Stuttgart 1974. The protecting groups may be removed at a convenient subsequent stage using methods known from the art. The disclosures of all articles and references mentioned in this application, including patents, are incorporated herein by reference in their entirety.


Starting materials can be obtained from commercial sources or prepared by well-established literature methods known to those skilled in the art.


All solvents, substrates and reagents that were commercially available were used without further purification. TLC analysis was performed using pre-coated glass plates (0.2±0.03 mm thickness, GF-254, particle size 0.01-0.04 mm) from Fluorochem Ltd, UK or using pre-coated glass plates (TLC silica gel 60 F254) or using pre-coated aluminium plates (TLC silica gel 60 F254) from Merck. TLC analysis was performed for compounds before their transferring in the appropriate salts (hydrochloride or TFA salt).


The column chromatography was performed using high-purity grade silica gel (pore size 60 Å, 220-440 mesh particle size, 35-75 μm particle size) from Fluka or using high-purity grade silica gel (pore size 60 Å, 230-400 mesh particle size, 40-63 μm particle size) from Merck.


Column chromatography (LCC) was performed using high-purity grade silica gel (pore size 60 Å, 220-440 mesh particle size, 35-75 μm particle size) purchased from Fluka.


Flash column chromatography (FLCC) was performed using Buchi Reverelis® X2-UV equipped in evaporative light scattering detector (ELSD), with UV detection in range UV 200-500 nm, purchased from BOCHI Labortechnik AG, using puriFlash® pre-packed cartridges with amorphous or spherical virgin silica gel, 50 μm, from InteiChim®.


Preparative HPLC were performed on preparative Shimadzu HPLC system containing two pumps LC-20AP, CBM-20A communication module and ELSD-LTII detector with the use of:

    • Phenomenex Luna 21.2/250 mm, 5 μm C-18(2) 100 Å column;
    • Thermo Scientific Hypersil GOLD 21.2/250 mm, 5 μm C-18 column;
    • Phenomenex Luna® 250×30 mm, 5 μm Phenyl-Hexyl 100 Å column;
    • Phenomenex Luna 250×21.2 mm, 5 μm Phenyl-Hexyl 100 Å column;
    • Phenomenex Lux® 250×21.2 mm Cellulose-4 column;


      or on preparative Shimadzu HPLC system containing LC-20AP pumps, CBM-20A communication module and SPD-20A UV/VIS detector and FRC-20 fraction collector with the use of:
    • Phenomenex Luna 250×21.2 mm, 5 μm Phenyl-Hexyl 100 Å column;
    • Phenomenex Luna® 250×30 mm, 5 μm Phenyl-Hexyl 100 Å column.


      The target compounds, when subjected to reversed-phase chromatographic purification in the presence of TFA, were usually obtained in the form of TFA salts.



1H NMR spectra were recorded on an Agilent 400-MR DD2 400 MHz spectrometer and on Bruker AVANCE DRX500, AVANCE DRX600 and on Bruker AVANCE II PLUS (respectively at 500, 600, or 700 MHz) NMR spectrometers.


All spectra were recorded in appropriate deuterated solvents (CDCl3, DMSO-d6, D2O, Methanol-d4, etc.) that were commercially available.


Resonances are given in parts per million (ppm) relative to tetramethylsilane internal standard. Data are reported as follows: chemical shift (δ), multiplicity (s=singlet, d=doublet, t=triplet, m=multiplet, bs=broad singlet), coupling constants (J in Hz) and integration.


ESI-MS spectra were obtained on:

    • A) Waters Alliance 2695 separation module with a PDA 1996 UV detector and Waters Micromass ZQ 2000 mass detector equipped with Phenomenex Kinetex 2.1/50 mm, 2.6 μm C-18 column eluted with 0.3 mL/min flow of 3-100% gradient (over 6 min) of acetonitrile in water,
    • B) Shimadzu LCMS system containing LCMS-2020 and SPD-M20A detectors, SIL-20AHT autosampler, LC-20AD pump with LPG module, DGU-20A3R degasser and CMB-20A communication module equipped with:
      • Phenomenex Kinetex® 2.1/50 mm, 2.6 μm C-18 column eluted with 0.5 mL/min flow of 10-90% gradient (over 6 or 8 min) of acetonitrile in water;
      • Phenomenex Kinetex® 2.6 mm XB C-18 100 Å LC Column 50×2.1 mm eluted with 0.5 mL/min flow of 10-90% gradient (over 10 or 16 m) of acetonitrile in water;
      • Phenomenex Luna® 3 mm Phenyl-Hexyl 100 Å LC Column 100×3 mm eluted with 0.5 mL/min flow of 10-90% gradient (over 10 or 16 or 25 m) of acetonitrile in water.
    • C) Shimadzu LCMS system containing LCMS-2020 and SPD-M20A detectors, SIL-30ACMP autosampler, two LC-30AD pumps, DGU-20A5R degasser and CTO-20AC column oven equipped with:
      • Phenomenex Luna, C18, 2 μm, 100A, 150×3 mm column eluted with 0.5 mL/min flow of 15-90% gradient (over 13 min) of acetonitrile in water or
      • Phenomenex Kinetex 2.1/30 mm, 1.7 μm XB-C18 100 Å LC column eluted with 1 mL/min flow of 10-90% gradient (over 3 mA) of acetonitrile in water.


ESI-MS spectra were obtained in one of the following Methods:















Method
UPLC, Solid phase: Kinetex ® 1.7 μm, XB-C18, LC


A
column 30 × 2.1 mm; Mobile phase: Acetonitrile in water,



1.0 mL/min, gradient










10


%




2.

min



90



%




1.

min



90


%









Method
UPLC, Solid phase: Kinetex ® 1.7 μm, XB-C18, LC


B
column 30 × 2.1 mm; Mobile phase: Acetonitrile in water,



1.0 mL/min, gradient










10


%




2.

min



70



%




0


min



90



%




1.

min



90


%









Method
HPLC, Solid phase: Luna ® 3 μm, Phenyl-Hexyl 100 A,


C
LC column 100 × 3 mm; Mobile Phase: Acetonitrile in



water, 1.5 mL/min, gradient










10


%




4.5

min



70



%




0


min



90



%




2.5

min



90



%




0


min



10



%




3.

min



10


%









Method
UPLC, Solid phase: Kinetex ® 1.7 μm, XB-C18, LC


D
column 30 × 2.1 mm; Mobile phase: Acetonitrile in water,



1.5 mL/min, gradient










10


%




5.

min



90



%




1.

min



90



%




0


min



10



%




2.

min



10


%









Method
UPLC, Solid phase: Kinetex ® 1.7 μm, XB-C18, LC


E
column 30 × 2.1 mm; Mobile phase: Acetonitrile in water,



1.0 mL/min, gradient










0


%




2.

min



50



%




1.

min



50


%









Method
UPLC, Solid phase: SecurityGuard ™ ULTRA Cartridges


F
UHPLC C18 2.1 mm ID Columns; Kinetex ® 1.7 μm,



XB-C18, LC column 30 × 2.1 mm, 100 Å; Mobile phase:



Acetonitrile in water, 0.5 mL/min, gradient










10


%




5.

min



90



%




1.

min



90



%




0


min



10



%




2.

min



10


%













Formic acid was used at a concentration of 0.5 weight % (1.1 mL of HCOOH per 2.5 L of the solvent) in all HPLC systems.


Abbreviations used are those conventional in the art or the following:

    • Ac=acetyl,
    • AcCl=acetyl chloride,
    • AcOH=acetic acid,
    • Alloc=allyloxycarbonyl,
    • aq=aqueous,
    • BINAP=2,2′-bis(diphenylphosphino)-1,1′-binaphthyl,
    • Bn=benzyl,
    • Boc=tert-butoxycarbonyl,
    • tBu=tert-butyl,
    • ° C.=degree Celsius,
    • CDI=carbonyldiimidazole,
    • COD=1,5-cyclooctadiene,
    • DAST=diethylaminosulfur trifluoride,
    • dba=dibenzylideneacetone,
    • DCE=1,2-dichloroethane,
    • DCM=dichloromethane,
    • DIAD=diisopropyl azodicarboxylate,
    • DIPEA=N,N-diisopropylethylamine,
    • DMAP=4-dimethylaminopyridine,
    • DMF=N,N-dimethylformamide,
    • DMS=dimethyl sulfide,
    • DMSO=dimethyl sulfoxide,
    • dppf=1,1′-ferrocenediyl-bis(diphenylphosphine),
    • EDCI=1-ethyl-3-(3-dimethylaminopropyl)carbodiimide,
    • ELSD=evaporative light scattering detector,
    • ESI-MS=electrospray ionization mass spectrometry,
    • Et3N=triethylamine,
    • EtOAc or AcOEt=ethyl acetate,
    • EtOH=ethanol,
    • FLCC=flash column chromatography,
    • g=gram,
    • h=hour(s),
    • HATU=1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide
    • hexafluorophosphate,
    • HPLC=high pressure liquid chromatography,
    • iPrOH=isopropyl alcohol,
    • K=kelvin,
    • L=liter,
    • LCC=liquid column chromatography,
    • LC-MS=liquid chromatography and mass spectrometry,
    • MeCN=acetonitrile,
    • MeOH=methanol,
    • min=minutes,
    • mL=milliliter(s),
    • M=molar,
    • mmol=millimoles,
    • MOMCl=chloromethyl methyl ether,
    • Ms=mesyl (methanesulfonyl),
    • MTBE=methyl tert-butyl ether,
    • m/z=mass to charge ratio,
    • μg=microgram,
    • μL=microliter,
    • μM=micromolar,
    • nM=nanomolar,
    • NMM=N-methylmorpholine,
    • NMP=N-methyl-2-pyrrolidone,
    • NMR=nuclear magnetic resonance,
    • N=normal,
    • PEG11=polyoxyethylene glycol,
    • RT or rt=room temperature,
    • Rt=retention time,
    • TBDMSCl=tert-butyldimethylsilyl chloride,
    • TBTU=2-(1H-benzotriazole-1-yl)-1,1,3,3-tetramethylaminium tetrafluoroborate,
    • TFA=trifluoroacetic acid,
    • TfCl=trifluoromethanesulfonyl chloride,
    • THF=tetrahydrofuran,
    • TLC=thin layer chromatography,
    • TMSCl=chlorotrimethylsilane.


If not otherwise defined, purity of a solid substance is expressed as a ratio of the weight of the component in question to the total weight, multiplied by 100 (weight %); purity of a liquid is expressed as a ratio of the volume of the component in question to the total volume, multiplied by 100 (volume %); concentration of a solution is expressed as a ratio of the weight of the solute (in grams) to the total volume (in mL) of the solution, multiplied by 100 (w/v %). Yield of a reaction is expressed as a ratio of the weight of the product in question to the theoretical yield of this product, multiplied by 100(%). Composition of a mixed solvent is expressed as a proportion of volume parts of the component solvents (e.g., 3:1).


YKL-40 Assay

Full details of the assay were described in the Polish patent application No. P.440558, filed on Mar. 4, 2022, and in the U.S. provisional patent application No. 63/316,477, filed on Mar. 4, 2022. The specific biotinylated compound used in the above-mentioned patent applications for this assay, “Example 4”, was ((2R,5S)-5-(4-chlorobenzyl)-4-(1-(pyridin-2-yl)piperidin-4-yl)morpholin-2-yl)methyl 41-oxo-45-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-4,7,10,13,16,19,22,25,28,31,34,37-dodecaoxa-40-azapentatetracontanoate. Apart from this state-of-the-art compound “Example 4”, the biotinylated compound Example 14 from this patent application was also alternatively used as an assay probe.


The standard experimental protocol for YKL-40 indirect binding AlphaScreen assay comprised of 5 step additions to wells of 96 well plate: (i) in the 1st step, 8 μL of the biotinylated compound (“Example 4”) was added at 5×15 nM concentration in MST buffer containing 1% DMSO; (ii) in the 2nd step, 8 μL of a small molecule compound inhibitor was added at 5×1 μM or 5×100 μM concentration in MST buffer containing 2% DMSO; (iii) in the 3rd step, 8 μL of YKL-40-histag was added at concentration 5×5 nM in MST buffer containing 0% DMSO, after which the plate was spun down for 2 minutes at 1500 g and incubated 1 h at 37° C.; (iv) in the 4th step, 8 μL of Nickel Chelate Acceptor beads were added at concentration 5×10 μg/mL in MST buffer containing 1% DMSO, after which the plate was spun down for 1 minute at 250 g and incubated in dark for 1 hour at RT; (v) in the 5th step, 8 μL of Streptavidin Donor beads were added at concentration 5×10 μg/mL in MST buffer containing 1% DMSO, after which the plate was spun down for 1 minute at 250 g and incubated in dark for 1 hour at RT. Positive and negative control wells received just 8 μL of MST buffer containing 2% DMSO without inhibitor in the 2nd step (ii). In addition, negative control received just 8 μL of MST buffer containing 0% DMSO without YKL-40-histag in the 3rd step (iii). Finally, the plate luminescence was excited at 680 nm and the emission read at 520-620 nm using the AlphaScreen module in the Spark™ 10 M multimode microplate reader (Tecan Trading AG, Männedorf, Switzerland). Inhibition percentage and IC50 values were determined using GraphPad Prism 7.0 software (GraphPad Software, San Diego, CA, USA).


Orthogonal YKL-40 AlphaScreen Assay Using Biotinylated Asialofetuin (AF-Biot) in Place of Example 14.


During our studies we found out that YKL-40, being a lectin, can bind oligosaccharides other than oligomers of chitin. Specifically the screening method based on interaction of YKL-40 with heparan sulfate and influence of small molecule binders of YKL-40 on this interaction was described in the Polish patent application No. P.440558, filed on Mar. 4, 2022, in the U.S. provisional patent application No. 63/316,477, filed on Mar. 4, 2022, and in the U.S. patent application Ser. No. 18/117,193, filed on Mar. 3, 2023. During more extensive search for other oligosaccharides we found out that immobilized YKL40-his on NiNTA BLITz sensors specifically interacts with biotinylated asialofetuin AF-biot (branched triantennary LacNac) with Kd=2.6 nM in PBS buffer containing 1 mg/ml BSA and 0.05% Tween20, while AF-biot does not interact with bare NiNTA BLITz sensor or same sensor covered with non-YKL40 his-tagged protein. Based on these findings, an alternative AlphaScreen assay using same biotinylated asialofetuin (AF-biot) in place of Example 14 was established.


The standard experimental protocol for YKL-40 indirect binding AlphaScreen assay comprised of 5 step additions to wells of 96 well plate: (i) in the 1 step, the biotinylated asialofetuin was added at 5×1.6 nM concentration in assay buffer, i.e. PBS containing 1 mg/ml BSA and 0.05% Tween20; (ii) in the 2nd step, 8 μL of a small molecule compound inhibitor was added at starting dose 5×1 μM, 3× serial dilution, 10 data points in the assay buffer containing 2% DMSO; (iii) in the 3rd step, 8 μL of YKL-40-histag was added at concentration 5×20 nM in the assay buffer containing 0% DMSO, after which the plate was spun down for 2 minutes at 500 g and incubated 1 h at 37° C.; (iv) in the 4th step, 8 μL of Nickel Chelate Acceptor beads were added at concentration 5×10 μg/mL in the assay buffer containing 0% DMSO, after which the plate was spun down for 1 minute at 250 g and incubated in dark for 1 hour at RT; (v) in the 5th step, 8 μL of Streptavidin Donor beads were added at concentration 5×10 μg/mL in the assay buffer containing 0% DMSO, after which the plate was spun down for 1 minute at 250 g and incubated in dark for 1 hour at RT. Positive and negative control wells received just 8 μL of the assay buffer containing 2% DMSO without inhibitor in the 2nd step (ii). In addition, negative control received just 8 μL of the assay buffer containing 0% DMSO without YKL-40-histag in the 3Y step (iii). Finally, the plate luminescence was excited at 680 nm and the emission read at 520-620 nm using the AlphaScreen module in the Spark™ 10 M multimode microplate reader (Tecan Trading AG, Männedorf, Switzerland). Inhibition percentage and IC50 values were determined using GraphPad Prism 7.0 software (GraphPad Software, San Diego, CA, USA). Compounds from Examples 4 and 11 (both of this patent application) can substantially interfere with YKL-40 and asialofetuin (triantennary LacNac) interaction—62% and 64%, with IC50 values 22 nM and 20 nM, respectively.


BLITz system (Sartorius) that used Bio-layer interferometry technology was applied to assess Kd of biotinylated compounds. 10 min before Kd measurement, streptavidin biosensors were dehydrated in the MST buffer. During dehydration process the 10 mM stock of biotinylated compound in DMSO was diluted in MST buffer to final concentration 0.5 μM. In addition, protein stock of recombinant YKL40 (un-tagged, purified in-house) was also prepared in MST buffer at two concentrations: 200 μM and 400 μM. The dehydrated biosensor was applied on the BLITz system, followed by 5-step procedure: 1) initial baseline for 30 s with MST buffer in tube, 2) loading of 4 μl of biotinylated compound on drop, 3) baseline for 30 s with MST buffer in tube, 4) association of 4 μl of protein stock at one concentration or MST buffer (negative control), 5) dissociation with MST buffer in tube. The same multi-step measurement was done twice for each protein stock concentration and once for negative control. Kd was measured by the BLITz software based on global fitting using the negative control as a reference.


The compounds disclosed in Table 1 below have the IC50 values towards YKL-40 generally ranging from about 0.001 μM to about 100 μM. Their ranges of activity have been assigned as follows:

    • A: <0.1 μM;
    • B: 0.1-1 μM;
    • C: 1-10 μM;
    • D: 10-100 μM; and
    • E: >100 μM.


In Table 1, whenever the symbol of range of activity is preceded by asterisk (*), this symbol represents the Kd range rather than the IC50 value range. The ranges of activity have been assigned as follows:

    • *A: <0.1 μM;
    • *B: 0.1-1 μM;
    • *C: 1-10 μM;
    • *D: 10-100 μM; and
    • *E: >100 μM.












TABLE 1







YKL-40,



Ex. #
Structure
IC50/Kd
IUPAC Name


















1.


embedded image

  A single isomer—cis-trans relationship unknown

A
(25,5S)-5-(4-chlorobenzyl)-2- methyl-4-(4-(5-methyloxazol- 2-yl)cyclohexyl)morpholine hydrochloride





2.


embedded image

  A single isomer—cis-trans relationship unknown

A
(25,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-2- methylmorpholine 2,2,2- trifluoroacetate





3.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(5-methyloxazol-2-yl- cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine 2,2,2- trifluoroacetate





4.


embedded image

  A single isomer—trans isomer according to X-Ray

A
(2R,5S)-5-(4-chlorobenzyl)-4- ((1r,4S)-4-(1,5-dimethyl-1H- pyrazol-3-yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine dihydrochloride





5.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-2-(3- methylisoxazol-5- yl)morpholine 2,2,2- trifluoroacetate





6.


embedded image

  A single isomer—cis-trans relationship unknown

A
(7S,9aR)-7-(4-chlorobenzyl)- 8-(4-(5-methyloxazol-2-yl)- cyclohexyl)octahydro- pyrazino[2,1-c][1,4]oxazine 2,2,2-trifluoroacetate





7.


embedded image

  A single isomer—different to Ex. 8; cis-trans relationship unknown

A
(7S,9aR)-7-(4-chlorobenzyl)- 8-(4-(1,5-dimethyl-1H- pyrazol-3-yl)- cyclohexyl)octahydro- pyrazino[2,1-c][1,4]oxazine 2,2,2-trifluoroacetate





8.


embedded image

  A single isomer—different to Ex. 7; cis-trans relationship unknown

A
(7S,9aR)-7-(4-chlorobenzyl)- 8-(4-(1,5-dimethyl-1H- pyrazol-3- yl)cyclohexyl)octahydro- pyrazino[2,1-c][1,4]oxazine 2,2,2-trifluoroacetate





9.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(5-methyloxazol-2-yl)- cyclohexyl)morpholine-2- carboxylic acid 2,2,2- trifluoroacetate





10.


embedded image

  A single isomer—cis-trans relationship unknown

A
methyl (2R,5S)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3-yl)- cyclohexyl)morpholine-2- carboxylate 2,2,2- trifluoroacetate





11.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)morpholine-2- carboxylic acid 2,2,2- trifluoroacetate





12.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-2-(1-( 2,2,2- trifluoroethyl)-1H-1,2,3- triazol-4-yl)morpholine 2,2,2- trifluoroacetate





13.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(5-methyloxazol-2-yl)- cyclohexyl)-2-(1-( 2,2,2- trifluoroethyl)-1H-1,2,3- triazol-4-yl)morpholine 2,2,2- trifluoroacetate





14.


embedded image

  A single isomer—cis-trans relationship unknown

*A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-N-(37-oxo- 41-((3aS,4S,6aR)-2- oxohexahydro-1H-thieno[3,4- d]imidazol-4-yl)- 3,6,9,12,15,18,21,24,27,30,33- undecaoxa-36- azahentetracontyl)morpholine- 2-carboxamide 2,2,2- trifluoroacetate





15.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-1,2,4- triazol-3-yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine 2,2,2- trifluoroacetate





16.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-2- (fluoromethyl)morpholine 2,2,2-trifluoroacetate





17.


embedded image

  A single isomer—cis-trans relationship unknown

A
((2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)morpholin-2- yl)methanol 2,2,2- trifluoroacetate





18.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-N- ethyl-4-(4-(5-methyloxazol-2- yl)cyclohexyl)morpholine-2- carboxamide 2,2,2- trifluoroacetate





19.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-N- ethylmorpholine-2- carboxamide 2,2,2- trifluoroacetate





20.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-N,N- dimethylmorpholine-2- carboxamide 2,2,2- trifluoroacetate





21.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)- N,N-dimethyl-4-(4-(5- methyloxazol-2- yl)cyclohexyl)morpholine-2- carboxamide 2,2,2- trifluoroacetate





22.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-2-(5-methyl- 1,3,4-oxadiazol-2- yl)morpholine 2,2,2- trifluoroacetate





23.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-N′-acetyl-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholine-2- carbohydrazide 2,2,2- trifluoroacetate





24.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-2- (5-methyl-1,3,4-oxadiazol-2- yl)-4-(4-(5-methyloxazol-2- yl)cyclohexyl)morpholine 2,2,2-trifluoroacetate





25.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-N′-acetyl-5-(4- chlorobenzyl)-4-(4-(5- methyloxazol-2- yl)cyclohexyl)morpholine-2- carbohydrazide 2,2,2- trifluoroacetate





26.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4,5-dimethyloxazol-2- yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine hydrochloride





27.


embedded image

  A single isomer—cis-trans relationship unknown

A
N-(((2S,5S)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholin-2- yl)methyl)-1,5-dimethyl-1H- pyrazole-3-carboxamide hydrochloride





28.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2S,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-2- ethynylmorpholine 2,2,2- trifluoroacetate





29.


embedded image

  A single isomer—cis-trans relationship unknown

*A
N-(35-(4-((2R,5S)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholin-2- yl)-1H-1,2,3-triazol-1-yl)- 3,6,9,12,15,18,21,24,27,30,33- undecaoxapentatriacontyl)-5- ((3aS,4S,6aR)-2-oxo- hexahydro-1H-thieno[3,4- d]imidazol-4-yl)pentanamide 2,2,2-trifluoroacetate





30.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-N- isopropylmorpholine-2- carboxamide hydrochloride





31.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-N- methylmorpholine-2- carboxamide dihydrochloride





32.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4,5-dimethyloxazol-2- yl)cyclohexyl)-N,N- dimethylmorpholine-2- carboxamide hydrochloride





33.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4,5-dimethyloxazol-2- yl)cyclohexyl)-N- isopropylmorpholine-2- carboxamide hydrochloride





34.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-N- methyl-4-(4-(5-methyloxazol- 2-yl)cyclohexyl)morpholine-2- carboxamide hydrochloride





35.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4,5-dimethyloxazol-2- yl)cyclohexyl)-N- ethylmorpholine-2- carboxamide hydrochloride





36.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(5-fluoro-1-methyl-1H- pyrazol-3-yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine hydrochloride





37.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4,5-dimethylthiazol-2- yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine hydrochloride





38.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-2- ((methylsulfonyl)methyl)-4-(4- (5-methylthiazol-2-yl)- cyclohexyl)morpholine hydrochloride





39.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4,5-dimethyloxazol-2-yl)- cyclohexyl)morpholine-2- carboxylic acid hydrochloride





40.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4,5-dimethylthiazol-2- yl)cyclohexyl)morpholine-2- carboxylic acid hydrochloride





41.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-2-((prop-2- yn-1-yloxy)methyl)morpholine hydrochloride





42.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4,5-dimethylthiazol-2- yl)cyclohexyl)-N- ethylmorpholine-2- carboxamide hydrochloride





43.


embedded image

  A single isomer—cis-trans relationship unknown

*A
N-(35-(4-((((2R,5S)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3-yl)- cyclohexyl)morpholin-2- yl)methoxy)methyl)-1H-1,2,3- triazol-1-yl)- 3,6,9,12,15,18,21,24,27,30,33- undecaoxapentatriacontyl)-5- ((3aS,4S,6aR)-2-oxo- hexahydro-1H-thieno[3,4- d]imidazol-4-yl)pentanamide hydrochloride





44.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-2-(azidomethyl)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholine hydrochloride





45.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-N-(prop-2- yn-1-yl)morpholine-2- carboxamide hydrochloride





46.


embedded image

  A single isomer—cis-trans relationship unknown

A
N-(((2S,5S)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholin-2- yl)methyl)-1H-pyrazole-3- carboxamide hydrochloride





47.


embedded image

  A single isomer—cis-trans relationship unknown

*A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-N-((1-(37- oxo-41-((3aS,4S,6aR)-2- oxohexahydro-1H-thieno[3,4- d]imidazol-4-yl)- 3,6,9,12,15,18,21,24,27,30,33- undecaoxa-36-aza- hentetracontyl)-1H-1,2,3- triazol-4-yl)methyl)- morpholine-2-carboxamide hydrochloride





48.


embedded image

  A single isomer—cis-trans relationship unknown

A
N-(((2S,5S)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholin-2- yl)methyl)-2H-1,2,3-triazole- 4-carboxamide hydrochloride





49.


embedded image

  A single isomer—cis-trans relationship unknown

A
3-(((2R,5S)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholin-2- yl)methyl)imidazolidine-2,4- dione hydrochloride





50.


embedded image

  A single isomer—cis-trans relationship unknown

B
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-1,2,4- triazol-3-yl)cyclohexyl)-N- ethylmorpholine-2- carboxamide hydrochloride





51.


embedded image

  A single isomer—cis-trans relationship unknown

A
N-(((2S,5S)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholin-2- yl)methyl)-1H-imidazole-4- carboxamide hydrochloride





52.


embedded image

  Probably a mixture of two isomers due to the delicate issue of chirality at cyclohexene ring

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4,5-dimethylthiazol-2- yl)cyclohex-3-en-1-yl)-2- ((methylsulfonyl)methyl)- morpholine 2,2,2- trifluoroacetate





53.


embedded image

  Probably a mixture of two isomers due to the delicate issue of chirality at cyclohexene ring

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4,5-dimethylthiazol-2- yl)cyclohex-3-en-1-yl)-N- ethylmorpholine-2- carboxamide 2,2,2- trifluoroacetate





54.


embedded image

  A single isomer—cis-trans relationship unknown

*A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4,5-dimethylthiazol-2- yl)cyclohexyl)-N-(37-oxo-41- ((3aS,45,6aR)-2-oxo- hexahydro-1H-thieno[3,4- d]imidazol-4-yl)- 3,6,9,12,15,18,21,24,27,30,33- undecaoxa-36-aza- hentetracontyl)morpholine-2- carboxamide hydrochloride





55.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-2-(azidomethyl)-5-(4- chlorobenzyl)-4-(4-(4,5- dimethylthiazol-2- yl)cyclohexyl)morpholine hydrochloride





56.


embedded image

  A single isomer—cis-trans relationship unknown

*A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4,5-dimethylthiazol-2- yl)cyclohexyl)-N-((1-(37-oxo- 41-((3aS,4S,6aR)-2-oxo- hexahydro-1H-thieno[3,4- d]imidazol-4-yl)- 3,6,9,12,15,18,21,24,27,30,33- undecaoxa-36-aza- hentetracontyl)-1H-1,2,3- triazol-4-yl)methyl)- morpholine-2-carboxamide hydrochloride





57.


embedded image

  A single isomer—cis-trans relationship unknown

A
N-(((2S,5S)-5-(4- chlorobenzyl)-4-(4-(4,5- dimethylthiazol-2- yl)cyclohexyl)morpholin-2- yl)methyl)-1H-1,2,3-triazole- 5-carboxamide hydrochloride





58.


embedded image

  A single isomer—cis-trans relationship unknown

A
N-(((2S,5S)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholin-2- yl)methyl)-1-methyl-1H- imidazole-4-carboxamide hydrochloride





59.


embedded image

  A single isomer—cis-trans relationship unknown

A
methyl 2-((2S,5S)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholin-2- yl)acetate hydrochloride





60.


embedded image

  A single isomer—cis-trans relationship unknown

A
2-((2S,5S)-5-(4-chlorobenzyl)- 4-(4-(1,5-dimethyl-1H- pyrazol-3- yl)cyclohexyl)morpholin-2- yl)acetic acid hydrochloride





61.


embedded image

  A single isomer—cis-trans relationship unknown

A
2-((2S,5S)-5-(4-chlorobenzyl)- 4-(4-(1,5-dimethyl-1H- pyrazol-3- yl)cyclohexyl)morpholin-2- yl)-N-ethylacetamide hydrochloride





62.


embedded image

  A single isomer—cis-trans relationship unknown

B
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-1,2,4- triazol-3- yl)cyclohexyl)morpholine-2- carboxylic acid hydrochloride





63.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-bromobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine hydrochloride





64.


embedded image

  A single isomer—cis-trans relationship unknown

A
N-(((2R,5S)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholin-2- yl)methyl)methanesulfonamide hydrochloride





65.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-bromobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)morpholine-2- carboxylic acid hydrochloride





66.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-bromobenzyl)-4- (4-(5-methyloxazol-2- yl)cyclohexyl)morpholine-2- carboxylic acid 2,2,2- trifluoroacetate





67.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-bromobenzyl)-4- (4-(5-methyloxazol-2- yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine 2,2,2- trifluoroacetate





68.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4- bromobenzyl)-4-(4-(1,5- dimethyl-1H-1,2,4-triazol-3- yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine hydrochloride





69.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-bromobenzyl)-4- (4-(1,5-dimethyl-1H-1,2,4- triazol-3-yl)cyclohexyl)- morpholine-2-carboxylic acid hydrochloride





70.


embedded image

  A single isomer—cis-trans relationship unknown

A
N-(((2R,5S)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholin-2- yl)methyl)-1,1,1- trifluoromethanesulfonamide hydrochloride





71.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-bromobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-N- ethylmorpholine-2- carboxamide hydrochloride





72.


embedded image

  A single isomer—cis-trans relationship unknown

B
(2R,5S)-N-(2-(3-(but-3-yn-1- yl)-3H-diazirin-3-yl)ethyl)-5- (4-chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholine-2- carboxamide hydrochloride





73.


embedded image

  A single isomer—cis-trans relationship unknown

A
2-(((2R,5S)-5-(4- chlorobenzyl)-4-(4-(4,5- dimethylthiazol-2- yl)cyclohexyl)morpholin-2- yl)methoxy)acetic acid hydrochloride





74.


embedded image

  A single isomer—cis-trans relationship unknown

A
2-((2R,5S)-5-(4-chlorobenzyl)- 4-(4-(1,5-dimethyl-1H- pyrazol-3- yl)cyclohexyl)morpholin-2- yl)propan-2-ol





75.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-2-(5-methyl- 1H-pyrazol-3-yl)morpholine hydrochloride





76.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5R)-2-(4-chlorobenzyl)-1- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-5- (methylsulfonyl)piperidine hydrochloride





77.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)morpholine-2- carboxamide





78.


embedded image

  A single isomer—cis-trans relationship unknown

A
2-(4-((2R,5R)-2-(4- chlorobenzyl)-5-(methyl- sulfonyl)piperidin-1-yl)- cyclohexyl)-5-methyloxazole hydrochloride





79.


embedded image

  A single isomer—cis-trans relationship unknown (unknown sulfur chirality)

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-2- ((methylsulfinyl)methyl)- morpholine





80.


embedded image

  A single isomer—cis-trans relationship unknown (unknown sulfur chirality)

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-2- ((methylsulfinyl)methyl)- morpholine





81.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-2- ((methylthio)methyl)- morpholine hydrochloride





82.


embedded image

  A single isomer—cis-trans relationship unknown

A
1-(((2R,5S)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholin-2- yl)methyl)pyrrolidine-2,5- dione 2,2,2-trifluoroacetate





83.


embedded image

  A single isomer—cis-trans relationship unknown

A
(25,5S)-5-(4-chlorobenzyl)-4- (4-(4,5-dimethylthiazol-2- yl)cyclohexyl)-2- ethynylmorpholine hydrochloride





84.


embedded image

  A single isomer—cis-trans relationship unknown

A
2-(4-((2R,5S)-5-(4- chlorobenzyl)-4-(4-(4,5- dimethylthiazol-2- yl)cyclohexyl)morpholin-2- yl)-1H-1,2,3-triazol-1-yl)acetic acid hydrochloride





85.


embedded image

  A single isomer—cis-trans relationship unknown

A
3-(4-((2R,5S)-5-(4- chlorobenzyl)-4-(4-(4,5- dimethylthiazol-2- yl)cyclohexyl)morpholin-2- yl)-1H-1,2,3-triazol-1- yl)propanoic acid hydrochloride





86.


embedded image


A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohex-3-en-1-yl)-2- ((methylsulfonyl)methyl)- morpholine hydrochloride





87.


embedded image

  A single isomer—cis-trans relationship unknown

*A
N-(35-(4-((2R,5S)-5-(4- chlorobenzyl)-4-(4-(4,5- dimethylthiazol-2-yl)- cyclohexyl)morpholin-2-yl)- 1H-1,2,3-triazol-1-yl)- 3,6,9,12,15,18,21,24,27,30,33- undecaoxapentatriacontyl)-5- ((3aS,4S,6aR)-2-oxo- hexahydro-1H-thieno[3,4- d]imidazol-4-yl)pentanamide hydrochloride





88.


embedded image

  A single isomer—cis-trans relationship unknown

A
2-((2S,5S)-5-(4-chlorobenzyl)- 4-(4-(4,5-dimethylthiazol-2- yl)cyclohexyl)morpholin-2- yl)acetic acid hydrochloride





89.


embedded image

  A single isomer—cis-trans relationship unknown

A
3-((((2R,5S)-5-(4- chlorobenzyl)-4-(4-(4,5- dimethylthiazol-2- yl)cyclohexyl)morpholin-2- yl)methyl)sulfonyl)propanoic acid hydrochloride





90.


embedded image

  A single isomer—cis-trans relationship unknown

A
3-(((2R,5S)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholin-2- yl)methyl)-6,6-dimethyl-3- azabicyclo[3.1.0]hexane-2,4- dione hydrochloride





91.


embedded image

  A single isomer—cis-trans relationship unknown

A
3-(((2R,5S)-5-(4- chlorobenzyl)-4-(4-(4,5- dimethylthiazol-2- yl)cyclohexyl)morpholin-2- yl)methyl)-6,6-dimethyl-3- azabicyclo[3.1.0]hexane-2,4- dione hydrochloride





92.


embedded image

  A single isomer—cis-trans relationship unknown

A
3-((((2S,5S)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholin-2- yl)methyl)amino)-4- hydroxycyclobut-3-ene-1,2- dione hydrochloride





93.


embedded image

  A single isomer—cis-trans relationship unknown

A
3-amino-4-((((2S,5S)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholin-2- yl)methyl)amino)cyclobut-3- ene-1,2-dione hydrochloride





94.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4,5-dimethylthiazol-2- yl)cyclohexyl)-2- ((ethylsulfonyl)methyl)- morpholine hydrochloride





95.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-2- ((ethylsulfonyl)methyl)- morpholine hydrochloride





96.


embedded image

  A single isomer—cis-trans relationship unknown

A
3-(((2R,5S)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholin-2- yl)methyl)-1- methylimidazolidine-2,4-dione hydrochloride





97.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4,5-dimethylthiazol-2- yl)cyclohexyl)-N-(1-methyl- 1H-imidazol-4-yl)morpholine- 2-carboxamide hydrochloride





98.


embedded image

  A single isomer—cis-trans relationship unknown

A
4-(4-((2R,5S)-5-(4- chlorobenzyl)-4-(4-(4,5- dimethylthiazol-2- yl)cyclohexyl)morpholin-2- yl)-1H-1,2,3-triazol-1- yl)butanoic acid hydrochloride





99.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4,5-dimethylthiazol-2- yl)cyclohexyl)-N- hydroxymorpholine-2- carboxamide hydrochloride





100.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-N- (1,5-dimethyl-1H-pyrazol-3- yl)-4-(4-(4,5-dimethylthiazol- 2-yl)cyclohexyl)morpholine-2- carboxamide hydrochloride





101.


embedded image

  A single isomer—cis-trans relationship unknown

A
5-(4-((2R,5S)-5-(4- chlorobenzyl)-4-(4-(4,5- dimethylthiazol-2- yl)cyclohexyl)morpholin-2- yl)-1H-1,2,3-triazol-1- yl)pentanoic acid hydrochloride





102.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-N- (1,5-dimethyl-1H-pyrazol-3- yl)-4-(4-(1,5-dimethyl-1H- pyrazol-3-yl)cyclohexyl)- morpholine-2-carboxamide hydrochloride





103.


embedded image

  A single isomer—cis-trans relationship unknown

A
N-((2R,5S)-5-(4- chlorobenzyl)-4-(4-(4,5- dimethylthiazol-2- yl)cyclohexyl)morpholine-2- carbonyl)-N-methylglycine hydrochloride





104.


embedded image

  A single isomer—cis-trans relationship unknown

B
(2R,5S)-4-(4-(1-methyl-1H- 1,2,4-triazol-3-yl)cyclohexyl)- 2-((methylsulfonyl)methyl)-5- (4-(trifluoromethyl)benzyl)- morpholine 2,2,2- trifluoroacetate





105.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1-methyl-1H-imidazol-4- yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine 2,2,2- trifluoroacetate





106.


embedded image

  A single isomer—cis-trans relationship unknown

B
(2R,5S)-4-(4-(1-methyl-1H- 1,2,4-triazol-3-yl)cyclohexyl)- 5-(4-(trifluoromethyl)benzyl)- morpholine-2-carboxylic acid 2,2,2-trifluoroacetate





107.


embedded image

  A single isomer—cis-trans relationship unknown

B
(2S,5S)-5-(4-chlorobenzyl)-2- methyl-4-(4-(5-methyl-1H- pyrazol-3- yl)cyclohexyl)morpholine 2,2,2-trifluoroacetate





108.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1-methyl-1H-1,2,4-triazol- 3-yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine 2,2,2- trifluoroacetate





109.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1-methyl-1H-1,2,4-triazol- 3-yl)cyclohexyl)morpholine-2- carboxylic acid 2,2,2- trifluoroacetate





110.


embedded image

  A single isomer—cis-trans relationship unknown

C
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1-methyl-1H-1,2,3-triazol- 4-yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine 2,2,2- trifluoroacetate





111.


embedded image

  A single isomer—cis-trans relationship unknown

C
methyl (2R,5S)-5-(4- chlorobenzyl)-4-(4-(1-methyl- 1H-1,2,3-triazol-4- yl)cyclohexyl)morpholine-2- carboxylate 2,2,2- trifluoroacetate





112.


embedded image

  A single isomer—cis-trans relationship unknown

C
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1-methyl-1H-1,2,3-triazol- 4-yl)cyclohexyl)morpholine-2- carboxylic acid 2,2,2- trifluoroacetate





113.


embedded image

  A single isomer—cis-trans relationship unknown

E
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 4-yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine 2,2,2- trifluoroacetate





114.


embedded image

  A single isomer—cis-trans relationship unknown

B
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1-methyl-1H-pyrazol-3- yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine 2,2,2- trifluoroacetate





115.


embedded image

  A single isomer—cis-trans relationship unknown

B
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(2-methyl-2H-1,2,3-triazol- 4-yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine 2,2,2- trifluoroacetate





116.


embedded image

  A single isomer—cis-trans relationship unknown

B
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(2-methyl-2H-tetrazol-5- yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine 2,2,2- trifluoroacetate





117.


embedded image

  A single isomer—cis-trans relationship unknown

C
(2R,5S)-5-(4-chlorobenzyl)- N,N-dimethyl-4-(4-(5-methyl- 1,3,4-thiadiazol-2- yl)cyclohexyl)morpholine-2- carboxamide 2,2,2- trifluoroacetate





118.


embedded image

  A single isomer—cis-trans relationship unknown

C
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(5-methyl-1,3,4-thiadiazol- 2-yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine hydrochloride





119.


embedded image

  A single isomer—cis-trans relationship unknown

C
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(5-methoxy-1-methyl-1H- 1,2,4-triazol-3-yl)cyclohexyl)- 2-((methylsulfonyl)methyl)- morpholine 2,2,2- trifluoroacetate





120.


embedded image

  A single isomer—cis-trans relationship unknown

C
3-(4-((2R,5S)-5-(4- chlorobenzyl)-2- ((methylsulfonyl)methyl)- morpholino)cyclohexyl)-1- methyl-1H-1,2,4-triazol-5-ol 2,2,2-trifluoroacetate





121.


embedded image

  A single isomer—cis-trans relationship unknown

D
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1-isopropyl-1H-pyrazol-3- yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine 2,2,2- trifluoroacetate





122.


embedded image

  A single isomer—cis-trans relationship unknown

C
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(5-methoxy-1-methyl-1H- 1,2,4-triazol-3-yl)cyclohexyl)- N,N-dimethylmorpholine-2- carboxamide 2,2,2- trifluoroacetate





123.


embedded image

  A single isomer—cis-trans relationship unknown


(2R,5S)-5-(4-chlorobenzyl)-4- (4-(5-hydroxy-1-methyl-1H- 1,2,4-triazol-3-yl)cyclohexyl)- N,N-dimethylmorpholine-2- carboxamide 2,2,2- trifluoroacetate





124.


embedded image

  A single isomer—cis-trans relationship unknown

B
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,2-dimethyl-1H-imidazol- 4-yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine hydrochloride





125.


embedded image

  A single isomer—cis-trans relationship unknown

B
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1-methyl-5- (trifluoromethyl)-1H-pyrazol- 3-yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine hydrochloride





126.


embedded image

  A single isomer—cis-trans relationship unknown

C
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4,5-dimethylthiophen-2- yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine hydrochloride





127.


embedded image

  A single isomer—cis-trans relationship unknown

E
4-(4-((2R,5S)-5-(4- chlorobenzyl)-2- ((methylsulfonyl)methyl)- morpholino)cyclohexyl)-1- methyl-1H-pyrrole-2- carbonitrile hydrochloride





128.


embedded image

  A single isomer—cis-trans relationship unknown

E
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(5-cyano-1-methyl-1H- pyrrol-3- yl)cyclohexyl)morpholine-2- carboxylic acid hydrochloride





129.


embedded image

  A single isomer—cis-trans relationship unknown

B
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1-ethyl-5-methyl-1H- pyrazol-3-yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine hydrochloride





130.


embedded image

  A single isomer—cis-trans relationship unknown

B
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1-ethyl-5-methyl-1H- pyrazol-3- yl)cyclohexyl)morpholine-2- carboxylic acid hydrochloride





131.


embedded image

  A single isomer—cis-trans relationship unknown

C
(2R,5S)-5-(4-chlorobenzyl)-N- ethyl-4-(4-(1-ethyl-5-methyl- 1H-pyrazol-3- yl)cyclohexyl)morpholine-2- carboxamide hydrochloride





132.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4-methyl-1H-pyrazol-1- yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine hydrochloride





133.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4-methyl-1H-pyrazol-1- yl)cyclohexyl)morpholine-2- carboxylic acid hydrochloride





134.


embedded image

  A single isomer—cis-trans relationship unknown

*A
N-(39-((((2R,5S)-5-(4- chlorobenzyl)-4-(4-(4,5- dimethylthiazol-2- yl)cyclohexyl)morpholin-2- yl)methyl)sulfonyl)-37-oxo- 3,6,9,12,15,18,21,24,27,30,33- undecaoxa-36- azanonatriacontyl)-5- ((3aS,4S,6aR)-2- oxohexahydro-1H-thieno[3,4- d]imidazol-4-yl)pentanamide hydrochloride





135.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-N-(1H- tetrazol-5-yl)morpholine-2- carboxamide hydrochloride





136.


embedded image

  A single isomer—cis-trans relationship unknown


(2R,5S)-4-(4-(1H-tetrazol-5- yl)cyclohexyl)-5-(4- chlorobenzyl)-2- ((methylsulfonyl)methyl)- morpholine hydrochloride





137.


embedded image

  A single isomer—cis-trans relationship unknown

E
(2R,5S)-4-(4-(1H-1,2,4-triazol- 1-yl)cyclohexyl)-5-(4- chlorobenzyl)-2- ((methylsulfonyl)methyl)- morpholine hydrochloride





138.


embedded image

  A single isomer—cis-trans relationship unknown

C
(2R,5S)-4-(4-(1H-pyrazol-1- yl)cyclohexyl)-5-(4- chlorobenzyl)-2- ((methylsulfonyl)methyl)- morpholine hydrochloride





139.


embedded image

  A single isomer—cis-trans relationship unknown

B
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(4-fluoro-1H-pyrazol-1- yl)cyclohexyl)-2- ((methylsulfonyl)methyl)- morpholine hydrochloride





140.


embedded image

  A single isomer—cis-trans relationship unknown

A
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(5-fluoro-1-methyl-1H- pyrazol-3- yl)cyclohexyl)morpholine-2- carboxylic acid hydrochloride





141.


embedded image

  A single isomer—cis-trans relationship unknown

A
N1-(1,31- bis(((2R,3R,4R,5R,6R)-3- acetamido-4,5-dihydroxy-6- (hydroxymethyl)tetrahydro- 2H-pyran-2-yl)oxy)-16-((3- ((3-(5-(((2R,3R,4R,5R,6R)-3- acetamido-4,5-dihydroxy-6- (hydroxymethyl)tetrahydro- 2H-pyran-2- yl)oxy)pentanamido)propyl)- amino)-3- oxopropoxy)methyl)- 5,11,21,27-tetraoxo-14,18- dioxa-6,10,22,26- tetraazahentriacontan-16-yl)- N12-(3-(4-(((2R,5S)-5-(4- chlorobenzyl)-4-(4-(1,5- dimethyl-1H-pyrazol-3- yl)cyclohexyl)morpholine-2- carboxamido)methyl)-1H- 1,2,3-triazol-1- yl)propyl)dodecanediamide hydrochloride





142.


embedded image

  A single isomer—cis-trans relationship unknown

B
(2R,5S)-5-(4-chlorobenzyl)-4- (4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)-N-(5-(2-(2,6- dioxopiperidin-3-yl)-3- oxoisoindolin-4- yl)pentyl)morpholine-2- carboxamide hydrochloride









Based on NMR spectra, polarity, and activity in AlphaScreen assay, the instant inventors believe that the majority of most active compounds according to the invention have trans relationship of the 1,4 substituents of the cyclohexane ring.


General Synthetic Procedures
General Procedure Ia
Reduction of α-Amino Acid to the Corresponding Amino Alcohol.



embedded image


To a suspension of amino acid in anhydrous tetrahydrofuran (THF) (3 mL/mmol) borane-dimethylsulfide complex (BH3×DMS; 3 equivalents) was added dropwise at 0° C. (Caution: foaming!) The cooling bath was removed and reaction mixture was refluxed overnight, after which time TLC or LC-MS control indicated completion of the reaction. The mixture was cooled to room temperature and 6 M HCl (8 equivalents with respect to the starting material) was carefully added dropwise (Caution: foaming!) and the mixture was again refluxed for 1.5 hours. The mixture was cooled to room temperature and pH was brought up to 10 with suitable addition of 4 M NaOH. Product was extracted several times with ethyl acetate (AcOEt), extracts were combined, dried over anhydrous MgSO4, filtered and concentrated in vacuo. The crude product was triturated with ethyl ether (Et2O) and filtered off.


General Procedure Ib
Reduction of Morpholin-3-One to Morpholine or Amide to Amine.



embedded image


To the solution of either morpholin-3-one or 2-piperazinone or amide in THF (3 mL/mmol) borane-dimethylsulfide complex (BH3×DMS; 3 equivalents) was added and the reaction mixture was refluxed for 3 hours, after which time the TLC or LC-MS control indicated completed consumption of the starting material. Reaction mixture was cooled to room temperature and 2 M or 6 M HCl was cautiously added (6 equivalents with respect to the starting material). The resulting reaction mixture was refluxed for 2 hours and cooled back to room temperature. The pH of the solution was then adjusted to strongly alkaline (˜10) by a dropwise addition of 6 M NaOH. The organic layer was separated and the aqueous layer was additionally extracted with diethyl ether or AcOEt. The combined organic extracts were then dried over anhydrous MgSO4, filtered and the solvents were evaporated. The crude product obtained was, in most cases, sufficiently pure to be used to the next step without any additional purification.


General Procedure II
Cyclization of α-Haloamide to Morpholin-3-One.



embedded image


To the solution of the α-haloamide (i.e., α-chloro- or α-bromoamide) in THF (10 mL/mmol) 3 equivalents of sodium hydride (NaH) was added in one portion (cooling the solution prior to the addition of NaH may was advisable when working on larger scale) and the reaction mixture was allowed to stir at room temperature for 2 hours. The excess of NaH was then carefully quenched by dropwise addition of brine and then additional volume of brine (equal to the initial volume of THF) was added causing phases separation. The organic layer was separated and the aqueous layer was additionally extracted with diethyl ether. The combined organic extracts were then dried over anhydrous MgSO4, filtered and the solvents were evaporated. The crude product was in most cases sufficiently pure to be used to the next step without any additional purification.


General Procedure III

Amino-Selective Acylation of Amino Alcohol with α-Bromoacid with the Use of an Amide-Forming Reagent.




embedded image


To the solution of α-bromoacid in dichloromethane (7 mL/mmol) diisopropylethylamine (DIPEA, 1 equivalent with respect to the starting α-bromoacid), coupling reagent (1 equivalent; typically TBTU or HATU, but other commonly used coupling reagents may be used as well) and amino alcohol (1 equivalent) were added sequentially and the reaction mixture was stirred for 1.5 hours or overnight at room temperature. After this time TLC control showed completed consumption of the starting materials and the reaction mixture was transferred to the separatory funnel and washed sequentially with 1 M HCl, 1 M NaOH, and brine. The organic phase was dried over anhydrous MgSO4, filtered and evaporated to give the crude product which was further purified by crystallization or by silica-gel column chromatography.


General Procedure IVa

Removal of the Tert-Butoxycarbonyl (Boc-) Group from Amine with HCl.


The N-Boc protected amine was treated with a 4 M solution of HCl (5 mL/mmol of starting material) in an appropriate organic solvent (e.g., AcOEt, 1,4-dioxane, MeOH, DCM) for the time necessary for complete consumption of the starting material (typically 30 minutes—2 hours). The volatiles were then removed in vacuo providing de-protected amine in the form of its hydrochloride salt. The crude product was usually sufficiently pure to be used in the following step, but additional trituration with diethyl ether may help to remove any colored impurities or the crude product was purified by preparative reversed-phase column chromatography to give the corresponding product.


General Procedure IVb

Removal of the Tert-Butoxycarbonyl (Boc-) Group from Amine with TFA.


The N-Boc protected amine was treated with solution of TFA (6 equivalents) in DCM for the time necessary for complete consumption of the starting material (typically 30 minutes—2 hours). The volatiles were then removed in vacuo providing de-protected amine in the form of its TFA salt. The crude product was usually purified by preparative reversed-phase column chromatography to give the corresponding product.


General Procedure V
Activation of Carboxylic Group by Mixed Anhydride Followed by Formation of an Amide.



embedded image


Carboxylic acid was dissolved in dichloromethane (DCM) (3-8 mL/mmol depending on the solubility) and N-methylmorpholine (1.2 equivalent) was added. The solution was cooled to −15° C. and alkyl (typically methyl, ethyl or isobutyl) chloroformate (1.2 equivalent) was added and the mixture was stirred for additional 10 minutes at which time the appropriate amine (neat, 1.2 equivalent) was added. The reaction mixture was allowed to warm to room temperature and was typically stirred overnight, though in the cases of reactive amines the coupling was usually completed within minutes. The crude product was isolated by washing of organic phase (DCM) subsequentially with 1 M HCl, 1 M NaOH, and brine. The organic phase was dried over anhydrous MgSO4, filtered and evaporated to give the crude product which was further purified by crystallization or by silica-gel column chromatography or by preparative reversed-phase column chromatography.


General Procedure VI
Addition of Grignard Reagent to Carbonyl Group.

An appropriate Grignard reagent (3 equivalents) was added dropwise to a solution of carbonyl compound in Et2O or THF (6 mL/mmol) at −40° C. After this reaction was allowed to warm up to room temperature. The reaction progress was monitored by TLC and LC-MS analysis of small aliquots of the crude reaction mixture. When analyses were indicated completion of the reaction, the mixture was poured into saturated solution of NH4Cl. An organic phase was separated, and the aqueous phase was extracted with AcOEt. The combined organic phases were dried over anhydrous MgSO4, concentrated in vacuo. The crude product was purified by silica-gel column chromatography or by flash column chromatography on silica or by preparative reversed-phase column chromatography.


General Procedure VII

Reductive Amination of the Cyclic Ketone with Appropriate Amine.




embedded image


An appropriate amine or an amine hydrochloride (1 equivalent) was dissolved in DCE and acetic acid (AcOH, 2 equivalents) and appropriate ketone (2 equivalents) were added and the mixture was heated at 50° C. for 2 days. The reaction mixture was cooled to room temperature and sodium triacetoxyborohydride (NaBH(OAc)3) (4 equivalents) was then added in one portion and the mixture was stirred overnight at room temperature. After this time a 5% aqueous solution of sodium bicarbonate (NaHCO3) and dichloromethane (DCM) were added and the mixture was stirred for 30 minutes. The layers were separated and the aqueous layer was additionally extracted with dichloromethane. The organic extracts were combined, washed with brine, dried over anhydrous MgSO4, filtered and evaporated to dryness. The crude product was purified by flash column chromatography on silica or by preparative reversed-phase column chromatography.


General procedure VIII


Mesylation of Appropriate Hydroxyl Group.



embedded image


To a cooled to 0° C. solution of alcohol, triethylamine (Et3N, 1.6 equivalent) in DCM (20 mL/mmol) a solution of methanesulfonic anhydride (1.5 equivalent) or methanesulfonic chloride (1.2 equivalent) in DCM (6 mL/mmol) was added dropwise and the mixture was stirred at room temperature for 30 minutes. After this time the reaction was washed with 1 M K2CO3, brine, dried over anhydrous MgSO4, filtered and the solvents were evaporated in vacuo. The crude product was in most cases sufficiently pure to be used to the next step without any additional purification.


General Procedure IX

Formation of Weinreb Amide from Carboxylic Acid and N,O-Dimethylhydroxylamine Hydrochloride with the Use of the Amide Forming Reagent.




embedded image


To a solution of carboxylic acid in dichloromethane (2 mL/mmol), DIPEA (only when TBTU was used) (2.1 equivalents) was added, followed by N,O-dimethylhydroxylamine hydrochloride (1.1 equivalent). TBTU (1.1 equivalent) or CDI (1.1 equivalent) was added to the reaction mixture and reaction was stirred at room temperature for the time necessary for the complete consumption of the starting material (usually 16-24 hours) as judged by TLC or LC-MS. The reaction mixture was diluted with dichloromethane, then washed with 2 M HCl and brine. An organic layer was dried over anhydrous MgSO4, filtered and concentrated in vacuo. Product was purified by silica-gel column chromatography.


General Procedure X
Installation of the Appropriate R-Group on the Primary or Secondary Amine or Alcohol



embedded image


To a solution of the primary or secondary amine or alcohol in DCM (10 mL/mmol) or THF (4 mL/mmol), Et3N (2-4 equivalents) or DIPEA (2 equivalents) followed by DMAP (0.1 equivalent) were added and then N-succinimidyl-N-methyl carbamate (2.5 equivalents) or appropriate carbamoyl chloride (2.5 equivalents) or appropriate isocyanate (1.1-2.5 equivalents) or acid chloride (1-2 equivalents) or appropriate anhydride (1.1-2 equivalents) or MOMCl (2 equivalents) was added at room temperature. The reaction progress was monitored by TLC and LC-MS. When analyses indicated completion of the reaction, the mixture was quenched by addition of 4 M NaOH or 5% NaHCO3 (for tosylation). Product was extracted with DCM (2 times). The combined organic solutions were dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was purified by preparative reversed-phase column chromatography or by silica-gel column chromatography.


General Procedure XI

Biotynylation (with BiotinPEG11NH2) of Appropriate Carboxylic Acid.




embedded image


To the solution of appropriate carboxylic acid (1-1.2 equivalent) in DMF or DCM (20 mL/mmol), Et3N (6 equivalents) or DIPEA (6 equivalents) and BiotinPEG11NH2 (1-2 equivalents) were added followed by addition of HATU (1.1 equivalent) or EDCI hydrochloride (1 equivalent) and the reaction mixture was stirred overnight at room temperature. After this time LC-MS control showed complete consumption of the starting materials and the reaction mixture was evaporated to dryness and the crude product was further purified by preparative reversed-phase column chromatography.


General Procedure XII
Fluorination of Alcohols and Carbonyl Compounds



embedded image


To a cooled to −20° or −70 solution of alcohol or aldehyde in dry THF or DCM (6 mL/mmol) DAST (2-2.5 equivalents) was added dropwise and the reaction was allowed to warm to room temperature and stirring was continued overnight. The reaction was poured into 5% NaHCO3 and the resulting mixture was stirred for 15 minutes. The organic phase was separated and an aqueous one was extracted with DCM, washed with brine, dried over anhydrous MgSO4, filtered and concentrated in vacuo. The crude product was purified by silica-gel column chromatography or by flash column chromatography on silica or by preparative reversed-phase column chromatography.


Exemplary Synthetic Procedures
Example 1
Synthesis of (2S,5S)-5-(4-chlorobenzyl)-2-methyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine hydrochloride (1)



embedded image


Step 1.
Synthesis of (2S)-2-amino-3-(4-chlorphenyl)propan-1-ol (1a)



embedded image


The title compound (1a) was obtained from optically pure L-p-chlorophenylalanine ((2S)-2-amino-3-(4-chlorophenyl)propanoic acid) (10.67 g; 53.45 mmol) according to the General Procedure Ia in 87% yield (8.61 g; 46.50 mmol).


ESI-MS m/z for C9H12ClNO found 185.7/187.7 [M+H]+; Rt=0.50 min (Method F); 1H NMR (500 MHz, CDCl3) δ 7.28 (d, J=8.3 Hz, 2H), 7.11 (d, J=8.3 Hz, 2H), 3.62 (dd, J=10.6, 3.8 Hz, 1H), 3.37 (dd, J=10.5, 6.9 Hz, 1H), 3.11-3.07 (brs, 1H), 2.76 (dd, J=13.6, 5.4 Hz, 1H), 2.50 (dd, J=13.6, 8.6 Hz, 1H).


Step 2.
Synthesis of (R)-2-bromo-N—((S)-1-(4-chlorophenyl)-3-hydroxypropan-2-yl)propanamide (1b)



embedded image


(2S)-2-amino-3-(4-chlorophenyl)propan-1-ol (1a) (1 g; 5.38 mmol) was coupled with (R)-2-bromopropanoic acid according to the General Procedure III using TBTU as an amide bond forming reagent. The title compound 1b was obtained as a white solid in 64% yield (1.1 g; 3.44 mmol).


ESI-MS m/z for C12H16BrClNO2 found 320.7/322.7 [M+H]+; Rt=2.49 min (Method F); 1H NMR (500 MHz, CDCl3) δ 7.25 (d, J=8.2 Hz, 2H), 7.14 (d, J=8.2 Hz, 2H), 6.58 (d, J=6.7 Hz, 1H), 4.33 (dd, J=14.1, 7.1 Hz, 1H), 4.13-4.06 (m, 1H), 3.69-3.64 (m, 1H), 3.61-3.57 (m, 1H), 2.90-2.81 (m, 2H), 2.17 (brs, 1H), 1.82 (d, J=6.9 Hz, 3H).


Step 3.
Synthesis of (2S,5S)-5-(4-chlorobenzyl)-2-methylmorpholin-3-one (1c)



embedded image


The title compound (1c) was obtained from 1b (1.1 g; 3.44 mmol) according to the General Procedure II in 79% yield (650 mg; 2.72 mmol).


ESI-MS m/z for C12H15ClNO2 found 240.1/242.1 [M+H]+; Rt=2.45 min (Method F); 1H NMR (500 MHz, CDCl3) δ 7.27 (d, J=8.2 Hz, 2H), 7.10 (d. J=8.2 Hz, 2H), 6.13 (brs, 1H), 4.18 (m, 1H), 3.76 (d, J=3.0 Hz, 2H), 3.55-3.50 (m, 1H), 2.91-2.83 (m, 2H), 1.46 (d, J=6.9 Hz, 3H).


Step 4.
Synthesis of (2S,5S)-5-(4-chlorobenzyl)-2-methylmorpholine (1d)



embedded image


The title compound (1d) was obtained from 1c (1.1 g; 4.6 mmol) according to the General Procedure Ib in 90% yield (932 mg; 4.14 mmol).


ESI-MS m/z for C12H17ClNO found 226.4/228.4 [M+H]+; Rt=1.30 min (Method F); 1H NMR (500 MHz, DMSO-d6+D2O) δ 7.33 (d, J=8.3 Hz, 2H), 7.23 (d, J=8.3 Hz, 2H), 3.50-3.48 (m, 1H), 3.49-3.47 (m, 2H), 2.87-2.81 (m, 1H), 2.80-2.76 (m, 2H), 2.65 (dd, J=12.4 Hz, 8.3 Hz, 1H), 2.58 (dd, J=12.4 Hz, 3.0 Hz, 1H), 1.09 (d, J=6.2 Hz, 3H).


Step 5.
Synthesis of N-(prop-2-yn-1-yl)-1,4-dioxaspiro[4.5]decane-8-carboxamide (1e)



embedded image


The title compound (1e) was obtained from 1,4-dioxaspiro[4.5]decane-8-carboxylic acid (560 mg; 3.00 mmol) and prop-2-yn-1-amine (288 μL; 4.50 mmol) according to the General Procedure V in 84% yield (560 mg; 2.51 mmol).


ESI-MS m/z for C12H18NO3 found 224.0 [M+H]+; Rt=0.49 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 3.86-3.83 (m, 4H), 3.82-3.78 (m, 2H), 2.98 (s, 1H), 2.16-2.11 (m, 1H), 1.69-1.62 (m, 4H), 1.59-1.52 (m, 2H), 1.46-1.37 (m, 2H).


Step 6.
Synthesis of 5-methyl-2-(1,4-dioxaspiro[4.5]decan-8-yl)oxazole (1f)



embedded image


To the solution of 1e (400 mg; 1.80 mmol) in DCM (8 mL) placed into a reactor AuCl3 (27 mg; 0.09 mmol) was added and whole was stirred at room temperature for 2 days. The reaction progress was monitored by LC-MS analysis. After analytical control indicated completion of the reaction, a whole was concentrated in vacuo. The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 10:1 to 2:1, v/v). Compound if was obtained in 87% yield (349 mg; 1.56 mmol).


ESI-MS m/z for C12H18NO3 found 224.0 [M+H]+; Rt=0.93 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 6.66 (q, J=1.3 Hz, 1H), 3.84 (s, 4H), 2.81-2.76 (m, 1H), 2.22 (d, J=1.3 Hz, 3H), 1.93-1.88 (m, 2H), 1.71-1.65 (m, 4H), 1.59-1.52 (m, 2H).


Step 7.
Synthesis of 4-(5-methyloxazol-2-yl)cyclohexan-1-one (1g)



embedded image


The solution of if (346 mg; 1.55 mmol) in 1 N HCl (10 mL) was stirred at room temperature for 2 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, to this mixture aqueous solution of NaOH (4 M) was added to pH 12 and then extracted with DCM (2×). The combined organic solutions were dried over anhydrous MgSO4, filtered and concentrated in vacuo. The crude product was used to the next step without additional purification. Compound 1g was obtained in 96% yield (267 mg; 1.49 mmol).


ESI-MS m/z for C10H14NO2 found 180.0 [M+H]+; Rt=0.71 min (Method A); 1H NMR (700 MHz, DMSO-d6+H2O, 348 K) δ 6.68 (q, J=1.3 Hz, 1H), 3.26-3.21 (m, 1H), 2.49-2.43 (m, 2H), 2.36-2.29 (m, 2H), 2.27-2.21 (m, 5H), 1.98-1.91 (m, 2H).


Step 8.
Synthesis of (2S,5S)-5-(4-chlorobenzyl)-2-methyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine hydrochloride (1)



embedded image


The title compound (1) was obtained as a single isomer as a hydrochloride salt from 1d (270 mg; 1.20 mmol) and ketone 1g (163 mg; 0.90 mmol) according to the General Procedure VII in 14% yield (57 mg; 0.13 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 4:1 to 1:2, v/v) and then transformed into hydrochloride salt with using HCl/AcOEt.


ESI-MS m/z for C22H30ClN2O2 found 389.3/391.3 [M+H]+; Rt=0.99 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.5 Hz, 2H), 7.32 (d, JBB′=8.5 Hz, 2H), 6.66 (q, J=1.2 Hz, 1H), 3.90-3.89 (m, 1H), 3.79-3.77 (m, 1H), 3.72-3.70 (m, 1H), 3.59-3.57 (m, 1H), 3.47-3.42 (m, 2H), 3.16-3.10 (m, 2H), 3.01-2.99 (m, 1H), 2.73 (tt, J=11.8 Hz, J=3.6 Hz, 1H), 2.34-2.31 (m, 1H), 2.28-2.26 (m, 1H), 2.23 (d, J=1.2 Hz, 3H), 2.19-2.15 (m, 2H), 1.65-1.52 (m, 4H), 1.22 (d, J=6.3 Hz, 3H).


Example 2
Synthesis of (2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-methylmorpholine 2,2,2-trifluoroacetate (2)



embedded image


Step 1.
Synthesis of N-methoxy-N-methyl-1,4-dioxaspiro[4.5]decane-8-carboxamide (2a)



embedded image


The title compound (2a) was obtained from 1,4-dioxaspiro[4.5]decane-8-carboxylic acid (6 g; 32.2 mmol) according to the General Procedure IX in 99% yield (7.31 g; 31.9 mmol).


ESI-MS m/z for C11H20NO4 found 230.1 [M+H]+; Rt=0.67 min (Method A)


Step 2.
Synthesis of 1-(1,4-dioxaspiro[4.5]decan-8-yl)but-2-yn-1-one (2b)



embedded image


The title compound (2b) was obtained from 2a (690 mg; 3.00 mmol) and from prop-1-yn-1-ylmagnesium bromide (0.5 M; 18 mL; 9.00 mmol) according to the General Procedure VI in 99% yield (618 mg; 2.97 mmol).


ESI-MS m/z for C12H17O3 found 209.0 [M+H]+; Rt=0.98 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 3.85-3.83 (m, 4H), 2.44-2.38 (m, 1H), 2.03 (s, 3H), 1.89-1.83 (m, 2H), 1.68-1.58 (m, 4H), 1.54-1.48 (m, 2H).


Step 3.
Synthesis of 1,5-dimethyl-3-(1,4-dioxaspiro[4.5]decan-8-yl)-1H-pyrazole (2c)



embedded image


To the suspension of 2b (208 mg; 1.00 mmol) in MeCN (2 mL) methylhydrazine (105 μL; 2.00 mmol) was added and the reaction mixture was stirred at room temperature for 1 hour. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the whole was concentrated in vacuo and the crude product was used to the next step without additional purification. Compound 2c was obtained in 99% yield (234 mg; 0.99 mmol).


ESI-MS m/z for C13H21N2O2 found 237.0 [M+H]+; Rt=0.81 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 5.79 (s, 1H), 3.87 (s, 4H), 3.63 (s, 3H), 2.57-2.51 (m, 1H), 2.19 (s, 3H), 1.87-1.80 (m, 2H), 1.75-1.69 (m, 2H), 1.68-1.60 (m, 2H), 1.58-1.51 (m, 2H).


Step 4.
Synthesis of 4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexan-1-one (2d)



embedded image


The solution of 2c (234 mg; 0.99 mmol) in 1 N HCl (10 mL) was stirred at room temperature for 2 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, to this mixture aqueous solution of NaOH (4 M) was added to pH 13 and then extracted with DCM (2×). The combined organic solutions were dried over anhydrous MgSO4, filtered and concentrated in vacuo. The crude product was used to the next step without additional purification. Compound 2d was obtained in 99% yield (188 mg; 0.98 mmol).


ESI-MS m/z for C11H17N2O found 193.1 [M+H]+; Rt=0.71 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 5.89 (s, 1H), 3.60 (s, 3H), 3.00-2.93 (m, 1H), 2.50-2.44 (m, 2H), 2.27-2.21 (m, 2H), 2.16 (s, 3H), 2.14-2.08 (m, 2H), 1.79-1.70 (m, 2H).


Step 5.
Synthesis of (2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-methylmorpholine 2,2,2-trifluoroacetate (2)



embedded image


The title compound (2) was obtained as a single isomer as a TFA salt from 1d (228 mg; 1.02 mmol) and from ketone 2d (177 mg; 0.92 mmol) according to the General Procedure VII in 10% yield (47 mg; 0.09 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 2:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 20:1, v/v) and then purified by preparative reversed-phase column chromatography (C-18, water/MeCN+1‰ TFA, 90:10 to 10:90, 30 min, 20 mL/min).


ESI-MS m/z for C23H33ClN3O found 402.4/404.4 [M+H]+; Rt=0.94 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.39 (d, JAA′=8.5 Hz, 2H), 7.31 (d, JBB′=8.5 Hz, 2H), 5.84 (s, 1H), 3.86-3.84 (m, 1H), 3.74-3.70 (m, 2H), 3.61-3.59 (m, 4H), 3.49-3.39 (m, 3H), 3.18-3.14 (m, 1H), 3.09 (dd, J=13.8 Hz, J=4.0 Hz, 1H), 3.04-3.00 (m, 1H), 2.31-2.28 (m, 1H), 2.26-2.23 (m, 1H), 2.17 (s, 3H), 2.06-2.02 (m, 2H), 1.55-1.48 (m, 2H), 1.47-1.40 (m, 2H), 1.22 (d, J=6.2 Hz, 3H).


Example 3
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine 2,2,2-trifluoroacetate (3)



embedded image


Step 1.
Synthesis of (R)-2-bromo-3-(tert-butoxy)-N—((S)-1-(4-chlorophenyl)-3-hydroxypropan-2-yl)propanamide (3a)



embedded image


(2S)-2-amino-3-(4-chlorophenyl)propan-1-ol (1a) (1.89 g; 10.2 mmol) was coupled with (R)-2-bromo-3-(tert-butoxy)propanoic acid (3a′) (2.28 g; 10.2 mmol) according to the General Procedure III using TBTU as an amide bond forming reagent. The title compound 3a was obtained in 70% yield (2.96 g; 7.14 mmol).


ESI-MS m/z for C16H23BrClNO3Na found 414.3/416.3 [M+Na]+; Rt=1.43 min (Method A)


Step 2.
Synthesis of (R)-2-bromo-3-(tert-butoxy)propanoic acid (3a′)



embedded image


To a solution of O-(tert-butyl)-D-serine (26.8 g; 166 mmol) and KBr (69.2 g; 582 mmol) in water (134 mL) HBr (48%; 45 mL) was added at room temperature. Then the mixture was cooled to −10° C. and a solution of NaNO2 (13.8 g; 200 mmol) in water (20 mL) was added by syringe pump over the period of 2 hours keeping the temperature below −5° C. Then the mixture was stirred for 2 hours at 0° C. The yellow foam appeared. To this mixture, a 10% solution of Na2SO3 in water (50 mL) was added (to remove an excess of bromine). The phases were separated and an aqueous one was extracted with MTBE (4×250 mL). The combined organic phases were dried over anhydrous Na2SO4, filtered, and concentrated in vacuo and the crude bromoacid was then dissolved in a solution of 1 M NaOH (340 mL) and extracted with MTBE (100 mL). To the aqueous phase, a solution of 1 M HCl (340 mL) was added (the white precipitate appeared) and the whole was transferred to the separatory funnel and extracted with MTBE (4×500 mL). The combined organic phases were dried over anhydrous Na2SO4, filtered, and concentrated in vacuo and the obtained product was used to the next step without additional purification. Compound 3a′ was obtained in 95% yield (35.6 g; 158 mmol).


ESI-MS m/z for C7H12BrO3 found 223.0/225.0 [M−H]; Rt=1.01 min (Method A)


Step 3.
Synthesis of (2S,5S)-2-(tert-butoxymethyl)-5-(4-chlorobenzyl)morpholin-3-one (3b)



embedded image


The title compound (3b) was obtained from 3a (1.2 g, 3.05 mmol) according to the General Procedure II in 89% yield (0.85 g; 2.71 mmol).


ESI-MS m/z for C16H22ClNO3Na found 334.1/336.1 [M+Na]+; Rt=1.39 min (Method A)


Step 4.
Synthesis of ((2R,5S)-5-(4-chlorobenzyl)morpholin-2-yl)methanol (3c)



embedded image


The title compound (3c) was obtained from 3b (0.85 g; 2.71 mmol) according to the General Procedure Ib in 74% yield (0.48 g; 2.01 mmol).


ESI-MS m/z for C12H17ClNO2 found 242.2/244.2 [M+H]+; Rt=0.93 min (Method B)


Step 5.
Synthesis of tert-butyl (2R,5S)-5-(4-chlorobenzyl)-2-(hydroxymethyl)morpholine-4-carboxylate (3d)



embedded image


To a solution of amino alcohol 3c (2.87 g; 11.9 mmol) in dichloromethane (110 mL), di-tert-butyl dicarbonate (Boc2O) (2.46 g; 11.3 mmol) was added and the reaction mixture was stirred at room temperature for 2 hours, after which time TLC showed almost complete consumption of the starting material. Volatiles were removed in vacuo and the residue was purified by column chromatography (hexane/AcOEt, 1:1 v/v). Compound 3d was obtained as a colorless oil in 77% yield (3.14 g; 9.16 mmol).


ESI-MS m/z for C12H17ClNO2 found 242.1/246.1 [M+H−Boc]+; Rt=1.41 min (Method A)


Step 6.
Synthesis of tert-butyl (2R,5S)-5-(4-chlorobenzyl)-2-(((methylsulfonyl)oxy)methyl)morpholine-4-carboxylate (3e)



embedded image


The title compound (3e) was obtained from 3d (16.9 g; 49.44 mmol) according to the General Procedure VIII in 99% yield (20.52 g; 48.95 mmol).


ESI-MS m/z for C23H31ClN3O4S found 420.1/422.1 [M+H]+: Rt=1.60 min (Method A)


Step 7.
Synthesis of (2R,5S)-tert-butyl 5-(4-chlorobenzyl)-2-((methylthio)methyl)morpholine-4-carboxylate (3f)



embedded image


To a solution of 3e (1.3 g; 3.09 mmol) in acetonitrile (15 mL) MeSNa (550 mg; 7.74 mmol) was added and the reaction mixture was stirred at 80° C. overnight. The reaction progress was monitored by LC-MS analysis. After analytical control indicated completion of the reaction, the mixture was dissolved in DCM and then washed with 2 M NaOH and brine, dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was used to the next step without additional purification. Compound 3f was obtained in 79% yield (0.9 g; 2.43 mmol).


ESI-MS m/z for C18H27ClNO3S found 372.1/374.1 [M+H]+: Rt=1.89 min (Method A)


Step 8.
Synthesis of (2R,5S)-tert-butyl 5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)morpholine-4-carboxylate (3)



embedded image


The reaction of 3f (0.32 g; 0.86 mmol) with peracetic acid (35% in AcOH; 0.33 mL; 1.72 mmol) was stirred at room temperature overnight. Then the reaction was concentrated in vacuo and the residue was transferred to AcOEt/1 M NaOH. Phases were separated and the organic one was washed with brine, dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was used to the next step without additional purification. Compound 3g was obtained in 99% yield (0.34 g; 0.85 mmol).


ESI-MS m/z for C18H27ClNO5S found 404.1/406.1 [M+H]+; Rt=1.55 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.33-7.26 (m, 2H), 7.24-7.18 (m, 2H), 4.11-4.05 (m, 1H), 3.87-3.82 (m, 1H), 3.81-3.77 (m, 1H), 3.77-3.73 (m, 1H), 3.61-3.54 (m, 1H), 3.34-3.29 (m, 1H), 3.06-2.94 (m, 5H), 2.86-2.79 (m, 1H), 1.98-1.95 (m, 1H), 1.27-1.14 (m, 9H).


Step 9.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (3h)



embedded image


The title compound (3h) was obtained as a hydrochloride salt from 3g (16.5 g; 40.84 mmol) according to the General Procedure IVa in 99% yield (13.73 g; 40.43 mmol).


ESI-MS m/z for C13H18ClNO3S found 303.8/305.8 [M+H]+; Rt=0.51 min (Method A)


Step 10.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine 2,2,2-trifluoroacetate (3)



embedded image


The title compound (3) was obtained as a single isomer as a TFA salt from 3h (170 mg; 0.50 mmol) and ketone 1g (104 mg; 0.58 mmol) according to the General Procedure VII in 24% yield (72 mg; 0.12 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 5:1 to 2:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 120:1, v/v) and then purified by preparative reversed-phase column chromatography (C-18, water/MeCN+1‰ TFA, 90:10 to 10:90.30 min, 20 mL/min).


ESI-MS m/z for C23H32ClN2O4S found 467.3/469.3 [M+H]+; Rt=0.95 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.5 Hz, 2H), 7.31 (d, JBB′=8.5 Hz, 2H), 6.65 (q, J=1.2 Hz, 1H), 4.26-4.23 (m, 1H), 3.76-3.73 (m, 2H), 3.67-3.65 (m, 1H), 3.58 (dd, J=15.0 Hz, J=7.9 Hz, 1H), 3.53-3.51 (m, 1H), 3.48-3.46 (m, 1H), 3.40-3.37 (m, 1H), 3.27-3.24 (m, 1H), 3.17 (dd, J=13.8 Hz, J=10.9 Hz, 1H), 3.09 (dd, J=13.6 Hz, J=4.0 Hz, 1H), 3.02 (s, 3H), 2.73 (tt, J=11.9 Hz, J=3.5 Hz, 1H), 2.27-2.25 (m, 2H), 2.22 (d, J=1.2 Hz, 3H), 2.18-2.15 (m, 2H), 1.60-1.49 (m, 4H).


Example 4
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-((1r,4S)-4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine dihydrochloride (4)



embedded image


The title compound (4) was obtained as a single isomer as a dihydrochloride salt from 3h (168 mg; 0.49 mmol) and ketone 2d (90 mg; 0.47 mmol) according to the General Procedure VII in 17% yield (44 mg; 0.08 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 5:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 20:1, v/v) and then transformed into dihydrochloride salt with using HCl/AcOEt. The obtained solid of the dihydrochloride salt was then lyophilized.


ESI-MS m/z for C24H35ClN3O3S found 480.4/482.4 [M+H]+; Rt=1.00 min (Method A); 1H NMR (400 MHz, D2O) δ 7.30-7.25 (m, 2H), 7.20-7.14 (m, 2H), 6.19 (s, 1H), 4.32-4.19 (m, 1H), 3.85-3.70 (m, 5H), 3.70-3.54 (m, 3H), 3.51-3.35 (m, 2H), 3.33-3.23 (m, 1H), 3.19-3.00 (m, 5H), 2.76-2.63 (m, 1H), 2.38-2.27 (m, 2H), 2.21 (s, 3H), 2.13-2.05 (m, 2H), 1.61-1.37 (m, 4H).


Example 5
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(3-methylisoxazol-5-yl)morpholine 2,2,2-trifluoroacetate (5)



embedded image


Step 1.
Synthesis of (2R,5S)-4-(tert-butoxycarbonyl)-5-(4-chlorobenzyl)morpholine-2-carboxylic acid (5a)



embedded image


To a solution of alcohol 3d (1.8 g; 5.26 mmol) in acetone (40 mL) at 0° C., Jones reagent (12 mL; 2.6 M) was added dropwise. The reaction mixture was stirred at 0° C. for 1 hour, and then isopropanol (iPrOH) (5 mL) was added. After 10 minutes ethyl acetate (150 mL) was added and the mixture was filtered through a pad of Celite. The filtrate was washed with brine, dried over anhydrous MgSO4 and evaporated affording the title compound 5a as a white foam in 91% yield (1.7 g; 4.79 mmol).


ESI-MS m/z for C17H22ClNO5Na found 378.3/380.3 [M+Na]+, 256.1/258.1 [M+H−Boc]+; Rt=1.45 min (Method A)


Step 2.
Synthesis of (2R,5S)-tert-butyl 5-(4-chlorobenzyl)-2-(methoxy(methyl)carbamoyl)morpholine-4-carboxylate (5b)



embedded image


The title compound (5b) was obtained as a white crystal from 5a (0.5 g; 1.4 mmol) according to the General Procedure IX in 67% yield (0.37 g; 0.94 mmol).


ESI-MS m/z for C14H20ClN2O3 found 298.9/300.9 [M+H−Boc]+; Rt=1.51 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.33-7.25 (m, 2H), 7.25-7.15 (m, 2H), 4.26-4.21 (m, 1H), 4.13-4.04 (m, 1H), 3.91-3.83 (m, 1H), 3.78-3.74 (m, 1H), 3.69 (s, 3H), 3.62-3.57 (m, 1H), 3.18-3.10 (m, 4H), 3.01-2.91 (m, 1H), 2.88-2.81 (m, 1H), 1.21 (s, 9H).


Step 3.
Synthesis of (2R,5S)-tert-butyl 5-(4-chlorobenzyl)-2-(3-methylisoxazol-5-yl)morpholine-4-carboxylate (5c)



embedded image


To a cooled to 0° C. solution of an oxime (55 mg; 074 mmol) in THF (1.5 mL) a solution of n-butyllithium (2.5 M in hexanes; 0.6 mL; 1.49 mmol) was added dropwise and the mixture was stirred for 30 minutes. Then the solution of 5b (246 mg; 0.62 mmol) in THF (3 mL) was added dropwise over 15 minutes. After 30 minutes to this mixture a solution of H2SO4 (0.13 mL) in THF/water (1.5 mL/0.4 mL) was added and the resulting mixture was refluxed for 1 hour. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the reaction mixture was cooled to 0° C. and neutralized with 5% NaHCO3. Then water was added and the mixture was extracted with Et2O. An organic layer was then dried over anhydrous MgSO4, filtered and concentrated in vacuo and the crude product was used to the next step without additional purification. Compound 5c was obtained in 97% yield (235 mg; 0.6 mmol).


ESI-MS m/z for C20H26ClN2O4 found 393.2/395.2 [M+H]+; Rt=1.47 min (Method A)


Step 4.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-2-(3-methylisoxazol-5-yl)morpholine hydrochloride (5d)



embedded image


The title compound (5d) was obtained as a hydrochloride salt from 5c (235 mg; 0.6 mmol) according to the General Procedure IVa in 99% yield (194 mg; 0.59 mmol).


ESI-MS m/z for C12H16ClN4O found 293.1/295.1 [M+H]+; Rt=0.76 min (Method A)


Step 5.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(3-methylisoxazol-5-yl)morpholine 2,2,2-trifluoroacetate 5



embedded image


The title compound (5) was obtained as a single isomer as a TFA salt from 5d (181 mg; 0.550 mmol) and ketone 2d (100 mg; 0.520 mmol) according to the General Procedure VII in 10% yield (30 mg; 0.055 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 10:1, v/v, then AcOEt, 100%) and then transformed into TFA salt with using TFA/DCM and then purified by preparative reversed-phase column chromatography (C-18, water/MeCN+1‰ TFA, 90:10 to 20:80.30 min, 20 mL/min).


ESI-MS m/z for C26H34ClN4O2 found 469.3/471.3 [M+H]+; Rt=1.10 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.5 Hz, 2H), 7.31 (d, JBB′=8.5 Hz, 2H), 6.53 (s, 1H), 5.81 (s, 1H), 5.05-5.00 (m, 1H), 3.87-3.86 (m, 1H), 3.80-3.78 (m, 1H), 3.73-3.71 (m, 1H), 3.68-3.66 (m, 1H), 3.60 (s, 3H), 3.53-3.49 (m, 1H), 3.45-3.43 (m, 1H), 3.16-3.14 (m, 2H), 2.49-2.45 (m, 1H), 2.27-2.22 (m, 5H), 2.17 (s, 3H), 2.05-2.02 (m, 2H), 1.56-1.50 (m, 2H), 1.46-1.40 (m, 2H).


Example 6
Synthesis of (7S,9aR)-7-(4-chlorobenzyl)-8-(4-(5-methyloxazol-2-yl)cyclohexyl)octahydropyrazino[2,1-c][1,4]oxazine 2,2,2-trifluoroacetate (6)



embedded image


Step 1.
Synthesis of methyl (S)-4-((R)-2-((tert-butoxycarbonyl)amino)-3-(4-chlorophenyl)propanoyl)-morpholine-3-carboxylate (6a)



embedded image


The title compound (6a) was obtained from methyl (S)-morpholine-3-carboxylate hydrochloride (1.6 g; 8.81 mmol) and Boc-4-chloro-L-phenylalanine (2.64 g; 8.81 mmol) according to the General Procedure III in 77% yield (2.9 g; 6.81 mmol).


ESI-MS m/z for C20H28ClN2O6 found 427.2/429.2 [M+H]+; Rt=1.46 min (Method A); 1H NMR (250 MHz, CDCl3) δ 7.29-7.22 (m, 2H), 7.20-7.02 (m, 2H), 5.32-5.22 (m, 1H), 5.12-5.07 (m, 1H), 4.94-4.83 (m, 1H), 4.49-4.37 (m, 1H), 4.25-3.99 (m, 1H), 3.90-3.81 (m, 1H), 3.81-3.71 (m, 3H), 3.72-3.48 (m, 2H), 3.20-3.06 (m, 1H), 2.93-2.77 (m, 1H), 1.39 (s, 9H).


Step 2.
Synthesis of methyl (S)-4-((R)-2-amino-3-(4-chlorophenyl)propanoyl)morpholine-3-carboxylate hydrochloride (6b)



embedded image


The title compound (6b) was obtained as a hydrochloride salt from 6a (2.9 g; 6.81 mmol) according to the General Procedure IVa in 99% yield (2.44 g; 6.74 mmol).


ESI-MS m/z for C15H20ClN2O4 found 326.8/328.8 [M+H]+; Rt=0.56 min (Method A)


Step 3.
Synthesis of (7S,9aS)-7-(4-chlorobenzyl)hexahydropyrazino[2,1-c][1,4]oxazine-6,9-dione (6c)



embedded image


To a solution of crude 6b (2.44 g; 6.74 mmol) in MeOH (70 mL) Et3N (4.8 mL; 33.96 mmol) was added and the mixture was stirred at room temperature for 2 hours. LC-MS showed completion of the reaction. The mixture was concentrated in vacuo and the yellow oily residue was purified by flash column chromatography on silica (DCM/MeOH, 100:1 to 10:1, v/v). Compound 6c was obtained as a light yellow foam in 59% yield (1.18 g; 4.01 mmol).


ESI-MS m/z for C14H16ClN2O3 found 295.1/297.1 [M+H]+; Rt=0.84 min (Method A)


Step 4.
Synthesis of (7S,9aR)-7-(4-chlorobenzyl)octahydropyrazino[2,1-c][1,4]oxazine (6d)



embedded image


The title compound (6d) was obtained as a white solid from 6c (1.1 g; 3.73 mmol) according to the General Procedure Ib in 96% yield (950 mg; 3.57 mmol).


ESI-MS m/z for C14H20ClN2O found 267.1/269.1 [M+H]+; Rt=1.25 min (Method E)


Step 5.
Synthesis of (7S,9aR)-7-(4-chlorobenzyl)-8-(4-(5-methyloxazol-2-yl)cyclohexyl)octahydro-pyrazino[2,1-c][1,4]oxazine 2,2,2-trifluoroacetate (6)



embedded image


The title compound (6) was obtained as a single isomer as a TFA salt from 6d (150 mg; 0.560 mmol) and ketone 1g (100 mg; 0.560 mmol) according to the General Procedure VII in 14% yield (42 mg; 0.078 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 5:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 5:1, v/v) and then transformed into TFA salt with using TFA/DCM and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C24H33ClN3O2 found 430.3/432.3 [M+H]+; Rt=0.98 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 7.38-7.37 (m, 2H), 7.31-7.30 (m, 2H), 6.67 (q, J=1.2 Hz, 1H), 3.81-3.80 (m, 1H), 3.78-3.76 (m, 1H), 3.72 (dd, J=11.1 Hz, J=2.9 Hz, 1H), 3.59-3.56 (m, 1H), 3.42-3.38 (m, 2H), 3.30-3.27 (m, 1H), 3.21 (t, J=10.5 Hz, 1H), 3.00 (dd, J=13.2 Hz, J=3.9 Hz, 1H), 2.93-2.89 (m, 1H), 2.71 (tt, J=12.1 Hz, J=3.4 Hz, 1H), 2.52-2.49 (m, 2H), 2.48-2.36 (m, 2H), 2.28-2.26 (m, 2H), 2.22 (d, J=1.2 Hz, 3H), 2.20-2.17 (m, 1H), 2.14-2.12 (m, 2H), 1.58-1.43 (m, 4H).


Example 7
Synthesis of (7S,9aR)-7-(4-chlorobenzyl)-8-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)octahydropyrazino[2,1-c][1,4]oxazine 2,2,2-trifluoroacetate (7)



embedded image


The title compound (7) was obtained as a single isomer as a TFA salt from 6d (126 mg; 0.470 mmol) and ketone 2d (100 mg; 0.520 mmol) according to the General Procedure VII in 29% yield (76 mg; 0.136 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 3:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 10:1 to 5:1, v/v) and then transformed into TFA salt with using TFA/DCM and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C25H36ClN4O found 443.3/445.3 [M+H]+; Rt=1.02 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.36-7.35 (m, 2H), 7.29-7.27 (m, 2H), 5.90 (s, 1H), 3.78-3.71 (m, 3H), 3.62 (s, 3H), 3.60-3.57 (m, 1H), 3.38-3.35 (m, 1H), 3.31-3.27 (m, 2H), 3.21 (t, J=10.6 Hz, 1H), 3.00 (dd, J=13.5 Hz, J=3.8 Hz, 1H), 2.88-2.84 (m, 2H), 2.53-2.51 (m, 1H), 2.50-2.48 (m, 1H), 2.47-2.43 (m, 2H), 2.26-2.21 (m, 1H), 2.18 (s, 3H), 2.17-2.15 (m, 2H), 1.97-1.93 (m, 2H), 1.72-1.65 (m, 2H), 1.64-1.58 (m, 2H).


Example 8
Synthesis of (7S,9aR)-7-(4-chlorobenzyl)-8-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)octahydropyrazino[2,1-c][1,4]oxazine 2,2,2-trifluoroacetate (8)



embedded image


The title compound (8) was obtained during the synthesis of compound 7 as a second single isomer as a TFA salt from 6d (126 mg; 0.47 mmol) and ketone 2d (100 mg; 0.52 mmol) according to the General Procedure VII in 19% yield (52 mg; 0.09 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 3:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 10:1 to 5:1, v/v) and then transformed into TFA salt with using TFA/DCM and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C25H36ClN4O found 443.3/445.3 [M+H]+; Rt=1.00 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.37-7.36 (m, 2H), 7.31-7.30 (m, 2H), 5.81 (s, 1H), 3.82-3.77 (m, 2H), 3.74 (dd, J=11.1 Hz, J=3.0 Hz, 1H), 3.62-3.58 (m, 4H), 3.39-3.31 (m, 3H), 3.24 (t, J=10.5 Hz, 1H), 3.03 (dd, J=13.4 Hz, J=4.2 Hz, 1H), 2.93 (t, J=12.3 Hz, 1H), 2.55-2.51 (m, 3H), 2.48-2.43 (m, 2H), 2.27-2.22 (m, 3H), 2.17 (s, 3H), 2.04-2.02 (m, 2H), 1.53-1.39 (m, 4H).


Example 9
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid 2,2,2-trifluoroacetate (9)



embedded image


Step 1.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)morpholine-2-carboxylic acid hydrochloride (9a)



embedded image


The title compound (9a) was obtained as a hydrochloride salt from 5a (5.16 g; 14.5 mmol) according to the General Procedure IVa in 93% yield (3.93 g; 13.49 mmol).


ESI-MS m/z for C12H15ClNO3 found 256.1/258.1 [M+H]+; Rt=0.39 min (Method A)


Step 2.
Synthesis of methyl (2R,5S)-5-(4-chlorobenzyl)morpholine-2-carboxylate (9b)



embedded image


To a cooled to −12° C. (brine/CO2(s)) solution of an amino acid 9a (1.20 g; 4.10 mmol) in MeOH (25 mL) a solution of HCl/MeOH (3.2 M; 6.4 mL) was added dropwise and after 1 hour this reaction mixture was allowed to warm to room temperature and stirred overnight. After reaction was completed as judged by LC-MS, the solvent was evaporated. The residue was taken between 5% NaHCO3 and DCM. An organic layer was dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by flash column chromatography on silica (CHCl3/MeOH, 200:1 to 180:1, v/v). Compound 9b was obtained in 66% yield (726 mg; 2.70 mmol).


ESI-MS m/z for C13H17ClNO3 found 270.0/272.0 [M+H]+; Rt=0.58 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.30-7.27 (m, 2H), 7.21-7.18 (m, 2H), 4.19-4.14 (m, 1H), 3.67 (s, 3H), 3.54-3.46 (m, 2H), 3.19-3.14 (m, 1H), 2.90-2.84 (m, 2H), 2.58-2.55 (m, 2H).


Step 3.
Synthesis of methyl (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylate (9c and 9c′)



embedded image


The title compound (9c and 9c′) was obtained as a mixture of two isomers from 9b (150 mg; 0.560 mmol) and ketone 1g (100 mg; 0.560 mmol) according to the General Procedure VII in 61% yield (148 mg; 0.340 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 4:1 to 1:2, v/v) to give two separated isomers 9c in 30% yield (72 mg; 0.170 mmol) and 9c′ in 31% yield (76 mg; 0.176 mmol).


For 9c: ESI-MS m/z for C23H30ClN2O4 found 433.0/435.0 [M+H]+; Rt=1.08 min (Method A)


For 9c′: ESI-MS m/z for C23H30ClN2O4 found 433.0/435.0 [M+H]+; Rt=1.03 min (Method A)


Step 4.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid 2,2,2-trifluoroacetate (9)



embedded image


To the solution of 9c′ (76 mg; 0.176 mmol) in MeOH (10 mL) 1 M NaOH (2 mL) was added and whole was stirred at room temperature for 2 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the solvent was evaporated in vacuo and the residue was purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 20 mL/min). The title compound (9) was obtained as a single isomer as a TFA salt in 30% yield (28 mg; 0.053 mmol).


ESI-MS m/z for C22H28ClN2O4 found 419.2/421.2 [M+H]+; Rt=0.97 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 7.39 (d, JAA′=8.4 Hz, 2H), 7.30 (d, JBB′=8.4 Hz, 2H), 6.67 (q, J=1.1 Hz, 1H), 4.42-4.40 (m, 1H), 3.69-3.62 (m, 3H), 3.43-3.40 (m, 3H), 3.10-3.08 (m, 1H), 2.96-2.92 (m, 1H), 2.74-2.69 (m, 1H), 2.22 (d, J=1.2 Hz, 3H), 2.14-2.09 (m, 4H), 1.57-1.46 (m, 4H).


Example 10
Synthesis of methyl (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylate 2,2,2-trifluoroacetate (10)



embedded image


The title compound (10) was obtained as a single isomer as a TFA salt from 9b (207 mg; 0.77 mmol) and ketone 2d (150 mg; 0.78 mmol) according to the General Procedure VII in 24% yield (101 mg; 0.18 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 2:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 16:1, v/v) and then transformed into TFA salt with using TFA/DCM and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 90:10 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C24H33ClN3O3 found 446.1/448.1 [M+H]+; Rt=1.01 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.37 (d, JAA′=8.5 Hz, 2H), 7.30 (d, JBB′=8.5 Hz, 2H), 5.81 (s, 1H), 4.56 (dd, J=8.6 Hz, J=3.6 Hz, 1H), 3.76-3.70 (m, 6H), 3.60 (s, 3H), 3.55-3.53 (m, 1H), 3.50-3.44 (m, 2H), 3.13 (dd, J=14.0 Hz, J=4.5 Hz, 1H), 3.01 (dd, J=14.0 Hz, J=9.9 Hz, 1H), 2.50-2.45 (m, 1H), 2.15-2.11 (m, 5H), 2.05-1.99 (m, 2H), 1.62-1.50 (m, 2H), 1.44 (qd, J=13.2 Hz, J=3.3 Hz, 1H), 1.37 (qd, J=13.2 Hz, J=3.3 Hz, 1H).


Example 11
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid 2,2,2-trifluoroacetate (11)



embedded image


To the solution of 10 (95 mg; 0.17 mmol) in MeOH (10 mL) 1 M NaOH (3 mL) was added and whole was stirred at room temperature for 2 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the solvent was evaporated in vacuo and the residue was dissolved in water/MeCN with few drops of TFA and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 20:80.30 min, 20 mL/min). The title compound (11) was obtained as a single isomer as a TFA salt in 53% yield (48 mg; 0.09 mmol).


ESI-MS m/z for C2H31ClN3O3 found 432.2/434.2 [M+H]+; Rt=0.98 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38-7.36 (m, 2H), 7.31-7.30 (m, 2H), 5.80 (s, 1H), 4.46-4.44 (m, 1H), 3.75-3.72 (m, 3H), 3.59 (s, 3H), 3.53-3.50 (m, 1H), 3.46-3.43 (m, 2H), 3.13-3.11 (m, 1H), 3.02 (dd, J=14.1 Hz, J=9.6 Hz, 1H), 2.46 (tt, J=12.2 Hz, J=3.7 Hz, 1H), 2.18-2.12 (m, 5H), 2.05-1.98 (m, 2H), 1.61-1.50 (m, 2H), 1.43 (qd, J=13.0 Hz, J=3.3 Hz, 1H), 1.36 (qd, J=13.0 Hz, J=3.3 Hz, 1H).


Example 12
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(1-(2,2,2-trifluoroethyl)-1H-1,2,3-triazol-4-yl)morpholine 2,2,2-trifluoroacetate (12)



embedded image


Step 1.
Synthesis of (2R,5S)-tert-butyl 5-(4-chlorobenzyl)-2-formylmorpholine-4-carboxylate (12a)



embedded image


To a sulfur trioxide-pyridine complex (93 mg; 0.59 mmol), pyridine (46 mg; 0.59 mmol) and DMSO (0.11 mL; 1.45 mmol) were added. The resulting suspension was stirred for 15 minutes, then DCM (4 mL) was added. The reaction was cooled to 0° C. and solution of an aminoalcohol 3d (0.1 g; 0.29 mmol), DIPEA (177 μL; 1 mmol) and DMSO (0.11 mL; 1.45 mmol) were added. The reaction was stirred at 0° C. for 2 hours and after this time it was allowed to warm up to room temperature. The reaction progress was monitored by TLC and LC-MS analyses of small aliquots of the crude reaction mixture. When analyses indicated completion of the reaction, water (10 mL) was added. The organic phase was separated, washed with brine, dried over anhydrous MgSO4 and concentrated in vacuo. After evaporation of solvent, the crude product 12a (97 mg; 0.29 mmol) was obtained in 99% yield.


ESI-MS m/z for C17H23ClNO4 found 340.1/342.1 [M+H]+; Rt=1.25 min (Method A)


Step 2.
Synthesis of tert-butyl (2S,5S)-5-(4-chlorobenzyl)-2-ethynylmorpholine-4-carboxylate (12b)



embedded image


To a suspension of K2CO3 (726 mg; 5.25 mmol), p-toluenesulfonyl azide (415 mg; 2.1 mmol) in MeCN (30 mL) dimethyl-2-oxopropylphosphonate (0.29 mL; 2.1 mmol) was added and this mixture was stirred at room temperature for 2 hours. Then the solution of 12a (593 mg; 1.75 mmol) in MeOH (6 mL) was added and the resulting mixture was stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the reaction mixture was concentrated in vacuo and the residue was suspended in Et2O. After 3 minutes in ultrasound bath an ether was collected by decantation. This manner (action) has been repeated two times. A solid residue was taken between DCM/water followed by an organic layer was concentrated in vacuo. The crude product was purified by flash column chromatography on silica (hexane/AcOEt; 30:1 to 5:1, v/v). Compound 12b was obtained as a white powder in 63% yield (1.11 g; 3.31 mmol).


ESI-MS m/z for C18H23ClNO3 found 336.1/338.1 [M+H]+; Rt=1.77 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.31-7.27 (m, 2H), 7.21-7.18 (m, 2H), 4.16-4.11 (m, 1H), 4.08-4.03 (m, 1H), 3.90-3.84 (m, 1H), 3.68 (d, J=11.8 Hz, 1H), 3.52 (dd, J=11.8, 3.4 Hz, 1H), 3.36 (d, J=2.2 Hz, 1H), 3.12-3.06 (m, 1H), 2.99-2.90 (m, 1H), 2.87-2.78 (m, 1H), 1.22 (s, 9H).


Step 3.
Synthesis of tert-butyl (2R,5S)-5-(4-chlorobenzyl)-2-(1-(2,2,2-trifluoroethyl)-1H-1,2,3-triazol-4-yl)morpholine-4-carboxylate (12c)



embedded image


To a solution of 12b (370 mg; 1.10 mmol) in MeOH (4 mL) CuSO4×5 H2O (137 mg; 0.55 mmol) was added and after 10 minutes sodium ascorbate (218 mg; 1.10 mmol) was added in one portion and the mixture was stirred at room temperature for 20 minutes. Then to this mixture 2-azido-1,1,1-trifluoroethane (2.2 mL; 1.10 mmol) was added and the mixture was stirred at room temperature for 2 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the green suspension was concentrated in vacuo and the residue was taken between water/DCM. An organic layer was washed with brine, dried over anhydrous MgSO4, filtered and concentrated in vacuo and the crude product mixture was purified by flash column chromatography on silica (CHCl3/MeOH, 100:1 to 50:1, v/v). Compound 12c was obtained as a white foam in 88% yield (445 mg; 0.97 mmol).


ESI-MS m/z for C20H25ClF3N4O3 found 461.0/463.0 [M+H]+; Rt=1.78 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 8.26 (s, 1H), 7.31-7.28 (m, 2H), 7.24-7.21 (m, 2H), 5.43-5.35 (m, 2H), 4.62-4.57 (m, 1H), 4.18-4.12 (m, 1H), 4.04-3.95 (m, 1H), 3.83-3.78 (m, 1H), 3.73-3.65 (m, 1H), 3.36-3.26 (m, 1H), 3.08-3.00 (m, 1H), 2.93-2.85 (m, 1H), 1.23 (s, 9H); 19F NMR (235 MHz, DMSO-d6) δ −70.06-−70.82 (m).


Step 4.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-2-(1-(2,2,2-trifluoroethyl)-1H-1,2,3-triazol-4-yl)morpholine (12d)



embedded image


The title compound (12d) was obtained as a free amine in 91% yield (317 mg; 0.88 mmol) from 12c (445 mg; 0.97 mmol) according to the General Procedure IVa and then after basic (5% NaHCO3aq) extraction with DCM.


ESI-MS m/z for C15H17ClF3N4O found 360.9/362.9 [M+H]+; Rt=0.89 min (Method A)


Step 5.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(1-(2,2,2-trifluoroethyl)-1H-1,2,3-triazol-4-yl)morpholine 2,2,2-trifluoroacetate (12)



embedded image


The title compound (12) was obtained as a single isomer as a TFA salt from 12d (100 mg; 0.28 mmol) and ketone 2d (56 mg; 0.29 mmol) according to the General Procedure VII in 18% yield (33 mg; 0.05 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 1:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 20:1, v/v) and then transformed into TFA salt with using TFA/DCM and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 90:10 to 20:80, 30 min, 18 mL/min).


ESI-MS m/z for C26H33ClF3N6O found 537.2/539.2 [M+H]+; Rt=1.10 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 8.38 (s, 1H), 7.39 (d, JAA′=8.5 Hz, 2H), 7.33 (d, JBB′=8.5 Hz, 2H), 5.82 (s, 1H), 544 (q, JH-F=9.0 Hz, 2H), 5.10-5.08 (m, 1H), 3.93-3.91 (m, 1H), 3.88-3.85 (m, 1H), 3.75-3.72 (m, 2H), 3.65-3.63 (m, 1H), 3.60 (s, 3H), 3.55-3.51 (m, 1H), 3.25 (dd, J=13.9 Hz, J=10.6 Hz, 1H), 3.20-3.17 (m, 1H), 2.49-2.46 (m, 1H), 2.32-2.26 (m, 2H), 2.17 (d, J=0.6 Hz, 3H), 2.06-2.04 (m, 2H), 1.60-1.53 (m, 2H), 1.49-1.41 (m, 2H); 19F NMR (700 MHz, DMSO-d6+D2O) δ −70.1 (t, JF-H=8.7 Hz), —74.1 (s, TFA).


Example 13
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-(1-(2,2,2-trifluoroethyl)-1H-1,2,3-triazol-4-yl)morpholine 2,2,2-trifluoroacetate (13)



embedded image


The title compound (13) was obtained as a single isomer as a TFA salt from 12d (110 mg; 0.31 mmol) and ketone 1g (57 mg; 0.32 mmol) according to the General Procedure VII in 6% yield (13 mg; 0.02 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 1:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 40:1, v/v) and then transformed into TFA salt with using TFA/DCM and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 90:10 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C25H30ClF3N5O2 found 524.2/526.2 [M+H]+; Rt=1.11 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 8.36 (s, 1H), 7.38 (d, JAA′=8.5 Hz, 2H), 7.32 (d, JBB′=8.5 Hz, 2H), 6.65 (q, J=1.2 Hz, 1H), 5.42 (q, JH-F=9.0 Hz, 2H), 5.08-5.06 (m, 1H), 3.91-3.89 (m, 1H), 3.83-3.81 (m, 1H), 3.73-3.69 (m, 2H), 3.62-3.58 (m, 1H), 3.53-3.51 (m, 1H), 3.23 (dd, J=13.8 Hz, J=10.7 Hz, 1H), 3.17 (dd, J=13.8 Hz, J=4.2 Hz, 1H), 2.76-2.72 (m, 1H), 2.34-2.29 (m, 2H), 2.22 (d, J=1.2 Hz, 3H), 2.20-2.17 (m, 2H), 1.62-1.53 (m, 4H); 19F NMR (700 MHz, DMSO-d6+D2O) δ −70.1 (t, JF-H=8.7 Hz), —73.9 (s, TFA).


Example 14
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)morpholine-2-carboxamide 2,2,2-trifluoroacetate (14)



embedded image


The title compound (14) was obtained as a single isomer as a TFA salt from 11 (28 mg; 0.050 mmol) according to the General Procedure XI in 18% yield (12 mg; 0.009 mmol). The crude product was purified twice by preparative reversed-phase column chromatography (first: column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 90:10 to 10:90, 30 min, 20 mL/min; second: column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 45:55, 30 min, 20 mL/min).


ESI-MS m/z for C57H95ClN7O15S found 1184.6/1185.9 [M+H]+; Rt=1.06 min (Method A)


Example 15
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine 2,2,2-trifluoroacetate (15)



embedded image


Step 1.
Synthesis of 1,5-dimethyl-3-(1,4-dioxaspiro[4.5]dec-7-en-8-yl)-1H-1,2,4-triazole (15a)



embedded image


The solution of 4,4,5,5-tetramethyl-2-(1,4-dioxaspiro[4.5]dec-7-en-8-yl)-1,3,2-dioxaborolane (560 mg; 2.10 mmol), 3-bromo-1,5-dimethyl-1H-1,2,4-triazole (352 mg, 2.00 mmol), [PPh3]4Pd (116 mg; 0.10 mmol) in mixture of dioxane and 2 M aqueous solution of K2CO3 (10 mL; 4:1 v/v) was heated at 100° C. for 24 hours under argon atmosphere. TLC showed complete consumption of the starting material. Then AcOEt (40 mL) was added and this mixture was washed with water (3×). An organic layer was concentrated in vacuo and the residue was purified by flash column chromatography on silica (hexane/AcOEt, 1:3, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 14:1, v/v). Compound 15a was obtained in 28% yield (132 mg; 0.56 mmol).


ESI-MS m/z for C12H18N3O2 found 236.0 [M+H]+; Rt=0.58 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 6.43-6.40 (m, 1H), 3.88 (s, 4H), 3.68 (s, 3H), 2.50-2.47 (m, 2H), 2.33-2.30 (m, 5H), 1.77-1.68 (m, 2H).


Step 2.
Synthesis of 1,5-dimethyl-3-(1,4-dioxaspiro[4.5]decan-8-yl)-1H-1,2,4-triazole (15b)



embedded image


To the solution of 15a (132 mg; 0.56 mmol) in AcOEt (10 mL) 10% Pd/C (5 mol %; 27 mg, 0.025 mmol) was added and the reaction mixture was conducted under hydrogen atmosphere. The reaction progress was monitored by LC-MS. After 2.5 hours, when analysis indicated completion of the reaction, Pd/C was filtered off through the Celite and the solvent was concentrated in vacuo. The crude product was used to the next step without additional purification. Compound 15b was obtained in 93% yield (124 mg; 0.52 mmol).


ESI-MS m/z for C12H20N3O2 found 238.0 [M+H]+; Rt=0.36 min (Method A)


Step 3.
Synthesis of 4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexan-1-one (15c)



embedded image


The solution of 15b (124 mg; 0.52 mmol) in 2 M HCl (10 mL) was stirred at room temperature for 2.5 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, to this mixture aqueous solution of NaOH (4 M) was added to pH 13 and then extracted with DCM (2×). The combined organic solutions were dried over anhydrous MgSO4, filtered and concentrated in vacuo. The crude product was used to the next step without additional purification. Compound 15c was obtained in 94% yield (95 mg; 0.49 mmol).


ESI-MS m/z for C10H16N3O found 194.0 [M+H]+; Rt=0.93 min (Method A)


Step 4.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine 2,2,2-trifluoroacetate (15)



embedded image


The title compound (15) was obtained as a single isomer as a TFA salt from 3h (184 mg; 0.54 mmol) and ketone 15c (95 mg; 0.49 mmol) according to the General Procedure VII in 18% yield (53 mg; 0.09 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 1:1 to 1:4, v/v, then: AcOEt/MeOH, 100:0 to 4:1, v/v) and then transformed into TFA salt with using TFA/DCM and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C23H34ClN4O3S found 481.1/483.1 [M+H]+; Rt=0.80 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.5 Hz, 2H), 7.32 (d, JBB′=8.5 Hz, 2H), 4.29-4.26 (m, 1H), 3.80-3.77 (m, 2H), 3.69 (s, 3H), 3.68-3.66 (m, 1H), 3.60-3.56 (m, 2H), 3.49-3.44 (m, 1H), 3.41-3.39 (m, 1H), 3.31-3.28 (m, 1H), 3.19 (dd, J=13.7 Hz, J=11.0 Hz, 1H), 3.11 (dd, J=13.7 Hz, J=3.9 Hz, 1H), 3.03 (s, 3H), 2.64-2.60 (m, 1H), 2.36 (s, 3H), 2.28-2.26 (m, 2H), 2.14-2.11 (m, 2H), 1.58-1.53 (m, 4H).


Example 16
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(fluoromethyl)morpholine 2,2,2-trifluoroacetate (16)



embedded image


Step 1.
Synthesis of tert-butyl (2R,5S)-5-(4-chlorobenzyl)-2-(fluoromethyl)morpholine-4-carboxylate (16a)



embedded image


The title compound (16a) was obtained from 3d (160 mg; 0.46 mmol) according to the General Procedure XII in 60% yield (95 mg; 0.28 mmol).


ESI-MS C12H16ClFNO found 243.9/245.9 [M+H−Boc]+; Rt=1.76 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.31-7.24 (m, 2H), 7.23-7.12 (m, 2H), 4.54-4.48 (m, 1H), 4.46-4.40 (m, 1H), 4.08-4.02 (m, 1H), 3.79-3.68 (m, 2H), 3.63-3.57 (m, 1H), 3.54-3.50 (m, 1H), 3.01-2.90 (m, 2H), 2.84-2.79 (m, 1H), 1.31-1.13 (m, 9H).


Step 2.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-2-(fluoromethyl)morpholine hydrochloride (3b)



embedded image


The title compound (16b) was obtained as a hydrochloride salt from 16a (0.55 g; 1.59 mmol) according to the General Procedure IVa in 99% yield (439 mg; 1.57 mmol).


ESI-MS C12H16ClFNO found 243.9/245.9 [M+H]+; Rt=0.63 min (Method A)


Step 3.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(fluoromethyl)morpholine 2,2,2-trifluoroacetate (16)



embedded image


The title compound (16) was obtained as a single isomer as a TFA salt from 16b (61 mg; 0.220 mmol) and ketone 2d (47 mg; 0.240 mmol) according to the General Procedure VII in 8% yield (9 mg; 0.017 mmol). The crude product was purified by flash column chromatography on silica (DCM/MeOH, 100:0 to 9:1, v/v) and then transformed into TFA salt with using TFA/DCM and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C23H32ClFN3O found 420.1/422.1 [M+H]+; Rt=1.03 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.4 Hz, 2H), 7.32 (d, JBB′=8.4 Hz, 2H), 5.84 (s, 1H), 4.60 (qd, J=10.6 Hz, J=3.6 Hz, 1H), 4.53 (qd, J=10.6 Hz, J=3.6 Hz, 1H), 4.07-4.02 (m, 1H), 3.80-3.76 (m, 2H), 3.68-3.66 (m, 1H), 3.61 (s, 3H), 3.58-3.56 (m, 1H), 3.49-3.47 (m, 1H), 3.23-3.20 (m, 1H), 3.15-3.09 (m, 2H), 2.49-2.46 (m, 1H), 2.31-2.25 (m, 2H), 2.17 (s, 3H), 2.06-2.03 (m, 2H), 1.57-1.50 (m, 2H), 1.48-1.41 (m, 2H); 19F NMR (250 MHz, DMSO-d6+D2O) δ −74.8 (s, TFA), —232.1 (td, JF-H=47.2 Hz, JF-H=23.4 Hz).


Example 17
Synthesis of ((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methanol 2,2,2-trifluoroacetate (17)



embedded image


The title compound (17) was obtained as a single isomer from 3c (3.495 g; 14.5 mmol) and ketone 2d (2.900 g; 15.0 mmol) according to the General Procedure VII in 21% yield (1.305 g; 3.1 mmol). The sample of 32 mg of the product was transformed into TFA salt with using TFA/DCM and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 30:70, 30 min, 20 mL/min) to give a product as a TFA salt.


ESI-MS m/z for C23H33ClN3O2 found 418.1/420.1 [M+H]+; Rt=0.90 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.40-7.37 (m, 2H), 7.34-7.31 (m, 2H), 5.83 (d, J=0.9 Hz, 1H), 3.82-3.71 (m, 3H), 3.66-3.63 (m, 1H), 3.61 (s, 3H), 3.59-3.55 (m, 2H), 3.52-3.47 (m, 2H), 3.19-3.12 (m, 2H), 3.12-3.06 (m, 1H), 2.50-2.44 (m, 1H), 2.34-2.23 (m, 2H), 2.17 (s, 3H), 2.07-2.01 (m, 2H), 1.58-1.48 (m, 2H), 1.49-1.41 (m, 2H).


Example 18
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-N-ethyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxamide 2,2,2-trifluoroacetate (18)



embedded image


Step 1.
Synthesis of tert-butyl (2R,5S)-5-(4-chlorobenzyl)-2-(ethylcarbamoyl)morpholine-4-carboxylate



embedded image


The title compound (18a) was obtained from 5a (712 mg; 2.20 mmol) according to the General Procedure V in 39% yield (330 mg; 0.86 mmol).


ESI-MS m/z for C19H28ClN2O4 found 383.0/385.0 [M+H]+; Rt=1.54 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.30-7.26 (m, 2H), 7.21-7.18 (m, 2H), 4.10-3.92 (m, 2H), 3.81-3.74 (m, 2H), 3.61-3.53 (m, 1H), 3.21-3.09 (m, 2H), 2.97-2.90 (m, 2H), 2.88-2.80 (m, 1H), 1.21 (s, 8H), 1.05 (t, J=7.2 Hz, 3H).


Step 2.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-N-ethylmorpholine-2-carboxamide hydrochloride (18b)



embedded image


The title compound (18b) was obtained as a hydrochloride salt from 18a (330 mg; 0.86 mmol) according to the General Procedure IVa in 99% yield (240 mg; 0.85 mmol).


ESI-MS m/z for C14H20ClN2O2 found 283.1/285.1 [M+H]+; Rt=0.61 min (Method A)


Step 3.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-N-ethyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxamide 2,2,2-trifluoroacetate (18)



embedded image


The title compound (18) was obtained as a single isomer as a TFA salt from 18b (150 mg; 0.47 mmol) and ketone 1g (95 mg; 0.52 mmol) according to the General Procedure VII in 4% yield (13 mg; 0.02 mmol). The crude product was purified by silica-gel column chromatography (pentane/AcOEt, 1:1 to 1:5, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 25:1 to 20:1, v/v) and then transformed into TFA salt with using TFA/DCM and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 30:70, 30 min, 20 mL/min).


ESI-MS m/z for C24H33ClN3O3 found 446.2/448.2 [M+H]+; Rt=1.03 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.5 Hz, 2H), 7.35 (d, JBB′=8.5 Hz, 2H), 6.65 (q, J=1.2 Hz, 1H), 4.31-4.29 (m, 1H), 3.78-3.76 (m, 2H), 3.73-3.70 (m, 1H), 3.56-3.54 (m, 1H), 3.51-3.48 (m, 1H), 3.35-3.32 (m, 1H), 3.23-3.07 (m, 4H), 2.76-2.71 (m, 1H), 2.26-2.25 (m, 1H), 2.23 (d, J=1.2 Hz, 3H), 2.22-2.13 (m, 3H), 1.62-1.51 (m, 4H), 1.07 (t, J=7.2 Hz, 3H).


Example 19
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide 2,2,2-trifluoroacetate (19)



embedded image


The title compound (19) was obtained as a single isomer as a TFA salt from 18b (150 mg; 0.470 mmol) and ketone 2d (100 mg; 0.520 mmol) according to the General Procedure VII in 20% yield (55 mg; 0.096 mmol). The crude product was purified by silica-gel column chromatography (pentane/AcOEt, 2:3 to 1:6, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 120:1 to 10:1, v/v) and then transformed into TFA salt with using TFA/DCM and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C25H36ClN4O2 found 459.2/461.2 [M+H]+; Rt=1.00 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.5 Hz, 2H), 7.34 (d, JBB′=8.5 Hz, 2H), 5.82 (s, 1H), 4.31-4.30 (m, 1H), 3.79-3.76 (m, 2H), 3.73-3.71 (m, 1H), 3.59 (s, 3H), 3.58-3.56 (m, 1H), 3.51-3.49 (m, 1H), 3.36-3.32 (m, 1H), 3.23-3.12 (m, 2H), 3.11-3.09 (m, 2H), 2.48-2.44 (m, 1H), 2.23-2.16 (m, 5H), 2.05-1.99 (m, 2H), 1.59-1.50 (m, 2H), 1.46-1.36 (m, 2H), 1.07 (t, J=7.2 Hz, 3H).


Example 20
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N,N-dimethylmorpholine-2-carboxamide 2,2,2-trifluoroacetate (20)



embedded image


The title compound (20) was obtained as a TFA salt as a single isomer in 15% overall yield in a similar way to Example 18 with the exception that, in the first step of the synthesis, dimethylamine (2 M solution) was used instead of ethylamine hydrochloride and in the third step of the synthesis, the compound 2d was used instead of the compound 1g. The crude product was purified by silica-gel column chromatography (pentane/AcOEt, 2:3 to 1:6, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 6:1 to 5:1, v/v) and then transformed into TFA salt with using TFA/DCM and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 30:70, 30 min, 20 mL/min).


ESI-MS m/z for C25H36ClN4O2 found 459.2/461.2 [M+H]+; Rt=0.98 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.36 (d, JAA′=8.4 Hz, 2H), 7.29 (d, JBB′=8.4 Hz, 2H), 5.84 (s, 1H), 4.74-4.73 (m, 1H), 3.82-3.80 (m, 2H), 3.64-3.60 (m, 4H), 3.56-3.51 (m, 2H), 3.43-3.41 (m, 1H), 3.19-3.17 (m, 1H), 3.02-3.00 (m, 4H), 2.88 (brs, 3H), 2.49-2.45 (m, 1H), 2.20-2.16 (m, 4H), 2.12-2.09 (m, 1H), 2.05-1.98 (m, 2H), 1.61-1.51 (m, 2H), 1.44 (qd, J=13.0 Hz, J=3.3 Hz, 1H), 1.35-1.32 (m, 1H).


Example 21
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-N,N-dimethyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxamide 2,2,2-trifluoroacetate (21)



embedded image


The title compound (21) was obtained as a TFA salt as a single isomer in 3% overall yield in a similar way to Example 18 with the exception that, in the first step of the synthesis, dimethylamine (2 M solution) was used instead of ethylamine hydrochloride. The crude product was purified by silica-gel column chromatography (pentane/AcOEt, 7:3, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 180:1, v/v) and then transformed into TFA salt with using TFA/DCM and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 30:70, 30 min, 20 mL/min).


ESI-MS m/z for C24H33ClN3O3 found 446.1/448.1 [M+H]+; Rt=0.99 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.37 (d, JAA′=8.4 Hz, 2H), 7.30 (d, JBB′=8.4 Hz, 2H), 6.65-6.64 (m, 1H), 4.75-4.73 (m, 1H), 3.82-3.80 (m, 2H), 3.64-3.57 (m, 2H), 3.54-3.53 (m, 1H), 3.43-3.41 (m, 1H), 3.21-3.19 (m, 1H), 3.03-3.00 (m, 4H), 2.88 (brs, 3H), 2.72 (tt, J=12.1 Hz, J=3.5 Hz, 1H), 2.24-2.22 (m, 4H), 2.18-2.12 (m, 3H), 1.64-1.54 (m, 3H), 1.50-1.45 (m, 1H).


Example 22
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(5-methyl-1,3,4-oxadiazol-2-yl)morpholine 2,2,2-trifluoroacetate (22)



embedded image


Step 1.
Synthesis of 4-(tert-butyl) 2-methyl (2R,5S)-5-(4-chlorobenzyl)morpholine-2,4-dicarboxylate (22a)



embedded image


To the solution of compound 5a (1 g; 2.81 mmol) in DMF (15 mL) K2CO3 (777 mg; 5.62 mmol) and Mel (0.35 mL; 5.62 mmol) were added and the reaction mixture was stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, to this reaction mixture Et2O (200 mL) was added and a whole was washed with water (2×). Then dried over anhydrous MgSO4, filtered and concentrated in vacuo. The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 20:1 to 5:1, v/v). Compound 22a was obtained in 69% yield (717 mg; 1.94 mmol).


ESI-MS m/z for C18H25ClNO5 found 370.2/372.2 [M+H]+; Rt=1.59 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 7.32-7.26 (m, 2H), 7.22-7.17 (m, 2H), 4.11-3.97 (m, 3H), 3.79-3.77 (m, 1H), 3.69 (s, 3H), 3.59-3.53 (m, 1H), 3.18-3.08 (m, 1H), 3.04-2.90 (m, 1H), 2.80-2.68 (m, 1H), 1.28-1.02 (m, 9H).


Step 2.
Synthesis of tert-butyl (2R,5S)-5-(4-chlorobenzyl)-2-(hydrazinecarbonyl)morpholine-4-carboxylate (22b)



embedded image


To the suspension of 22a (715 mg; 1.93 mmol) in MeCN (30 mL) hydrazine monohydrate (1.16 mL; 23.20 mmol) was added and the reaction mixture was stirred under gentle reflux (94° C.) overnight. Then another portion of hydrazine monohydrate (0.5 mL; 10.00 mmol) was added and the reaction mixture was stirred under gentle reflux (94° C.) for 4 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the whole was concentrated in vacuo and the crude product was used to the next step without additional purification. Compound 22b was obtained in 99% yield (705 mg; 1.91 mmol).


ESI-MS m/z for C17H25ClN3O4 found 370.0/372.0 [M+H]+; Rt=1.28 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.29-7.25 (m, 2H), 7.21-7.18 (m, 2H), 4.10-4.04 (m, 1H), 4.00-3.91 (m, 1H), 3.89-3.83 (m, 1H), 3.78-3.74 (m, 1H), 3.58-3.52 (m, 1H), 3.04-2.96 (m, 1H), 2.96-2.91 (m, 1H), 2.89-2.82 (m, 1H), 1.23 (s, 9H).


Step 3.
Synthesis of tert-butyl (2R,5S)-5-(4-chlorobenzyl)-2-(5-methyl-1,3,4-oxadiazol-2-yl)morpholine-4-carboxylate (22c)



embedded image


To the suspension of 22b (705 mg; 1.91 mmol) in xylene (38 mL) trimethylorthoacetate (0.49 mL; 3.86 mmol) and acetic acid (0.14 mL; 2.30 mmol) were added and the reaction mixture was stirred under reflux for 6 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the reaction mixture was cooled to room temperature and whole was diluted with AcOEt (280 mL) and then washed with 5% NaHCO3 (3×70 mL) and brine (70 mL). The organic layer was dried over anhydrous MgSO4, filtered and concentrated in vacuo. The crude product was purified by silica-gel column chromatography (cyclohexane/AcOEt, 3:1 to 2:1, v/v). Compound 22c was obtained in 85% yield (640 mg; 1.63 mmol).


ESI-MS m/z for C19H25ClN3O4 found 394.0/396.0 [M+H]+; Rt=1.54 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.31-7.28 (m, 2H), 7.23-7.20 (m, 2H), 4.74-4.67 (m, 1H), 4.22-4.14 (m, 1H), 4.11-4.04 (m, 1H), 3.87-3.81 (m, 1H), 3.77-3.68 (m, 1H), 3.43-3.38 (m, 1H), 3.37 (s, 3H), 3.01-2.96 (m, 1H), 2.93-2.85 (m, 1H), 1.24 (s, 9H).


Step 4.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-2-(5-methyl-1,3,4-oxadiazol-2-yl)morpholine (22d)



embedded image


The title compound (22d) was obtained as a free amine in 49% yield (236 mg; 0.80 mmol) from 22c (638 mg; 1.62 mmol) according to the General Procedure IVa and then after basic (NaOH) extraction with DCM.


ESI-MS m/z for C14H17ClN3O2 found 294.0/296.0 [M+H]+; Rt=0.29 min (Method A)


Step 5.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(5-methyl-1,3,4-oxadiazol-2-yl)morpholine 2,2,2-trifluoroacetate (22)



embedded image


The title compound (22) was obtained as a single isomer as a TFA salt from 22d (118 mg; 0.400 mmol) and ketone 2d (82 mg; 0.420 mmol) according to the General Procedure VII in 5% yield (13 mg; 0.022 mmol). The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 1:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 100:1 to 10:1, v/v) and then transformed into TFA salt with using TFA/DCM and then purified twice by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 10:90, 30 min, 20 mL/min).


ESI-MS m/z for C25H33ClN5O2 found 470.2/472.2 [M+H]+; Rt=1.09 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.35 (d, JAA′=8.5 Hz, 2H), 7.28 (d, JBB′=8.5 Hz, 2H), 5.81 (s, 1H), 5.15-5.14 (m, 1H), 3.87-3.85 (m, 1H), 3.74-3.71 (m, 2H), 3.66-3.64 (m, 1H), 3.62-3.58 (m, 4H), 3.14-3.11 (m, 1H), 3.06 (dd, J=13.9 Hz, J=10.1 Hz, 1H), 2.51 (s, 3H), 2.49-2.44 (m, 2H), 2.19-2.14 (m, 5H), 2.04-1.99 (m, 2H), 1.56-1.48 (m, 2H), 1.46-1.36 (m, 2H).


Example 23
Synthesis of (2R,5S)—N′-acetyl-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carbohydrazide 2,2,2-trifluoroacetate (23)



embedded image


The title compound (23) was obtained during the synthesis of compound 22 as a by-product as a TFA salt as a single isomer from 22d (118 mg; 0.400 mmol) and ketone 2d (82 mg; 0.420 mmol) according to the General Procedure VII in 5% yield (12 mg; 0.020 mmol). The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 1:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 100:1 to 10:1, v/v) and then transformed into TFA salt with using TFA/DCM and then purified twice by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C25H35ClN5O3 found 488.2/490.2 [M+H]+; Rt=0.93 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.37 (d, JAA′=8.4 Hz, 2H), 7.33 (d, JBB′=8.4 Hz, 2H), 5.80 (s, 1H), 4.46-4.45 (m, 1H), 3.79-3.75 (m, 3H), 3.59 (s, 3H), 3.54-3.52 (m, 1H), 3.45-3.42 (m, 1H), 3.37-3.35 (m, 1H), 3.12-3.11 (m, 2H), 2.49-2.45 (m, 1H), 2.21-2.16 (m, 5H), 2.07-1.99 (m, 2H), 1.90 (s, 3H), 1.59-1.50 (m, 2H), 1.46-1.36 (m, 2H).


Example 24
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-2-(5-methyl-1,3,4-oxadiazol-2-yl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine 2,2,2-trifluoroacetate (24)



embedded image


The title compound (24) was obtained as a single isomer as a TFA salt from 22d (118 mg; 0.400 mmol) and ketone 1g (76 mg; 0.420 mmol) according to the General Procedure VII in 4% yield (9 mg; 0.016 mmol). The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 3:4, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 30:1 to 5:1, v/v) and then purified twice by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C24H30ClN4O3 found 457.4/459.4 [M+H]+; Rt=1.15 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.39 (d, JAA′=8.5 Hz, 2H), 7.29 (d, JBB′=8.5 Hz, 2H), 6.67 (q, J=1.1 Hz, 1H), 5.16-5.13 (m, 1H), 3.84-3.81 (m, 2H), 3.75-3.73 (m, 1H), 3.69-3.64 (m, 2H), 3.50-3.47 (m, 1H), 3.15-3.13 (m, 1H), 3.06-3.03 (m, 1H), 3.75-3.71 (m, 1H), 2.52 (s, 3H), 2.24-2.22 (m, 5H), 2.15-2.12 (m, 2H), 1.54-1.50 (m, 4H).


Example 25
Synthesis of (2R,5S)—N′-acetyl-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carbohydrazide 2,2,2-trifluoroacetate (25)



embedded image


The title compound (25) was obtained during the synthesis of compound 24 as a by-product as a TFA salt as a single isomer from 22d (118 mg; 0.400 mmol) and ketone 1g (76 mg; 0.420 mmol) according to the General Procedure VII in 2% yield (4 mg; 0.007 mmol). The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 3:4, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 30:1 to 5:1, v/v) and then purified twice by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C24H32ClN4O4 found 475.4/477.4 [M+H]+; Rt=0.93 min (Method A)


Example 26
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (26)



embedded image


Step 1.
Synthesis of N-(but-3-yn-2-yl)-1,4-dioxaspiro[4.5]decane-8-carboxamide (26a)



embedded image


The title compound (26a) was obtained from 1,4-dioxaspiro[4.5]decane-8-carboxylic acid (802 mg; 4.30 mmol) and but-3-yn-2-amine hydrochloride (0.50 g; 4.73 mmol) according to the General Procedure V in 99% yield (1.01 g; 4.26 mmol).


ESI-MS m/z for C13H20NO3 found 238.2 [M+H]+; Rt=0.67 min (Method A)


Step 2.
Synthesis of 4,5-dimethyl-2-(1,4-dioxaspiro[4.5]decan-8-yl)oxazole (26b)



embedded image


To the solution of 26a (1.01 g; 4.26 mmol) in DCM (20 mL) placed into a reactor AuCl3 (66 mg; 0.21 mmol) was added and whole was stirred at room temperature overnight. The reaction progress was monitored by LC-MS analysis. After analytical control indicated completion of the reaction, a whole was concentrated in vacuo. The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 10:1 to 5:3, v/v). Compound 26b was obtained in 65% yield (655 mg; 2.76 mmol).


ESI-MS m/z for C13H20NO3 found 238.2 [M+H]+; Rt=1.04 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 3.84-3.83 (m, 4H), 2.76-2.69 (m, 1H), 2.14 (s, 3H), 1.93 (s, 3H), 1.91-1.86 (m, 2H), 1.70-1.63 (m, 4H), 1.57-1.51 (m, 2H).


Step 3.
Synthesis of 4-(4,5-dimethyloxazol-2-yl)cyclohexan-1-one (26c)



embedded image


The solution of 26b (655 mg; 2.76 mmol) and 1 N HCl (15 mL) in dioxane (10 mL) was stirred at room temperature for 3 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, dioxane was evaporated in vacuo and to this mixture an aqueous solution of NaOH (4 M) was added to pH 12 and then extracted with DCM (2×). The combined organic solutions were dried over anhydrous MgSO4, filtered and concentrated in vacuo. The crude product was used to the next step without additional purification. Compound 26c was obtained in 93% yield (495 mg; 2.56 mmol).


ESI-MS m/z for C11H16NO2 found 194.2 [M+H]+; Rt=0.83 min (Method A); 1H NMR (700 MHz, DMSO-d6+H2O, 348 K) δ 3.21-3.15 (m, 1H), 2.48-2.39 (m, 2H), 2.35-2.28 (m, 2H), 2.24-2.19 (m, 2H), 2.16 (s, 3H), 1.96 (s, 3H), 1.96-1.89 (m, 2H).


Step 4.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (26)



embedded image


The title compound (26) was obtained as a single isomer as a hydrochloride salt from 3h (173 mg; 0.51 mmol) and ketone 26c (95 mg; 0.49 mmol) according to the General Procedure VII in 14% yield (36 mg; 0.07 mmol). The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 1:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 90:1 to 10:1, v/v) and then purified twice by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 90:10 to 10:90, 30 min, 20 mL/min).


ESI-MS m/z for C24H35ClN2O4S found 481.1/483.1 [M+H]+; Rt=1.03 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 7.37 (d, JAA′=8.4 Hz, 2H), 7.31 (d, JBB′=8.4 Hz, 2H), 4.33-4.30 (m, 1H), 3.84-3.82 (m, 1H), 3.79-3.77 (m, 1H), 3.69-3.67 (m, 1H), 3.59-3.54 (m, 2H), 3.49-3.46 (m, 1H), 3.42-3.39 (m, 1H), 3.32-3.28 (m, 1H), 3.19 (dd, J=13.8 Hz, J=10.8 Hz, 1H), 3.12 (dd, J=13.8 Hz, J=4.0 Hz, 1H), 3.02 (s, 3H), 2.69 (tt, J=10.7 Hz, J=3.1 Hz, 1H), 2.29-2.26 (m, 2H), 2.17-2.14 (m, 5H), 1.95 (d, J=0.8 Hz, 3H), 1.62-1.52 (m, 4H).


Example 27
Synthesis of N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1,5-dimethyl-1H-pyrazole-3-carboxamide hydrochloride (27)



embedded image


Step 1.
Synthesis of (2R,5S)-tert-butyl 2-(azidomethyl)-5-(4-chlorobenzyl)morpholine-4-carboxylate (27a)



embedded image


To a solution of 3e (0.4 g; 0.95 mmol) in DMF (8 mL) NaN3 (0.19 g; 2.86 mmol) was added and the mixture was heated at 50° C. overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the reaction mixture was taken into water and AcOEt. An organic layer was washed with brine and then dried over anhydrous MgSO4, filtered and concentrated in vacuo and the crude product mixture was used to the next step without additional purification. Compound 27a was obtained in 99% yield (344 mg; 0.94 mmol).


ESI-MS m/z for C17H24ClN4O3 found 367.2/369.2 [M+H]+; Rt=6.60 min (Method C)


Step 2.
Synthesis of tert-butyl (2S,5S)-2-(aminomethyl)-5-(4-chlorobenzyl)morpholine-4-carboxylate (27b)



embedded image


An obtained azide 27a (225 mg; 0.61 mmol) was dissolved in THF/H2O (10 mL; 9:1, v/v) and then 1 M solution of Me3P in THF (1 mL; 0.92 mmol) was added and a whole was stirred at room temperature for 2 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the reaction was quenched with 5% NaHCO3 and a whole was extracted with diethyl ether (2×). The combined organic solutions were washed with water and then dried over anhydrous MgSO4, filtered and concentrated in vacuo. The crude product mixture was used to the next step without additional purification. Compound 27b was obtained in 90% yield (187 mg; 0.55 mmol).


ESI-MS m/z for C17H26ClN2O3 found 341.0/343.0 [M+H]+; Rt=1.06 min (Method A)


Step 3.
Synthesis of tert-butyl (2S,5S)-5-(4-chlorobenzyl)-2-((1,5-dimethyl-1H-pyrazole-3-carboxamido)methyl)morpholine-4-carboxylate (27c)



embedded image


To the solution of 27b (187 mg; 0.55 mmol) in dry DCM (3 mL) diisopropylethylamine (DIPEA; 0.14 mL; 0.83 mmol), 1,5-dimethyl-1H-pyrazole-3-carboxylic acid (84 mg; 0.60 mmol) and HATU (228 mg; 0.60 mmol) were added and the reaction mixture was stirred at room temperature overnight. The reaction progress was monitored by TLC and LC-MS. When analyses indicated completion of the reaction, the mixture was diluted with DCM and washed with an aqueous solution of HCl, and aqueous solution of NaOH, water and brine. The organic phase was dried over anhydrous MgSO4, filtered and concentrated in vacuo. The crude product was purified by flash column chromatography on silica (hexane/MeCN, 100:0 to 2:8, v/v). Compound 27c was obtained in 96% yield (247 mg; 0.53 mmol).


ESI-MS m/z for C23H32ClN4O4 found 463.2/465.1 [M+H]+; Rt=1.58 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.29-7.26 (m, 2H), 7.20-7.16 (m, 2H), 6.41 (s, 1H), 4.08-4.02 (m, 1H), 3.80-3.68 (m, 5H), 3.53-3.45 (m, 2H), 2.97-2.90 (m, 1H), 2.90-2.80 (m, 2H), 2.71-2.70 (m, 1H), 2.70-2.69 (m, 1H), 2.25 (s, 3H), 1.21 (s, 9H).


Step 4.
Synthesis of N-(((2R,5S)-5-(4-chlorobenzyl)morpholin-2-yl)methyl)-1,5-dimethyl-1H-pyrazole-3-carboxamide (27d)



embedded image


The title compound (27d) was obtained as a free amine in 77% yield (147 mg; 0.41 mmol) from 27c (247 mg; 0.53 mmol) according to the General Procedure IVa and then after basic (5% NaHCO3) extraction with DCM.


ESI-MS m/z for C18H24ClN4O2 found 363.2/365.0 [M+H]+; Rt=0.83 min (Method A)


Step 5.
Synthesis of N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1,5-dimethyl-1H-pyrazole-3-carboxamide hydrochloride (27)



embedded image


The title compound (27) was obtained as a single isomer as a hydrochloride salt from 27d (144 mg; 0.40 mmol) and ketone 2d (85 mg; 0.44 mmol) according to the General Procedure VII in 17% yield (43 mg; 0.07 mmol). The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 1:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 25:1 to 9:1, v/v) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C29H40ClN6O2 found 539.0/541.0 [M+H]+; Rt=1.06 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 7.40 (d, JAA′=8.5 Hz, 2H), 7.31 (d, JBB′=8.5 Hz, 2H), 6.44 (s, 1H), 5.86 (s, 1H), 3.90-3.87 (m, 1H), 3.80-3.78 (m, 1H), 3.73 (s, 3H), 3.68-3.66 (m, 1H), 3.60 (s, 3H), 3.59-3.53 (m, 2H), 3.47-3.38 (m, 3H), 3.11-3.03 (m, 3H), 2.47-2.43 (m, 1H), 2.26-2.23 (m, 5H), 2.16 (m, 3H), 2.02-1.99 (m, 2H), 1.53-1.37 (m, 4H).


Example 28
Synthesis of (2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-ethynylmorpholine 2,2,2-trifluoroacetate (28)



embedded image


Step 1.
Synthesis of (2S,5S)-5-(4-chlorobenzyl)-2-ethynylmorpholine (28a)



embedded image


The title compound (28a) was obtained as a free amine in 99% yield (313 mg; 1.33 mmol) from 12c (448 mg; 1.34 mmol) according to the General Procedure IVb and then after basic (5% NaHCO3aq) extraction with DCM.


ESI-MS m/z for C13H15ClNO found 236.1/238.0 [M+H]+; Rt=0.66 min (Method A)


Step 2.
Synthesis of (2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-ethynylmorpholine 2,2,2-trifluoroacetate (28)



embedded image


The title compound (28) was obtained as a single isomer as a TFA salt from 28a (313 mg; 1.330 mmol) and ketone 2d (280 mg; 1.450 mmol) according to the General Procedure VII in 4% yield (30 mg; 0.057 mmol). The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 6:1 to 1:4, v/v) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 10:90, 30 min, 20 mL/min).


ESI-MS m/z for C24H31ClN3O found 412.4/414.3 [M+H]+; Rt=1.06 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.37 (d, JAA′=8.4 Hz, 2H), 7.30 (d, JBB′=8.4 Hz, 2H), 5.80 (s, 1H), 4.69-4.68 (m, 1H), 3.73-3.69 (m, 3H), 3.60 (s, 3H), 3.50-3.48 (m, 2H), 3.44-3.41 (m, 1H), 3.36-3.33 (m, 1H), 3.16-3.13 (m, 1H), 3.04 (dd, J=14.0 Hz, J=9.5 Hz, 1H), 2.45 (tt, J=12.1 Hz, J=3.7 Hz, 1H), 2.18-2.16 (m, 4H), 2.12-2.10 (m, 1H), 2.05-1.98 (m, 2H), 1.58-1.47 (m, 2H), 1.43 (qd, J=13.0 Hz, J=3.3 Hz, 1H), 1.36 (qd, J=13.0 Hz, J=3.3 Hz, 1H).


Example 29
Synthesis of N-(35-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxapentatriacontyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide 2,2,2-trifluoroacetate (29)



embedded image


To the solution of 28 (22 mg; 0.054 mmol) in MeOH (0.5 mL) CuSO4×5 H2O (6.3 mg; 0.025 mmol) was added and after 5 minutes to a light green solution a sodium L-ascorbate (10 mg; 0.050 mmol) was added in one portion. A whole was stirred for 15 minutes at room temperature and then a solution of an azide (39 mg; 0.049 mmol) in MeOH (0.5 mL) was added and the reaction mixture was stirred at room temperature overnight. After analytical control (LC-MS) indicated completion of the reaction, the reaction mixture was concentrated in vacuo and the residue was mixed with water/MeCN and TFA and then this mixture was filtered and purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 10:90, 30 min, 20 mL/min). Compound 29 was obtained as a single isomer as a TFA salt in 41% yield (27 mg; 0.020 mmol).


ESI-MS m/z for C58H95ClN9O14S found 1208.5/1210.0 [M+H]+; Rt=1.08 min (Method A)


Example 30
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-isopropylmorpholine-2-carboxamide hydrochloride (30)



embedded image


Step 1.
Synthesis of tert-butyl (2R,5S)-5-(4-chlorobenzyl)-2-(isopropylcarbamoyl)morpholine-4-carboxylate (30a)



embedded image


To the solution of 5a (2.5 g; 7.03 mmol) in dry DCM (30 mL) diisopropylethylamine (DIPEA; 1.8 mL; 10.50 mmol), propan-2-amine (0.72 mL; 8.43 mmol) and HATU (3 g; 7.70 mmol) were added and the reaction mixture was stirred at room temperature overnight. The reaction progress was monitored by TLC and LC-MS. When analyses indicated completion of the reaction, the mixture was diluted with DCM and washed with an aqueous solution of HCl, and aqueous solution of NaOH, water and brine. The organic phase was dried over anhydrous MgSO4, filtered and concentrated in vacuo. The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 4:6, v/v). Compound 30a was obtained in 99% yield (2.76 g; 6.96 mmol).


ESI-MS m/z for C20H30ClN2O4 found 397.1/399.0 [M+H]+; Rt=1.69 min (Method A)


Step 2.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-N-isopropylmorpholine-2-carboxamide (30b)



embedded image


The title compound (30b) was obtained as a free amine in 86% yield (1.78 g; 6.01 mmol) from 27c (2.76 g; 6.96 mmol) according to the General Procedure IVa and then after basic (5% NaHCO3) extraction with DCM.


ESI-MS m/z for C15H22ClN2O2 found 297.2/299.0 [M+H]+; Rt=0.80 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.30-7.27 (m, 2H), 7.23-7.20 (m, 2H), 3.93-3.87 (m, 1H), 3.85-3.81 (m, 1H), 3.61-3.55 (m, 1H), 3.52-3.47 (m, 1H), 3.06-2.99 (m, 1H), 2.89-2.83 (m, 1H), 2.83-2.78 (m, 1H), 2.74-2.69 (m, 2H), 1.12-1.06 (m, 6H).


Step 3.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-isopropylmorpholine-2-carboxamide hydrochloride (30)



embedded image


The title compound (30) was obtained as a single isomer as a hydrochloride salt from 30b (119 mg; 0.400 mmol) and ketone 2d (85 mg; 0.440 mmol) according to the General Procedure VII in 24% yield (49 mg; 0.096 mmol). The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 1:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 70:1 to 10:1, v/v) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C26H38ClN4O2 found 473.5/475.5 [M+H]+; Rt=1.09 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.5 Hz, 2H), 7.33 (d, JBB′=8.5 Hz, 2H), 5.80 (s, 1H), 4.29-4.28 (m, 1H), 3.92 (hept, J=6.7 Hz, 1H), 3.80-3.79 (m, 1H), 3.74-3.72 (m, 2H), 3.60 (s, 3H), 3.53-3.51 (m, 1H), 3.45-3.41 (m, 1H), 3.35-3.33 (m, 1H), 3.13-3.07 (m, 2H), 2.48-2.44 (m, 2H), 2.22-2.19 (m, 1H), 2.16-2.13 (m, 4H), 2.06-2.00 (m, 2H), 1.61-1.51 (m, 2H), 1.46-1.36 (m, 2H), 1.14 (d, J=6.6 Hz, 3H), 1.11 (d, J=6.6 Hz, 3H).


Example 31
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-methylmorpholine-2-carboxamide dihydrochloride (31)



embedded image


The title compound (31) was obtained as a dihydrochloride salt as a single isomer in 2% overall yield in a similar way to Example 30 with the exception that, in the first step of the synthesis, methylamine was used instead of propan-2-amine. The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 1:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 100:1 to 70:4, v/v) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C24H34ClN4O2 found 445.4/447.2 [M+H]+; Rt=0.98 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.37 (d, JAA′=8.4 Hz, 2H), 7.34 (d, JBB′=8.4 Hz, 2H), 5.81 (s, 1H), 4.35-4.33 (m, 1H), 3.82-3.80 (m, 1H), 3.77-3.75 (m, 1H), 3.73-3.71 (m, 1H), 3.60 (s, 3H), 3.57-3.55 (m, 1H), 3.47-3.43 (m, 1H), 3.36-3.32 (m, 1H), 3.12-3.08 (m, 2H), 2.69 (s, 3H), 2.49-2.45 (m, 1H), 2.23-2.16 (m, 5H), 2.07-2.01 (m, 2H), 1.63-1.54 (m, 2H), 1.47-1.37 (m, 2H).


Example 32
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-N,N-dimethylmorpholine-2-carboxamide hydrochloride (32)



embedded image


The title compound (32) was obtained as a hydrochloride salt as a single isomer in 8% overall yield in a similar way to Example 30 with the exception that, in the first step of the synthesis, dimethylamine hydrochloride was used instead of propan-2-amine and in the third step of the synthesis, ketone 26c was used instead of ketone 2d. The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 3:4, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 140:1 to 70:4, v/v) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C25H35ClN3O3 found 460.5/462.3 [M+H]+; Rt=1.06 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.37 (d, JAA′=8.5 Hz, 2H), 7.31 (d, JBB′=8.5 Hz, 2H), 4.84-4.82 (m, 1H), 3.96-3.94 (m, 1H), 3.83-3.80 (m, 1H), 3.63-3.57 (m, 2H), 3.53-3.50 (m, 1H), 3.44-3.42 (m, 1H), 3.22-3.20 (m, 1H), 3.06-3.04 (m, 4H), 3.89 (brs, 3H), 2.69 (tt, J=12.2 Hz, J=3.6 Hz, 1H), 2.25-2.24 (m, 1H), 2.17-2.11 (m, 6H), 1.95 (s, 3H), 1.75-1.63 (m, 2H), 1.59-1.46 (m, 2H).


Example 33
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-N-isopropylmorpholine-2-carboxamide hydrochloride (33)



embedded image


The title compound (33) was obtained as a single isomer as a hydrochloride salt from 30b (157 mg; 0.529 mmol) and ketone 26c (102 mg; 0.527 mmol) according to the General Procedure VII in 15% yield (40 mg; 0.078 mmol). The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 3:4, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 140:1 to 20:1, v/v) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C26H37ClN3O3 found 474.4/476.4 [M+H]+; Rt=1.14 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.5 Hz, 2H), 7.33 (d, JBB′=8.5 Hz, 2H), 4.32-4.30 (m, 1H), 3.92 (hept, J=6.6 Hz, 1H), 3.83-3.81 (m, 1H), 3.76-3.72 (m, 2H), 3.54-3.52 (m, 1H), 3.49-3.46 (m, 1H), 3.34-3.30 (m, 1H), 3.14-3.08 (m, 2H), 2.68 (tt, J=12.0 Hz, J=3.6 Hz, 1H), 2.26-2.23 (m, 1H), 2.20-2.11 (m, 6H), 1.94 (s, 3H), 1.65-1.48 (m, 4H), 1.13 (d, J=6.6 Hz, 3H), 1.11 (d, J=6.6 Hz, 3H).


Example 34
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-N-methyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxamide hydrochloride (34)



embedded image


The title compound (34) was obtained as a hydrochloride salt as a single isomer in 4% overall yield in a similar way to Example 30 with the exception that, in the first step of the synthesis, methylamine was used instead of propan-2-amine and in the third step of the synthesis, ketone 1g was used instead of ketone 2d. The crude product was purified by silica-gel column chromatography (AcOEt/MeOH, 100:1 to 10:1, v/v) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 30:70, 30 min, 20 mL/min).


ESI-MS m/z for C23H31ClN3O3 found 432.4/434.2 [M+H]+; Rt=0.97 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.4 Hz, 2H), 7.35 (d, JBB′=8.5 Hz, 2H), 6.66 (s, 1H), 4.44 (dd, J=10.4 Hz, J=3.0 Hz, 1H), 3.92 (d, J=13.2 Hz, 1H), 3.81-3.79 (m, 1H), 3.72 (dd, J=13.2 Hz, J=1.9 Hz, 1H), 3.60-3.58 (m, 1H), 3.54-3.51 (m, 1H), 3.35-3.33 (m, 1H), 3.17-3.10 (m, 2H), 2.74 (tt, J=12.3 Hz, J=3.7 Hz, 1H), 2.69 (s, 3H), 2.28-2.23 (m, 5H), 2.20-2.15 (m, 2H), 1.76-1.66 (m, 2H), 1.59-1.52 (m, 2H).


Example 35
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide hydrochloride (35)



embedded image


The title compound (35) was obtained as a hydrochloride salt as a single isomer in 1% overall yield in a similar way to Example 30 with the exception that, in the first step of the synthesis, ethylamine hydrochloride was used instead of propan-2-amine and in the third step of the synthesis, ketone 26c was used instead of ketone 2d. The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 1:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 60:1 to 6:1, v/v) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 30:70, 30 min, 20 mL/min).


ESI-MS m/z for C25H35ClN3O3 found 460.4/462.4 [M+H]+; Rt=1.10 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.5 Hz, 2H), 7.35 (d, JBB′=8.5 Hz, 2H), 4.39-4.37 (m, 1H), 3.89-3.87 (m, 1H), 3.80-3.78 (m, 1H), 3.74-3.72 (m, 1H), 3.59-3.57 (m, 1H), 3.54-3.50 (m, 1H), 3.35-3.32 (m, 1H), 3.23-3.10 (m, 4H), 2.69 (tt, J=12.2 Hz, J=3.6 Hz, 1H), 2.28-2.21 (m, 2H), 2.18-2.13 (m, 5H), 1.95 (brs, 3H), 1.71-1.61 (m, 2H), 1.54 (pd, J=13.0 Hz, J=3.2 Hz, 2H), 1.08 (t, J=7.2 Hz, 3H).


Example 36
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-fluoro-1-methyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (36)



embedded image


The title compound (36) was obtained as a hydrochloride salt as a single isomer in 14% overall yield in a similar way to Example 15 with the exception that, in the first step of the synthesis, 3-bromo-5-fluoro-1-methyl-1H-pyrazole was used instead of 3-bromo-1,5-dimethyl-1H-1,2,4-triazole and Pd(dppf) was used instead of [PPh3]4Pd. The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 1:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 80:1 to 20:1, v/v) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C23H32ClFN3O3S found 484.2/486.2 [M+H]+; Rt=0.99 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.4 Hz, 2H), 7.32 (d, JBB′=8.4 Hz, 2H), 5.71 (d, J=5.7 Hz, 1H), 4.37-4.34 (m, 1H), 3.89 (d, J=13.3 Hz, 1H), 3.79-3.77 (m, 1H), 3.67 (d, J=13.3 Hz, 1H), 3.60-3.55 (m, 5H), 3.47-3.41 (m, 2H), 3.32-3.30 (m, 1H), 3.20 (dd, J=13.8 Hz, J=10.9 Hz, 1H), 3.14-3.11 (m, 1H), 3.02 (s, 3H), 2.49-2.45 (m, 1H), 2.27-2.24 (m, 2H), 2.07-2.04 (m, 2H), 1.65-1.59 (m, 2H), 1.48-1.41 (m, 2H); 19F NMR (250 MHz, DMSO-d6+D2O) δ −135.3 (s).


Example 37
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (37)



embedded image


The title compound (37) was obtained as a hydrochloride salt as a single isomer in 9% overall yield in a similar way to Example 15 with the exception that, in the first step of the synthesis, 2-bromo-4,5-dimethylthiazole was used instead of 3-bromo-1,5-dimethyl-1H-1,2,4-triazole and Pd(dppf) was used instead of [PPh3]4Pd. The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 1:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 100:1 to 95:5, v/v) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 10:90, 30 min, 20 mL/min).


ESI-MS m/z for C24H34ClN2O3S2 found 497.4/499.4 [M+H]+; Rt=1.07 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.39 (d, JAA′=8.4 Hz, 2H), 7.33 (d, JBB′=8.4 Hz, 2H), 4.31-4.28 (m, 1H), 3.83-3.77 (m, 2H), 3.66 (d, J=12.9 Hz, 1H), 3.61-3.56 (m, 2H), 3.51-3.48 (m, 1H), 3.42-3.40 (m, 1H), 3.30-3.26 (m, 1H), 3.19 (dd, J=13.5 Hz, J=11.0 Hz, 1H), 3.13 (dd, J=13.8 Hz, J=3.7 Hz, 1H), 3.03 (s, 3H), 2.88 (tt, J=11.9 Hz, J=3.7 Hz, 1H), 2.29-2.27 (m, 5H), 2.19-2.16 (m, 5H), 1.63-1.52 (m, 4H).


Example 38
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)-4-(4-(5-methylthiazol-2-yl)cyclohexyl)morpholine hydrochloride (38)



embedded image


The title compound (38) was obtained as a hydrochloride salt as a single isomer in 11% overall yield in a similar way to Example 15 with the exception that, in the first step of the synthesis, 2-bromo-5-methylthiazole was used instead of 3-bromo-1,5-dimethyl-1H-1,2,4-triazole and Pd(dppf) was used instead of [PPh3]4Pd. The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 5:1, v/v, then AcOEt, 100%) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 90:10 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C23H32ClN2O3S2 found 483.0/485.0 [M+H]+; Rt=1.05 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.5 Hz, 2H), 7.33-7.32 (m, 3H), 4.30-4.27 (m, 1H), 3.80-3.76 (m, 2H), 3.66 (d, J=12.6 Hz, 1H), 3.59 (dd, J=15.0 Hz, J=8.0 Hz, 1H), 3.56-3.54 (m, 1H), 3.49-3.46 (m, 1H), 3.40-3.39 (m, 1H), 3.29-3.25 (m, 1H), 3.18 (dd, J=13.7 Hz, J=10.8 Hz, 1H), 3.12 (dd, J=13.8 Hz, J=3.9 Hz, 1H), 3.03 (s, 3H), 2.94-2.90 (m, 1H), 2.38 (d, J=1.2 Hz, 3H), 2.29-2.28 (m, 2H), 2.21-2.18 (m, 2H), 1.62-1.55 (m, 4H).


Example 39
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid hydrochloride (39)



embedded image


The title compound (39) was obtained as a hydrochloride salt as a single isomer in 2% overall yield in a similar way to Example 9 with the exception that, in the third step of the synthesis, ketone 26c was used instead of ketone 1g and only one isomer was obtained. The crude product was purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C23H30ClN2O4 found 433.3/435.3 [M+H]+; Rt=1.04 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.34 (d, JAA′=8.5 Hz, 2H), 7.27 (d, JBB′=8.5 Hz, 2H), 4.31-4.29 (m, 1H), 3.64 (dd, J=12.4 Hz, J=4.0 Hz, 1H), 3.59 (dd, J=12.4 Hz, J=2.2 Hz, 1H), 3.42-3.40 (m, 1H), 3.23-3.20 (m, 2H), 3.16-3.13 (m, 1H), 3.00 (dd, J=13.7 Hz, J=4.3 Hz, 1H), 2.88 (dd, J=13.7 Hz, J=9.8 Hz, 1H), 2.65-2.61 (m, 1H), 2.14 (brd, J=0.9 Hz, 3H), 2.10-2.03 (m, 4H), 1.94 (brd, J=0.9 Hz, 3H), 1.54-1.39 (m, 4H).


Example 40
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid hydrochloride (40)



embedded image


The title compound (40) was obtained as a hydrochloride salt as a single isomer in 23% overall yield in a similar way to Example 9 with the exception that, in the third step of the synthesis, ketone 40c (synthesis of this ketone is described below) was used instead of ketone 1g and only one isomer was obtained and in the fourth step of the synthesis, 4 M NaOH was used instead of 1 M NaOH. The crude product was purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 10:90, 30 min, 20 mL/min).


ESI-MS m/z for C23H30ClN2O3S found 449.4/451.2 [M+H]+; Rt=1.07 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.36 (d, JAA′=8.5 Hz, 2H), 7.29 (d, JBB′=8.5 Hz, 2H), 4.36-4.35 (m, 1H), 3.67-3.63 (m, 2H), 3.52-3.51 (m, 1H), 3.32-3.26 (m, 3H), 3.05 (dd, J=13.9 Hz, J=4.4 Hz, 1H), 2.92 (dd, J=13.8 Hz, J=10.0 Hz, 1H), 2.82 (tt, J=11.6 Hz, J=3.3 Hz, 1H), 2.25 (brd, J=0.6 Hz, 3H), 2.18 (brd, J=0.8 Hz, 3H), 2.15-2.09 (m, 4H), 1.60-1.42 (m, 4H).


Synthesis of the ketone 40c
Step 1.
Synthesis of 4,5-dimethyl-2-(1,4-dioxaspiro[4.5]dec-7-en-8-yl)thiazole (40a)



embedded image


The solution of 4,4,5,5-tetramethyl-2-(1,4-dioxaspiro[4.5]dec-7-en-8-yl)-1,3,2-dioxaborolane (560 mg; 2.10 mmol), 2-bromo-4,5-dimethylthiazole (385 mg, 2.00 mmol), Pd(dppf) (163 mg; 0.10 mmol) in mixture of dioxane and 2 M aqueous solution of K2CO3 (12.5 mL; 4:1 v/v) was heated at 100° C. for 2.5 hours under argon atmosphere. TLC showed complete consumption of the starting material. Then a whole was concentrated in vacuo and the residue was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 4:6, v/v, 35 min, 30 mL/min). Compound 40a was obtained in 81% yield (408 mg; 1.62 mmol).


ESI-MS m/z for C13H18NO2S found 252.2 [M+H]+; Rt=1.11 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 6.32-6.29 (m, 1H), 3.89 (s, 4H), 2.57-2.54 (m, 2H), 2.36-2.33 (m, 2H), 2.27 (s, 3H), 2.20 (s, 3H), 1.79-1.73 (m, 2H).


Step 2.
Synthesis of 4,5-dimethyl-2-(1,4-dioxaspiro[4.5]decan-8-yl)thiazole (40b)



embedded image


To the solution of 40a (404 mg; 1.61 mmol) in AcOEt (15 mL) 10% Pd/C (5 mol %; 86 mg, 0.08 mmol) was added and the reaction mixture was conducted under hydrogen atmosphere. The reaction progress was monitored by LC-MS. After 4 hours, when analysis indicated completion of the reaction, Pd/C was filtered off through the Celite and the solvent was concentrated in vacuo. The crude product was used to the next step without additional purification. Compound 40b was obtained in 97% yield (396 mg; 1.56 mmol).


ESI-MS m/z for C13H20NO2S found 254.2 [M+H]+; Rt=1.00 min (Method A)


Step 3.
Synthesis of 4-(4,5-dimethylthiazol-2-yl)cyclohexan-1-one (40c)



embedded image


The solution of 40b (396 mg; 1.56 mmol) in 2 M HCl (15 mL) was stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, to this mixture aqueous solution of NaOH (4 M) was added to pH 12 and then extracted with DCM (2×). The combined organic solutions were dried over anhydrous MgSO4, filtered and concentrated in vacuo. The crude product was used to the next step without additional purification. Compound 40c was obtained in 95% yield (310 mg; 1.48 mmol).


ESI-MS m/z for C11H16NOS found 210.1 [M+H]+; Rt=0.84 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 2.64-2.58 (m, 1H), 2.49-2.45 (m, 2H), 2.35-2.30 (m, 2H), 2.30-2.24 (m, 5H), 2.20 (s, 3H), 1.94-1.85 (m, 2H).


Example 41
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((prop-2-yn-1-yloxy)methyl)morpholine hydrochloride (41)



embedded image


To the solution of an alcohol 17 (127 mg; 0.24 mmol) in DMF (2 mL) Cs2CO3 (234 mg; 0.72 mmol) and a solution of a propargyl bromide (80% in toluene; 32 μL; 0.29 mmol) were added and stirred at room temperature overnight, then at 55° C. for 3 hours. Then to this mixture NaH (60% in mineral oil; 6 mg; 0.25 mmol) was added and the reaction mixture was stirred at room temperature for 2 hours. The reaction progress was monitored by LC-MS. Then to this mixture water was added and then whole was diluted with DCM and washed with 10% Na2S2O3 and brine. An organic solution was dried over anhydrous MgSO4, filtered and concentrated in vacuo. The crude product was purified twice by preparative reversed-phase column chromatography (first: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 98:2 to 10:90, 30 min, 20 mL/min, second: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 30:70, 30 min, 20 mL/min). The title compound (41) was obtained as a hydrochloride salt as a single isomer in 21% yield (23 mg; 0.05 mmol).


ESI-MS m/z for C26H35ClN3O2 found 456.4/458.4 [M+H]+; Rt=1.06 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.5 Hz, 2H), 7.31 (d, JBB′=8.5 Hz, 2H), 5.87 (s, 1H), 4.20 (dd, J=2.4 Hz, J=0.7 Hz, 2H), 4.02-4.00 (m, 1H), 3.82-3.80 (m, 1H), 3.77-3.75 (m, 1H), 3.64-3.61 (m, 6H), 3.49-3.47 (m, 1H), 3.43-3.41 (m, 1H), 3.27 (t, J=2.4 Hz, 1H), 3.17-3.12 (m, 3H), 2.50-2.47 (m, 1H), 2.29-2.23 (m, 2H), 2.18 (s, 3H), 2.07-2.02 (m, 2H), 1.63-1.57 (m, 2H), 1.48-1.40 (m, 2H).


Example 42
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide hydrochloride (42)



embedded image


To the solution of 40 (22 mg; 0.045 mmol) in dry DCM (2 mL) diisopropylethylamine (DIPEA; 20 μL; 0.112 mmol), ethylamine hydrochloride (5 mg; 0.067 mmol) were added and after 10 minutes HATU (21 mg; 0.053 mmol) was added and the reaction mixture was stirred at room temperature for 4 hours. The reaction progress was monitored by TLC and LC-MS. When analyses indicated completion of the reaction, the mixture was concentrated in vacuo. The residue was dissolved in water/MeCN/HClaq, filtered off and taken for preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min). The title compound (42) was obtained as a hydrochloride salt as a single isomer in 64% yield (15 mg; 0.029 mmol).


ESI-MS m/z for C25H35ClN3O2S found 476.3/478.3 [M+H]+; Rt=1.12 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.39 (d, JAA′=8.6 Hz, 2H), 7.36 (d, JBB′=8.6 Hz, 2H), 4.37-4.36 (m, 1H), 3.85 (dd, J=13.2 Hz, J=2.1 Hz, 1H), 3.81-3.79 (m, 1H), 3.72-3.70 (m, 1H), 3.59-3.52 (m, 2H), 3.36-3.32 (m, 1H), 3.23-3.18 (m, 1H), 3.17-3.09 (m, 3H), 2.90 (tt, J=12.2 Hz, J=3.6 Hz, 1H), 2.29-2.23 (m, 5H), 2.20-2.15 (m, 5H), 1.72-1.62 (m, 2H), 1.58-1.51 (m, 2H), 1.07 (t, J=7.2 Hz, 3H).


Example 43
Synthesis of N-(35-(4-((((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methoxy)methyl)-1H-1,2,3-triazol-1-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxapentatriacontyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide hydrochloride (43)



embedded image


To the solution of 41 (27 mg; 0.055 mmol) in MeOH (1 mL) CuSO4×5 H2O (7 mg; 0.027 mmol) was added and after 5 minutes to a light green solution sodium L-ascorbate (11 mg; 0.055 mmol) was added in one portion. A whole was stirred for 20 minutes at room temperature and then a solution of an azide (40 mg; 0.050 mmol) in MeOH (1 mL) was added and the reaction mixture was stirred at room temperature for 3 hours. After analytical control (LC-MS) indicated completion of the reaction, the reaction mixture was concentrated in vacuo and the residue was dissolved in water/MeCN/HClaq, filtered off and taken for preparative reversed-phase column chromatography (first: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 10:90, 30 min, 20 mL/min, second: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 50:70, 30 min, 16 mL/min, third: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 50:50, 30 min, 16 mL/min). The title compound (43) was obtained as a hydrochloride salt as a single isomer in 28% yield (18 mg; 0.014 mmol).


ESI-MS m/z for C60H99ClN9O15S found 1252.2/1254.2 [M+H]+; Rt=1.09 min (Method A)


Example 44
Synthesis of (2R,5S)-2-(azidomethyl)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine hydrochloride (44)



embedded image


Step 1.
Synthesis of (2R,5S)-2-(azidomethyl)-5-(4-chlorobenzyl)morpholine (44a)



embedded image


The title compound (44a) was obtained as a free amine in 97% yield (1.5 g; 5.64 mmol) from 27a (2.13 g; 5.81 mmol) according to the General Procedure IVa and then after basic (NaOH) extraction with DCM.


ESI-MS m/z for C12H16ClN4O found 267.0/268.9 [M+H]+; Rt=0.72 min (Method A)


Step 2.
Synthesis of (2R,5S)-2-(azidomethyl)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine hydrochloride (44)



embedded image


The title compound (44) was obtained as a single isomer as a free amine from 44a (800 mg; 3.00 mmol) and ketone 2d (635 mg; 3.30 mmol) according to the General Procedure VII in 46% yield (612 mg; 1.38 mmol). The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 9:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 200:1 to 9:1, v/v). The sample of this product (22 mg) was then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min) to give 10 mg of the title product as a hydrochloride salt.


ESI-MS m/z for C23H32ClN6O found 443.2/445.2 [M+H]+; Rt=1.12 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.40 (d, JAA′=8.5 Hz, 2H), 7.30 (d, JBB′=8.5 Hz, 2H), 5.88 (s, 1H), 3.97-3.94 (m, 1H), 3.79-3.77 (m, 1H), 3.74-3.72 (m, 1H), 3.62-3.54 (m, 6H), 3.43-3.36 (m, 2H), 3.21-3.17 (m, 1H), 3.11-3.10 (m, 2H), 2.49-2.44 (m, 1H), 2.27-2.25 (m, 2H), 2.17 (s, 3H), 2.02-2.00 (m, 2H), 1.52-1.38 (m, 4H).


Example 45
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-(prop-2-yn-1-yl)morpholine-2-carboxamide hydrochloride (45)



embedded image


The title compound (45) was obtained as a hydrochloride salt as a single isomer in 2% overall yield in a similar way to Example 30 with the exception that, in the first step of the synthesis, propargylamine was used instead of propan-2-amine. The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 100:0 to 0:100, v/v, then: AcOEt/MeOH, 10:1, v/v) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 99:1 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C26H34ClN4O2 found 469.2/471.2 [M+H]+; Rt=0.96 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.39 (d, JAA′=8.5 Hz, 2H), 7.35 (d, JBB′=8.5 Hz, 2H), 5.83 (s, 1H), 4.44-4.42 (m, 1H), 3.97 (dd, J=17.4 Hz, J=2.5 Hz, 1H), 3.91 (dd, J=17.4 Hz, J=2.5 Hz, 1H), 3.87 (dd, J=13.2 Hz, J=2.3 Hz, 1H), 3.81-3.79 (m, 1H), 3.73-3.71 (m, 1H), 3.61 (s, 3H), 3.60-3.58 (m, 1H), 3.51-3.47 (m, 1H), 3.36-3.32 (m, 1H), 3.14-3.09 (m, 2H), 2.91 (t, J=2.5 Hz, 1H), 2.50-2.46 (m, 1H), 2.24-2.17 (m, 5H), 2.06-2.01 (m, 2H), 1.67-1.57 (m, 2H), 1.46-1.38 (m, 2H).


Example 46
Synthesis of N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1H-pyrazole-3-carboxamide hydrochloride (46)



embedded image


Step 1.
Synthesis of ((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methanamine (46a)



embedded image


An azide 44 (as a free amine)(590 mg; 1.33 mmol) was dissolved in THF/H2O (20 mL; 9:1, v/v) and then 1 M solution of Me3P in THF (2 mL; 2.00 mmol) was added and a whole was stirred at room temperature for 2 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the reaction was quenched with 5% NaHCO3 and a whole was extracted with diethyl ether (2×). The combined organic solutions were washed with water and then dried over anhydrous MgSO4, filtered and concentrated in vacuo. The all crude product mixture was used to the next step without additional purification.


ESI-MS m/z for C23H34ClN4O found 417.1/419.1 [M+H]+; Rt=0.76 min (Method A)


Step 2.
Synthesis of tert-butyl (((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)carbamate (46b)



embedded image


The title compound (46b) was obtained in 74% yield (504 mg; 0.98 mmol) from 46a (the crude mixture) according to the General Procedure X.


ESI-MS m/z for C28H42ClN4O3 found 517.2/519.2 [M+H]+; Rt=1.08 min (Method A)


Step 3.
Synthesis of ((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methanamine (46c)



embedded image


The title compound (46c) was obtained as a free amine in 87% yield (355 mg; 0.85 mmol) from 46b (504 mg; 0.98 mmol) according to the General Procedure IVa and then after basic (NaOH) extraction with DCM.


ESI-MS m/z for C23H34ClN4O found 417.2/419.1 [M+H]+; Rt=1.05 min (Method A)


Step 4.
Synthesis of N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1H-pyrazole-3-carboxamide hydrochloride (46)



embedded image


To the solution of 46c (50 mg; 0.120 mmol) in dry DCM (2 mL) diisopropylethylamine (DIPEA; 0.03 mL; 0.180 mmol), 1H-pyrazole-3-carboxylic acid (14 mg; 0.126 mmol) and HATU (48 mg; 0.126 mmol) were added and the reaction mixture was stirred at room temperature overnight. The reaction progress was monitored by TLC and LC-MS. When analyses indicated completion of the reaction, the mixture was concentrated in vacuo. The crude product was purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 98:2 to 10:90, 30 min, 20 mL/min). The title compound (46) was obtained as a hydrochloride salt as a single isomer in 37% yield (24 mg; 0.044 mmol).


ESI-MS m/z for C27H36ClN6O2 found 511.3/513.3 [M+H]+; Rt=0.88 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.72 (d, J=2.3 Hz, 1H), 7.37 (d, JAA′=8.5 Hz, 2H), 7.31 (d, JBB′=8.5 Hz, 2H), 6.70 (d, J=2.3 Hz, 1H), 5.86 (s, 1H), 4.01-3.99 (m, 1H), 3.79-3.75 (m, 2H), 3.64-3.62 (m, 4H), 3.54-3.51 (m, 4H), 3.16-3.09 (m, 3H), 2.51-2.46 (m, 1H), 2.28-2.23 (m, 2H), 2.18 (s, 3H), 2.06-2.02 (m, 2H), 1.60-1.55 (m, 2H), 1.47-1.40 (m, 2H).


Example 47
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-((1-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)-1H-1,2,3-triazol-4-yl)methyl)morpholine-2-carboxamide hydrochloride (47)



embedded image


To the solution of 45 (59 mg; 0.117 mmol) in MeOH (2 mL) CuSO4×5 H2O (15 mg; 0.060 mmol) was added and after 5 minutes to a light green solution sodium L-ascorbate (23 mg; 0.117 mmol) was added in one portion. A whole was stirred for 25 minutes at room temperature and then a solution of an azide (93 mg; 0.117 mmol) in MeOH (2 mL) was added and the reaction mixture was stirred at room temperature overnight. After analytical control (LC-MS) indicated completion of the reaction, the reaction mixture was concentrated in vacuo and the residue was dissolved in water/MeCN/HClaq, filtered off and taken for preparative reversed-phase column chromatography (first: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 98:2 to 10:90, 30 min, 20 mL/min, second: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 99:1 to 20:80, 30 min, 18 mL/min, third: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 99:1 to 10:90, 30 min, 18 mL/min). The title compound (47) was obtained as a hydrochloride salt as a single isomer in 20% yield (31 mg; 0.023 mmol).


ESI-MS m/z for C60H98ClN10O5S found 1265.7/1267.7 [M+H]+; Rt=1.06 min (Method A)


Example 48
Synthesis of N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-2H-1,2,3-triazole-4-carboxamide hydrochloride (48)



embedded image


To the solution of 46c (50 mg; 0.120 mmol) in dry DCM (2 mL) diisopropylethylamine (DIPEA; 0.03 mL; 0.180 mmol), 2H-1,2,3-triazole-4-carboxylic acid (15 mg; 0.126 mmol) and HATU (48 mg; 0.126 mmol) were added and the reaction mixture was stirred at room temperature overnight. The reaction progress was monitored by TLC and LC-MS. When analyses indicated completion of the reaction, the mixture was concentrated in vacuo. The crude product was purified twice by preparative reversed-phase column chromatography (first: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 99:1 to 20:80, 30 min, 18 mL/min, second: C-18, water+0.3‰ HCl (36%)/MeCN, 98:2 to 30:70, 30 min, 18 mL/min). The title compound (48) was obtained as a hydrochloride salt as a single isomer in 9% yield (6 mg; 0.011 mmol).


ESI-MS m/z for C26H35ClN7O2 found 512.2/514.2 [M+H]+; Rt=0.88 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 8.28 (brs, 1H), 7.38 (d, JAA′=8.5 Hz, 2H), 7.31 (d, JBB′=8.5 Hz, 2H), 5.84 (s, 1H), 4.02-4.00 (m, 1H), 3.79-3.76 (m, 2H), 3.65-3.63 (m, 1H), 3.61 (s, 3H), 3.56-3.52 (m, 3H), 3.46-3.44 (m, 1H), 3.16-3.10 (m, 3H), 2.50-2.45 (m, 1H), 2.29-2.23 (m, 2H), 2.17 (s, 3H), 2.06-2.02 (m, 2H), 1.60-1.54 (m, 2H), 1.47-1.40 (m, 2H).


Example 49
Synthesis of 3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)imidazolidine-2,4-dione hydrochloride (49)



embedded image


To the solution of an alcohol 17 (133 mg; 0.25 mmol) and hydantoin (30 mg; 0.30 mmol) in THF (3 mL) PPh3 (158 mg; 0.60 mmol) was added. Then a whole was cooled to −10° C. and DIAD (65 μL; 0.33 mmol) was added dropwise and stirred at room temperature for 2 days. Then to this mixture another portion of hydantoin (30 mg; 0.30 mmol), PPh3 (79 mg; 0.30 mmol) and DIAD (65 μL; 0.33 mmol) were added at −10° C. and the reaction mixture was stirred at room temperature overnight. The reaction progress was monitored by LC-MS. Then a solid was filtered off and a light yellow filtrate was concentrated in vacuo. The residue was dissolved in water/MeCN/HClaq, filtered off and taken for preparative reversed-phase column chromatography (first: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 98:2 to 30:70, 30 min, 20 mL/min, second: C-18, water+0.3‰ HCl (36%)/MeCN, 98:2 to 20:80, 30 min, 20 mL/min). The title compound (49) was obtained as a hydrochloride salt as a single isomer in 40% yield (53 mg; 0.10 mmol).


ESI-MS m/z for C26H35ClN5O3 found 500.2/502.2 [M+H]+; Rt=0.87 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.37 (d, JAA′=8.5 Hz, 2H), 7.29 (d, JBB′=8.5 Hz, 2H), 5.82 (s, 1H), 4.07-4.04 (m, 1H), 3.92 (s, 2H), 3.76-3.74 (m, 1H), 3.71-3.67 (m, 2H), 3.62-3.60 (m, 4H), 3.55-3.50 (m, 3H), 3.17-3.08 (m, 3H), 2.47 (tt, J=12.3 Hz, J=3.6 Hz, 1H), 2.26-2.20 (m, 2H), 2.16 (s, 3H), 2.06-2.01 (m, 2H), 1.57-1.51 (m, 2H), 1.45-1.39 (m, 2H).


Example 50
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide hydrochloride (50)



embedded image


The title compound (50) was obtained as a hydrochloride salt as a single isomer in 7% overall yield in a similar way to Example 30 with the exception that, in the first step of the synthesis, ethylamine hydrochloride was used instead of propan-2-amine and in the third step of the synthesis, ketone 15c was used instead of ketone 2d. The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 1:1, v/v, then AcOEt, 100%, then: AcOEt/MeOH, 50:1 to 6:1, v/v) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 18 mL/min).


ESI-MS m/z for C24H35ClN5O2 found 460.1/462.1 [M+H]+; Rt=0.77 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.5 Hz, 2H), 7.34 (d, JBB′=8.5 Hz, 2H), 4.39-4.37 (m, 1H), 3.87 (dd, J=13.2 Hz, J=2.3 Hz, 1H), 3.80-3.78 (m, 1H), 3.73-3.71 (m, 4H), 3.60-3.58 (m, 1H), 3.53-3.49 (m, 1H), 3.35-3.32 (m, 1H), 3.22-3.09 (m, 4H), 2.67 (tt, J=12.2 Hz, J=3.6 Hz, 1H), 2.41 (s, 3H), 2.28-2.22 (m, 2H), 2.16-2.11 (m, 2H), 1.71-1.62 (m, 2H), 1.58-1.50 (m, 2H) 1.07 (t, J=7.2 Hz, 3H).


Example 51
Synthesis of N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1H-imidazole-4-carboxamide hydrochloride (51)



embedded image


To the solution of 1H-imidazole-4-carboxylic acid (14 mg; 0.120 mmol) in dry DCM/DMF (2 mL; 1:1 v/v) an oxalyl chloride (11 μL; 0.130 mmol) was added (gas evolution). The reaction mixture was stirred at room temperature for 1 hour and then to this mixture a solution of an amine 46c (50 mg; 0.120 mmol) in dry DCM (3 mL) and triethylamine (0.05 mL; 0.360 mmol) were added and the reaction mixture was stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the mixture was concentrated in vacuo. The crude product was purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 98:2 to 10:90, 30 min, 19 mL/min). The title compound (51) was obtained as a hydrochloride salt as a single isomer in 26% yield (17 mg; 0.031 mmol).


ESI-MS m/z for C27H36ClN6O2 found 511.3/513.3 [M+H]+; Rt=0.82 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 8.58 (s, 1H), 8.01 (d, J=0.9 Hz, 1H), 7.38 (d, JAA′=8.5 Hz, 2H), 7.31 (d, JBB′=8.5 Hz, 2H), 5.86 (s, 1H), 4.04-4.02 (m, 1H), 3.84-3.83 (m, 1H), 3.78-3.76 (m, 1H), 3.64-3.62 (m, 4H), 3.56-3.49 (m, 3H), 3.47-3.43 (m, 1H), 3.17-3.10 (m, 3H), 2.51-2.47 (m, 1H), 2.29-2.24 (m, 2H), 2.18 (s, 3H), 2.07-2.02 (m, 2H), 1.66-1.59 (m, 2H), 1.47-1.40 (m, 2H).


Example 52
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohex-3-en-1-yl)-2-((methylsulfonyl)methyl)morpholine 2,2,2-trifluoroacetate (52)



embedded image


Step 1.
Synthesis of 4-(4,5-dimethylthiazol-2-yl)cyclohex-3-en-1-one (52a)



embedded image


The solution of 40a (861 mg; 3.40 mmol) in 1 M KHSO4aq (60 mL) was stirred at 30° C. for 5 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, to this mixture aqueous solution of NaOH (4 M) was added to pH 12 and then extracted with DCM (2×). The combined organic solutions were dried over anhydrous MgSO4, filtered and concentrated in vacuo. The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 45:55, v/v, 35 min). Compound 52a was obtained in 44% yield (310 mg; 1.50 mmol).


ESI-MS m/z for C11H14NOS found 208.1 [M+H]+; Rt=1.10 min (Method A)


Step 2.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohex-3-en-1-yl)-2-((methylsulfonyl)methyl)morpholine 2,2,2-trifluoroacetate (52)



embedded image


The title compound (52) was obtained as a TFA salt from 3h (85 mg; 0.25 mmol) and ketone 52a (50 mg; 0.24 mmol) according to the General Procedure VII in 37% yield (56 mg; 0.09 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 35:65, v/v) and then purified twice by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 10:90, 30 min, 20 mL/min).


ESI-MS m/z for C24H32ClN2O3S2 found 495.0/497.0 [M+H]+; Rt=1.34 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 7.39-7.37 (m, 2H), 7.34-7.32 (m, 2H), 6.38-6.36 (m, 1H), 4.29-4.24 (m, 1H), 3.82-3.63 (m, 4H), 3.61-3.56 (m, 1.5H), 3.51-3.49 (m, 0.5H), 3.38-3.37 (m, 1H), 3.31-3.26 (m, 1H), 3.19 (td, J=13.8 Hz, J=10.9 Hz, 1H), 3.13-3.09 (m, 1H), 3.03 (s, 3H), 2.81-2.72 (m, 2H), 2.48-2.33 (m, 3H), 2.29 (s, 3H), 2.22 (s, 3H), 1.74-1.66 (m, 1H).


Example 53
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohex-3-en-1-yl)-N-ethylmorpholine-2-carboxamide 2,2,2-trifluoroacetate (53)



embedded image


The title compound (53) was obtained as a TFA salt in 24% overall yield in a similar way to Example 30 with the exception that, in the first step of the synthesis, ethylamine hydrochloride was used instead of propan-2-amine and in the third step of the synthesis, ketone 52a was used instead of ketone 2d. The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 10:90, v/v, 30 min) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 98:2 to 10:90, 30 min, 20 mL/min).


ESI-MS m/z for C25H33ClN3O2S found 474.2/476.2 [M+H]+; Rt=1.25 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.39-7.34 (m, 4H), 6.37-6.36 (m, 1H), 4.30-4.27 (m, 1H), 3.81-3.74 (m, 2H), 3.71-3.66 (m, 2H), 3.61-3.52 (m, 1H), 3.34-3.30 (m, 1H), 3.23-3.11 (m, 4H), 2.80-2.71 (m, 2H), 2.48-2.38 (m, 2H), 2.36-2.29 (m, 4H), 2.21 (brs, 3H), 1.77-1.67 (m, 1H), 1.08-1.06 (m, 3H).


Example 54
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)morpholine-2-carboxamide hydrochloride (54)



embedded image


The title compound (54) was obtained as a single isomer as a hydrochloride salt from 40 (17 mg; 0.035 mmol) according to the General Procedure XI in 77% yield (34 mg; 0.027 mmol). The crude product was purified twice by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 18 mL/min).


ESI-MS m/z for C57H94ClN6O15S2 found 1201.7/1203.7 [M+H]+; Rt=1.22 min (Method A)


Example 55
Synthesis of (2R,5S)-2-(azidomethyl)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine hydrochloride (55)



embedded image


The title compound (55) was obtained as a single isomer as a free amine from 44a (682 mg; 2.56 mmol) and ketone 40c (563 mg; 2.69 mmol) according to the General Procedure VII in 42% yield (500 mg; 1.09 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 0:100, v/v, 40 min). The sample of this product (25 mg) was then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 10:90.30 min, 20 mL/min) to give 9 mg of the title product as a hydrochloride salt.


ESI-MS m/z for C23H31ClN5OS found 460.3/462.3 [M+H]+; Rt=1.31 min (Method A); 1H NMR (700 MHz, D2O, 333 K) δ 7.86 (d, JAA′=8.3 Hz, 2H), 7.74 (d, JBB′=8.3 Hz, 2H), 4.52-4.50 (m, 1H), 4.35-4.29 (m, 2H), 4.24-4.21 (m, 1H), 4.12 (dd, J=13.7 Hz, J=3.3 Hz, 1H), 4.04-3.99 (m, 2H), 3.91 (dd, J=13.7 Hz, J=5.3 Hz, 1H), 3.81-3.78 (m, 1H), 3.71-3.67 (m, 1H), 3.63-3.58 (m, 2H), 2.88-2.87 (m, 2H), 2.78-2.75 (m, 5H), 2.74 (s, 3H), 2.15-2.04 (m, 4H).


Example 56
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-((1-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)-1H-1,2,3-triazol-4-yl)methyl)morpholine-2-carboxamide hydrochloride (56)



embedded image


Step 1.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-(prop-2-yn-1-yl)morpholine-2-carboxamide (56a)



embedded image


To the solution of 40 (22 mg; 0.045 mmol) in dry DCM (2 mL) diisopropylethylamine (DIPEA; 23 μL; 0.135 mmol), propargylamine (4.5 μL; 0.068 mmol) were added and after 5 minutes HATU (21 mg; 0.055 mmol) was added and the reaction mixture was stirred at room temperature overnight. The reaction progress was monitored by TLC and LC-MS. When analyses indicated completion of the reaction, the mixture was concentrated in vacuo. The crude product was used to the next step without additional purification.


ESI-MS m/z for C26H33ClN3O2S found 486.0/488.0 [M+H]+; Rt=1.25 min (Method A)


Step 2.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-((1-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)-1H-1,2,3-triazol-4-yl)methyl)morpholine-2-carboxamide hydrochloride (56)



embedded image


To the solution of 56a (the crude product) in MeOH (2 mL) CuSO4×5 H2O (6 mg; 0.022 mmol) was added and after 5 minutes to a light green solution sodium L-ascorbate (9 mg; 0.045 mmol) was added in one portion. A whole was stirred for 25 minutes at room temperature and then a solution of an azide (36 mg; 0.045 mmol) in MeOH (2 mL) was added and the reaction mixture was stirred at room temperature for 3 hours. After analytical control (LC-MS) indicated completion of the reaction, the reaction mixture was concentrated in vacuo and the residue was dissolved in water/MeCN/HClaq, filtered off and taken for preparative reversed-phase column chromatography (first: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min, second: C-18, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min). The title compound (56) was obtained as a hydrochloride salt as a single isomer in 27% yield (per two steps)(16 mg; 0.012 mmol).


ESI-MS m/z for C60H97ClN9O15S2 found 1282.2/1284.2 [M+H]+; Rt=1.23 min (Method A)


Example 57
Synthesis of N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methyl)-1H-1,2,3-triazole-5-carboxamide hydrochloride (57)



embedded image


The title compound (57) was obtained as a hydrochloride salt as a single isomer in 3% overall yield in a similar way to Example 46 with the exception that, in the first step of the synthesis, the compound 55 was used instead of the compound 44 and in the fourth step of the synthesis, 2H-1,2,3-triazole-4-carboxylic acid was used instead of 1H-pyrazole-3-carboxylic acid. The crude product was purified twice by preparative reversed-phase column chromatography (first: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 98:2 to 30:70, 30 min, 20 mL/min, second: C-18, water+0.3‰ HCl (36%)/MeCN, 98:2 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C26H34ClN6O2S found 529.1/531.1 [M+H]+; Rt=1.17 min (Method A); 1H NMR (700 MHz, DMSO-d6) δ 10.60 (brs, 1H), 8.63-8.61 (m, 1H), 7.43 (d, JAA′=8.4 Hz, 2H), 7.35 (d, JBB′=8.4 Hz, 2H), 4.04-4.00 (m, 1H), 3.82-3.80 (m, 2H), 3.62-3.58 (m, 2H), 3.54-3.48 (m, 3H), 3.14-3.07 (m, 3H), 2.88 (tt, J=12.0 Hz, J=3.6 Hz, 1H), 2.33-2.28 (m, 5H), 2.21-2.14 (m, 5H), 1.70-1.59 (m, 3H), 1.53-1.47 (m, 1H).


Example 58
Synthesis of N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1-methyl-1H-imidazole-4-carboxamide hydrochloride (58)



embedded image


To the solution of 1-methyl-1H-imidazole-4-carboxylic acid (25 mg; 0.190 mmol) in dry DCM/DMF (2 mL; 1:1 v/v) an oxalyl chloride (18 μL; 0.210 mmol) was added (gas evolution). The reaction mixture was stirred at room temperature for 1 hour and then to this mixture a solution of an amine 46c (80 mg; 0.190 mmol) in dry DCM (3 mL) and triethylamine (0.08 mL; 0.600 mmol) were added and the reaction mixture was stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the mixture was concentrated in vacuo. The crude product was purified twice by preparative reversed-phase column chromatography (first: C-18, water+0.3‰ HCl (36%)/MeCN, 98:2 to 10:90, 30 min, 20 mL/min, second: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 98:2 to 30:70, 30 min, 20 mL/min). The title compound (58) was obtained as a hydrochloride salt as a single isomer in 11% yield (12 mg; 0.021 mmol).


ESI-MS m/z for C28H38ClN6O2 found 525.4/527.4 [M+H]+; Rt=1.03 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 8.01 (brs, 1H), 7.76 (s, 1H), 7.37 (d, JAA′=8.5 Hz, 2H), 7.31 (d, JBB′=8.5 Hz, 2H), 5.83 (s, 1H), 3.99-3.97 (m, 1H), 3.78-3.73 (m, 5H), 3.64-3.60 (m, 4H), 3.53-3.49 (m, 3H), 3.45-3.42 (m, 1H), 3.15-3.08 (m, 3H), 2.50-2.45 (m, 1H), 2.28-2.22 (m, 2H), 2.17 (s, 3H), 2.05-2.01 (m, 2H), 1.60-1.54 (m, 2H), 1.46-1.39 (m, 2H).


Examples 59 and 60
Synthesis of methyl 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)acetate hydrochloride (59) and 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)acetic acid hydrochloride (60)



embedded image


Step 1.
Synthesis of tert-butyl (2S,5S)-5-(4-chlorobenzyl)-2-(cyanomethyl)morpholine-4-carboxylate (59a)



embedded image


To a solution of 3e (1.5 g; 4.39 mmol) in an anhydrous DMSO (10 mL) KCN (530 mg; 4.61 mmol) was added and the reaction mixture was stirred at 85° C. for 3 hours and then at room temperature overnight. The reaction progress was monitored by LC-MS analysis. When analysis indicated completion of the reaction, the reaction mixture was diluted with water and extracted with Et2O (3×50 mL). The combined organic solutions were dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 75:25, v/v, 20 min). Compound 59a was obtained in 75% yield (1.16 g; 3.31 mmol).


ESI-MS m/z for C13H16ClN2O found 251.0/253.0 [M+H−Boc]+; Rt=6.35 min (Method C)


Step 2.
Synthesis of 2-((2S,5S)-4-(tert-butoxycarbonyl)-5-(4-chlorobenzyl)morpholin-2-yl)acetic acid (59b)



embedded image


To a solution of 59a (830 mg; 2.37 mmol) in AcOH (10 mL) a concentrated H2SO4 (5 mL) and water (5 mL) were added and the reaction mixture was stirred at 90° C. overnight. The reaction progress was monitored by LC-MS analysis. When analysis indicated completion of the reaction, the reaction mixture was cooled down to room temperature and alkalized to pH 11-12 with 4 M KOHaq. Then to this solution a solution of Boc2O (1.2 g; 5.50 mmol) in THF (50 mL) was added and stirred at room temperature for 4 hours. Then another portion of Boc2O (1.2 g; 5.50 mmol) in THF (50 mL) was added and stirred at room temperature for 2 days. When analysis indicated completion of the reaction, the reaction mixture was acidified to pH 2-3 with 2 M HClq and extracted with AcOEt. The combined organic solutions were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was used to the next step without additional purification. Compound 59b was obtained in 99% yield (867 mg; 2.35 mmol).


ESI-MS m/z for C18H23ClNO5 found 368.0/370.0 [M−H]; Rt=5.92 min (Method C)


Step 3.
Synthesis of methyl 2-((2S,5S)-5-(4-chlorobenzyl)morpholin-2-yl)acetate (59c)



embedded image


The title compound (59c) was obtained as a free amine in 43% yield (250 mg; 0.88 mmol) from 59b (750 mg; 2.03 mmol) according to the General Procedure IVa and then after basic (5% NaHCO3) extraction with DCM.


ESI-MS m/z for C14H19ClNO3 found 284.0/286.0 [M+H]+; Rt=3.53 min (Method C)


Step 4.
Synthesis of methyl 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)acetate (59d)



embedded image


The title compound (59d) was obtained as a TFA salt from 59c (155 mg; 0.55 mmol) and ketone 2d (105 mg; 0.55 mmol) according to the General Procedure VII in 22% yield (70 mg; 0.12 mmol). The crude product was purified by silica-gel column chromatography (AcOEt, 100%, then: AcOEt/MeOH, 95:45, v/v) and then by preparative reversed-phase column chromatography (Phenyl-Hexyl, water/MeCN+1‰ TFA, 95:5 to 5:95, 60 min, 15 mL/min).


ESI-MS m/z for C25H35ClN3O3 found 460.0/462.0 [M+H]+; Rt=4.37 min (Method C)


Step 5.
Synthesis of methyl 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)acetate hydrochloride (59) and 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)acetic acid hydrochloride (60)



embedded image


To the solution of 59d (70 mg; 0.120 mmol) in MeOH (3 mL) 1 M NaOH (0.5 mL) was added and a whole was stirred at room temperature for 1 hour. The reaction progress was monitored by LC-MS. Then the solvent was evaporated in vacuo and the residue was purified by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water/MeCN+1‰ TFA, 95:5 to 5:95, 30 min, 20 mL/min). Then the obtained products were transformed into hydrochloride salts with using HCl/AcOEt (2.6 M; 0.5 mL). The obtained solids of the hydrochloride salts were then lyophilized. The title compounds (59 and 60) were obtained as separated single isomers as hydrochloride salts in 10% yield (for 59)(6 mg; 0.012 mmol) and in 53% yield (for 60)(31 mg; 0.064 mmol).


For 59: ESI-MS m/z for C25H35ClN3O3 found 460.2/462.2 [M+H]+; Rt=4.42 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.41-7.26 (m, 4H), 6.45 (s, 1H), 4.38-4.22 (m, 1H), 3.95 (s, 3H), 3.90-3.80 (m, 2H), 3.76-3.66 (m, 5H), 3.63-3.53 (m, 1H), 3.40-3.33 (m, 1H), 3.28-3.11 (m, 2H), 2.95-2.83 (m, 1H), 2.81-2.67 (m, 2H), 2.56-2.37 (m, 5H), 2.30-2.20 (m, 2H), 1.90-1.63 (m, 4H).


For 60: ESI-MS m/z for C24H33ClN3O3 found 446.2/448.2 [M+H]+; Rt=4.03 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.40-7.29 (m, 4H), 6.48-6.40 (m, 1H), 4.34-4.19 (m, 1H), 3.95 (s, 3H), 3.90-3.83 (m, 2H), 3.78-3.69 (m, 2H), 3.64-3.55 (m, 1H), 3.40-3.33 (m, 1H), 3.30-3.14 (m, 2H), 2.94-2.81 (m, 1H), 2.78-2.61 (m, 2H), 2.59-2.45 (m, 2H), 2.42 (s, 3H), 2.31-2.16 (m, 2H), 1.85-1.63 (m, 4H).


Example 61
Synthesis of 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)-N-ethylacetamide hydrochloride (61)



embedded image


The title compound (61) was obtained as a hydrochloride salt as a single isomer in 2% overall yield in a similar way to Example 30 with the exception that, in the first step of the synthesis, ethylamine hydrochloride was used instead of propan-2-amine and the compound 59b was used instead of the compound 5a and in the second step of the synthesis, 1 M NaOH was used instead of 5% NaHCO3. The crude product was purified by flash column chromatography on silica (AcOEt, 100%, then: AcOEt/MeOH, 80:20, v/v) and then purified by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water/MeCN+1‰ TFA, 95:5 to 5:95, 30 min, 20 mL/min). Then the obtained product was transformed into hydrochloride salt with using HCl/AcOEt (3.6 M; 0.2 mL). The obtained solid of the hydrochloride salt was then lyophilized.


ESI-MS m/z for C26H38ClN4O2 found 473.2/475.2 [M+H]+; Rt=4.06 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.43-7.25 (m, 4H), 6.55-6.43 (m, 1H), 4.32-4.18 (m, 1H), 3.97 (s, 3H), 3.90-3.80 (m, 2H), 3.77-3.66 (m, 2H), 3.64-3.55 (m, 1H), 3.43-3.32 (m, 1H), 3.28-3.13 (m, 4H), 2.95-2.85 (m, 1H), 2.62-2.41 (m, 7H), 2.34-2.18 (m, 2H), 1.89-1.65 (m, 4H), 1.14 (t, J=7.3 Hz, 3H).


Example 62
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid hydrochloride (62)



embedded image


The title compound (62) was obtained as a hydrochloride salt as a single isomer in 1% overall yield in a similar way to Example 9 with the exception that, in the third step of the synthesis, ketone 15c was used instead of ketone 1g. The crude product was purified by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 40 min, 12 mL/min). Then the obtained product was transformed into hydrochloride salt with using of 1 M HCl (2 mL). The obtained solid of the hydrochloride salt was then lyophilized.


ESI-MS m/z for C22H30ClN4O3 found 433.2/435.2 [M+H]+; Rt=3.40 min (Method C); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.40-7.36 (m, 2H), 7.33-7.30 (m, 2H), 4.52-4.44 (m, 1H), 3.80-3.76 (m, 1H), 3.76-3.71 (m, 2H), 3.68 (s, 3H), 3.56-3.52 (m, 1H), 3.50-3.44 (m, 2H), 3.16-3.10 (m, 1H), 3.10-3.00 (m, 1H), 2.64-2.58 (m, 1H), 2.35 (s, 3H), 2.23-2.06 (m, 4H), 1.68-1.42 (m, 4H).


Example 63
Synthesis of (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (63)



embedded image


Step 1.
Synthesis of (2S)-2-amino-3-(4-bromophenyl)propan-1-ol (63a)



embedded image


The title compound (63a) was obtained from optically pure L-p-bromophenylalanine ((2S)-2-amino-3-(4-bromophenyl)propanoic acid) (2.8 g; 11.47 mmol) according to the General Procedure Ia in 87% yield (2.6 g; 11.36 mmol).


Step 2.
Synthesis of (R)-2-bromo-N—((S)-1-(4-bromophenyl)-3-hydroxypropan-2-yl)-3-(tert-butoxy)-propanamide (63b)



embedded image


The title compound (63b) was obtained from the compound 63a (7.8 g; 34.0 mmol) and (R)-2-bromo-3-(tert-butoxy)propanoic acid (3a′) (9.5 g; 42.0 mmol) according to the General Procedure V in 99% yield (14.7 g; 33.7 mmol).


Step 3.
Synthesis of (2S,5S)-5-(4-bromobenzyl)-2-(tert-butoxymethyl)morpholin-3-one (63c)



embedded image


The title compound (63c) was obtained from 63b (16.05 g, 37.0 mmol) according to the General Procedure II in 51% yield (6.7 g; 18.8 mmol).


ESI-MS m/z for C12H15BrNO3 found 300.0/302.0 [M+H−tBu]+; Rt=5.75 min (Method C)


Step 4.
Synthesis of ((2R,5S)-5-(4-bromobenzyl)morpholin-2-yl)methanol (63d)



embedded image


The title compound (63d) was obtained from 63c (5.7 g; 16.1 mmol) according to the General Procedure Ib in 68% yield (3.1 g; 10.9 mmol).


ESI-MS m/z for C12H17BrNO2 found 285.7/287.7 [M+H]+; Rt=2.96 min (Method C)


Step 5.
Synthesis of tert-butyl (2R,5S)-5-(4-bromobenzyl)-2-(tert-butoxymethyl)morpholine-4-carboxylate (63e)



embedded image


To a solution of amino alcohol 63d (2.25 g, 7.86 mmol) in dichloromethane (80 mL), di-tert-butyl dicarbonate (Boc2O) (1.62 g, 7.47 mmol) was added and the reaction mixture was stirred at room temperature overnight, after which time TLC showed almost complete consumption of the starting material. The volatiles were removed in vacuo and the residue was purified by silica-gel column chromatography (DCM/MeOH, 100:1, v/v). Compound 63e was obtained in 82% yield (2.48 g; 6.44 mmol).


ESI-MS m/z for C12H17BrNO2 found 286.0/288.0 [M+H−Boc]+; Rt=5.81 min (Method C); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.44-7.39 (m, 2H), 7.15-7.10 (m, 2H), 4.08-4.00 (m, 1H), 3.84-3.75 (m, 1H), 3.72-3.66 (m, 1H), 3.53-3.46 (m, 2H), 3.46-3.40 (m, 2H), 3.36-3.30 (m, 1H), 2.96-2.84 (m, 2H), 2.84-2.77 (m, 1H), 1.22 (s, 9H).


Step 6.
Synthesis of tert-butyl (2R,5S)-5-(4-bromobenzyl)-2-(((methylsulfonyl)oxy)methyl)morpholine-4-carboxylate (63f)



embedded image


The title compound (63f) was obtained from 63e (965 mg; 2.5 mmol) according to the General Procedure VIII in 98% yield (1.14 g; 2.46 mmol).


ESI-MS m/z for C18H26BrNO6SNa found 485.9/487.9 [M+Na]+; Rt=6.35 min (Method C)


Step 7.
Synthesis of tert-butyl (2R,5S)-5-(4-bromobenzyl)-2-((methylthio)methyl)morpholine-4-carboxylate (63g)



embedded image


To a solution of 63f (0.9 g; 1.94 mmol) in acetonitrile (3.5 mL) MeSNa (365 mg; 5.21 mmol) was added and the reaction mixture was stirred at 105° C. overnight. The reaction progress was monitored by LC-MS analysis. After analytical control indicated completion of the reaction, the mixture was dissolved in water (5.6 mL) and MeCN was removed in vacuo. An aqueous phase was extracted with DCM (4×). The combined organic solutions were washed with 1 M NaOH and brine, dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was used to the next step without additional purification. Compound 63g was obtained in 87% yield (0.7 g; 1.69 mmol).


ESI-MS m/z for C13H19BrNOS found 316.0/318.0 [M+H−Boc]+; Rt=6.65 min (Method C)


Step 8.
Synthesis of tert-butyl (2R,5S)-5-(4-bromobenzyl)-2-((methylsulfonyl)methyl)morpholine-4-carboxylate (63h)



embedded image


To a solution of 63g (0.7 g; 1.69 mmol) in DCM (9.9 mL) mCPBA (1.39 g; 8.0 mmol) was added portionwise at 0° C. Then the cooling bath was removed and the mixture was stirred at room temperature overnight. The reaction progress was monitored by LC-MS analysis. When analysis indicated completion of the reaction, the reaction was quenched with 10% Na2SO3 and stirred for 30 minutes at room temperature. Then phases were separated and the organic one was washed with 1 M NaOH (4×) and brine, dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was used to the next step without additional purification. Compound 63h was obtained in 86% yield (0.65 g; 1.45 mmol).


ESI-MS m/z for C13H19BrNO3S found 348.0/350.0 [M+H−Boc]+; Rt=6.19 min (Method C)


Step 9.
Synthesis of (2R,5S)-5-(4-bromobenzyl)-2-((methylsulfonyl)methyl)morpholine (631)



embedded image


The title compound (631) was obtained as a free amine in 91% yield (423 mg; 1.22 mmol) from 63h (600 mg; 1.34 mmol) according to the General Procedure IVa and then after basic (5% NaHCO3) extraction with DCM.


ESI-MS m/z for C13H19BrNO3S found 348.0/350.0 [M+H]+; Rt=3.29 min (Method C)


Step 10.
Synthesis of (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (63)



embedded image


The title compound (63) was obtained as a single isomer as a hydrochloride salt from 63i (180 mg; 0.52 mmol) and ketone 2d (100 mg; 0.52 mmol) according to the General Procedure VII in 15% yield (45 mg; 0.08 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 6:4 to 0:100, v/v, then: AcOEt/MeOH, 98:2 to 96:4, v/v) and then the obtained product was transformed into hydrochloride salt with using of 1 M HCl (1 mL). The obtained solid of the hydrochloride salt was then lyophilized.


ESI-MS m/z for C24H35BrN3O3S found 524.1/526.1 [M+H]+; Rt=4.19 min (Method C); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.54-7.50 (m, 2H), 7.28-7.24 (m, 2H), 5.82 (s, 1H), 4.33-4.27 (m, 1H), 3.87-3.75 (m, 2H), 3.70-3.66 (m, 1H), 3.61 (s, 3H), 3.60-3.55 (m, 2H), 3.47-3.35 (m, 3H), 3.23-3.16 (m, 2H), 3.12-3.08 (m, 1H), 3.03 (s, 3H), 2.28-2.22 (m, 2H), 2.18 (s, 3H), 2.09-2.02 (m, 2H), 1.60-1.52 (m, 2H), 1.52-1.41 (m, 2H).


Example 64
Synthesis of N-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)methanesulfonamide hydrochloride (64)



embedded image


The title compound (64) was obtained as a hydrochloride salt as a single isomer in 30% yield (44 mg; 0.09 mmol) from 46c (126 mg; 0.30 mmol) according to the General Procedure X. The crude product was purified by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 40 min, 12 mL/min). Then the obtained product was transformed into hydrochloride salt with using of 1 M HCl (2 mL). The obtained solid of the hydrochloride salt was then lyophilized.


ESI-MS m/z for C24H36ClN4O3S found 495.2/497.2 [M+H]+; Rt=4.10 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.43-7.28 (m, 4H), 6.44 (s, 1H), 4.02-3.96 (m, 1H), 3.94 (s, 3H), 3.92-3.75 (m, 3H), 3.69-3.55 (m, 2H), 3.42-3.33 (m, 2H), 3.27-3.16 (m, 2H), 3.21-3.11 (m, 1H), 2.99 (s, 3H), 2.95-2.80 (m, 1H), 2.56-2.44 (m, 2H), 2.42 (s, 3H), 2.30-2.18 (m, 2H), 1.88-1.60 (m, 4H).


Example 65
Synthesis of (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid hydrochloride (65)



embedded image


Step 1.
Synthesis of (2R,5S)-5-(4-bromobenzyl)-4-(tert-butoxycarbonyl)morpholine-2-carboxylic acid (65a)



embedded image


To a solution of an alcohol 63e (0.7 g; 1.80 mmol) in acetone (18.5 mL) at 0° C., Jones reagent (2.1 mL; 2.6 M) was added dropwise. The reaction mixture was stirred at 0° C. for 2 hours, and then isopropanol (iPrOH) (4.5 mL) was added. After 10 minutes ethyl acetate was added and the mixture was filtered through a pad of the Celite. The filtrate was washed with brine, dried over anhydrous Na2SO4 and evaporated in vacuo affording the title compound 65a in 79% yield (566 mg; 1.42 mmol).


ESI-MS m/z for C12H15BrNO3 found 300.0/302.0 [M+H−Boc]+; Rt=5.87 min (Method C)


Step 2.
Synthesis of methyl (2R,5S)-5-(4-bromobenzyl)morpholine-2-carboxylate (65b)



embedded image


To a solution of 65a (566 mg; 1.42 mmol) in MeOH (12 mL) a solution of HCl/MeOH (3.8 M; 2.2 mL) was added dropwise at 0° C. Then the cooling bath was removed and the reaction mixture was stirred at room temperature overnight. After reaction was completed as judged by LC-MS, the solvent was evaporated in vacuo. Then another portion of HCl/MeOH (3.8 M; 4 mL) was added and the reaction mixture was stirred at 45° C. for 3 hours. The residue was taken between 5% NaHCO3 and DCM and an aqueous phase was extracted with DCM (2×). The combined organic solutions were dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was used to the next step without additional purification. Compound 65b was obtained in 72% yield (321 mg; 1.02 mmol).


ESI-MS m/z for C13H17BrNO3 found 314.0/316.0 [M+H]+; Rt=3.37 min (Method C)


Step 3.
Synthesis of methyl (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylate (65c)



embedded image


The title compound (65c) was obtained from 65b (220 mg; 0.70 mmol) and ketone 2d (135 mg; 0.70 mmol) according to the General Procedure VII in 21% yield (74 mg; 0.15 mmol).


ESI-MS m/z for C24H33BrN3O3 found 490.0/492.0 [M+H]+; Rt=4.21 min (Method C)


Step 4.
Synthesis of (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid hydrochloride (65)



embedded image


To the solution of 65c (74 mg; 0.15 mmol) in MeOH (1 mL) 1 M NaOH (1 mL) was added and whole was stirred at room temperature for 2 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the solvent was evaporated in vacuo and the residue was dissolved in MeCN/water and to this solution 5% TFA was added to pH 3. The crude product was then purified by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 40 min, 12 mL/min) and then the obtained product was transformed into hydrochloride salt with using of 1 M HCl (2 mL). The title compound (65) was obtained as a single isomer as a hydrochloride salt in 73% yield (58 mg; 0.11 mmol).


ESI-MS m/z for C23H31BrN3O3 found 476.1/478.1 [M+H]+; Rt=4.12 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.60-7.47 (m, 2H), 7.33-7.22 (m, 2H), 6.33 (s, 1H), 4.71-4.49 (m, 1H), 4.07-3.79 (m, 7H), 3.71-3.41 (m, 2H), 3.27-3.03 (m, 2H), 2.90-2.76 (m, 1H), 2.59-2.34 (m, 5H), 2.28-2.16 (m, 2H), 1.92-1.57 (m, 4H).


Example 66
Synthesis of (2R,5S)-5-(4-bromobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid 2,2,2-trifluoroacetate (66)



embedded image


The title compound (66) was obtained as a TFA salt as a single isomer in 2% overall yield in a similar way to Example 65 with the exception that, in the third step of the synthesis, ketone 1g was used instead of ketone 2d. The crude product was purified by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water/MeCN+1‰ TFA, 95:5 to 20:80.40 min, 12 mL/min).


ESI-MS m/z for C22H28BrN2O4 found 463.1/465.1 [M+H]+; Rt=4.14 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.57-7.50 (m, 2H), 7.31-7.23 (m, 2H), 6.70 (s, 1H), 4.62-4.51 (m, 1H), 3.96-3.59 (m, 6H), 3.25-3.06 (m, 2H), 2.92-2.73 (m, 1H), 2.49-2.20 (m, 7H), 1.77-1.62 (m, 4H).


Example 67
Synthesis of (2R,5S)-5-(4-bromobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine 2,2,2-trifluoroacetate (67)



embedded image


The title compound (67) was obtained as a single isomer as a TFA salt from 63i (105 mg; 0.30 mmol) and ketone 1g (54 mg; 0.30 mmol) according to the General Procedure VII in 47% yield (87 mg; 0.14 mmol). The crude product was purified by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water/MeCN+1‰ TFA, 95:5 to 20:80.40 min, 12 mL/min).


ESI-MS m/z for C23H32BrN2O4S found 511.1/513.1 [M+H]+; Rt=4.19 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.58-7.49 (m, 2H), 7.31-7.22 (m, 2H), 6.71 (s, 1H), 4.42-4.28 (m, 1H), 3.96-3.52 (m, 6H), 3.47-3.36 (m, 2H), 3.29-3.23 (m, 1H), 3.20-3.12 (m, 1H), 3.10 (s, 3H), 2.91-2.79 (m, 1H), 2.54-2.40 (m, 2H), 2.38-2.27 (m, 5H), 1.80-1.52 (m, 4H).


Example 68
Synthesis of (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (68)



embedded image


The title compound (68) was obtained as a single isomer as a hydrochloride salt from 63i (118 mg; 0.34 mmol) and ketone 15c (61 mg; 0.34 mmol) according to the General Procedure VII in 15% yield (30 mg; 0.05 mmol). The crude product was purified by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 40 min, 12 mL/min) and then the obtained product was transformed into hydrochloride salt with using of 1 M HCl (2 mL).


ESI-MS m/z for C23H34BrN4O3S found 525.1/527.1 [M+H]+; Rt=3.67 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.58-7.42 (m, 2H), 7.35-7.17 (m, 2H), 4.53-4.35 (m, 1H), 4.02-3.85 (m, 5H), 3.85-3.52 (m, 4H), 3.47-3.35 (m, 2H), 3.28-3.22 (m, 1H), 3.21-3.12 (m, 1H), 3.07 (s, 3H), 3.03-2.92 (m, 1H), 2.68 (s, 3H), 2.55-2.14 (m, 4H), 2.06-1.69 (m, 4H).


Example 69
Synthesis of (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid hydrochloride (69)



embedded image


Step 1.
Synthesis of methyl (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)morpholine-2-carboxylate (69a)



embedded image


The title compound (69a) was obtained from 65b (154 mg; 0.49 mmol) and ketone 15c (95 mg; 0.49 mmol) according to the General Procedure VII in 47% yield (112 mg; 0.23 mmol).


ESI-MS m/z for C23H32BrN4O3 found 491.0/493.0 [M+H]+; Rt=3.78 min (Method C)


Step 2.
Synthesis of (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid hydrochloride (69)



embedded image


To the solution of 69a (112 mg; 0.23 mmol) in MeOH (1 mL) 1 M NaOH (1 mL) was added and whole was stirred at room temperature for 2 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the solvent was evaporated in vacuo and the residue was dissolved in MeCN/water and to this solution 5% TFA was added to pH 3. The crude product was then purified by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 40 min, 12 mL/min) and then the obtained product was transformed into hydrochloride salt with using of 1 M HCl (2 mL). The title compound (69) was obtained as a single isomer as a hydrochloride salt in 43% yield (53 mg; 0.10 mmol).


ESI-MS m/z for C22H30BrN4O3 found 477.1/479.1 [M+H]+; Rt=3.30 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.58-7.47 (m, 2H), 7.34-7.23 (m, 2H), 4.72-4.54 (m, 1H), 4.01-3.77 (m, 7H), 3.70-3.58 (m, 1H), 3.57-3.41 (m, 1H), 3.39-3.31 (m, 1H), 3.29-3.10 (m, 1H), 3.07-2.93 (m, 1H), 2.71 (s, 3H), 2.58-2.13 (m, 4H), 2.08-1.60 (m, 4H).


Example 70
Synthesis of N-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1,1,1-trifluoromethanesulfonamide hydrochloride (70)



embedded image


The title compound (70) was obtained as a hydrochloride salt as a single isomer in 24% yield (54 mg; 0.09 mmol) from 46c (153 mg; 0.37 mmol) according to the General Procedure X with the exception that, the TfCl was added to the reaction mixture at 0° C. and then the reaction was allowed to room temperature. The crude product was purified by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 40 min, 12 mL/min). Then the obtained product was transformed into hydrochloride salt with using HCl/AcOEt (3.6 M; 0.2 mL). The obtained solid of the hydrochloride salt was then lyophilized.


ESI-MS m/z for C24H33ClF3N4O3S found 549.2/551.2 [M+H]+; Rt=4.67 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.46-7.21 (m, 4H), 6.30 (s, 1H), 4.09-3.94 (m, 1H), 3.94-3.74 (m, 6H), 3.74-3.35 (m, 5H), 3.24-3.12 (m, 2H), 2.88-2.71 (m, 1H), 2.55-2.42 (m, 2H), 2.37 (s, 3H), 2.29-2.12 (m, 2H), 1.86-1.54 (m, 4H).


Example 71
Synthesis of (2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide hydrochloride (71)



embedded image


The title compound (71) was obtained as a hydrochloride salt as a single isomer in 10% overall yield in a similar way to Example 30 with the exception that, in the first step of the synthesis, ethylamine hydrochloride was used instead of propan-2-amine and the compound 65a was used instead of the compound 5a and in the second step of the synthesis, the General Procedure IVb was used instead of the General Procedure IVa. The crude product was purified by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 12 mL/min). Then the obtained product was transformed into hydrochloride salt with using HCl/AcOEt (3.6 M; 0.2 mL). The obtained solid of the hydrochloride salt was then lyophilized.


ESI-MS m/z for C25H36BrN4O2 found 503.2/505.2 [M+H]+; Rt=4.21 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.57-7.52 (m, 2H), 7.35-7.28 (m, 2H), 6.43 (s, 1H), 4.65-4.38 (m, 1H), 4.01-3.79 (m, 7H), 3.68-3.54 (m, 1H), 3.42-3.32 (m, 2H), 3.29-3.02 (m, 3H), 2.93-2.74 (m, 1H), 2.58-2.45 (m, 2H), 2.42 (s, 3H), 2.32-2.17 (m, 2H), 1.88-1.57 (m, 4H), 1.17 (t, J=7.2 Hz, 3H).


Example 72
Synthesis of (2R,5S)—N-(2-(3-(but-3-yn-1-yl)-3H-diazirin-3-yl)ethyl)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamide hydrochloride (72)



embedded image


To the solution of 11 (30 mg; 0.055 mmol) in dry DMF (0.33 mL) diisopropylethylamine (DIPEA; 38 μL; 0.220 mmol), 2-(3-(but-3-yn-1-yl)-3H-diazirin-3-yl)ethan-1-amine (10 mg; 0.072 mmol) were added and after 5 minutes HATU (23 mg; 0.060 mmol) was added and the reaction mixture was stirred at room temperature overnight. The reaction progress was monitored by TLC and LC-MS. When analyses indicated completion of the reaction, the mixture was concentrated in vacuo, diluted with water and the crude product was purified by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 12 mL/min). Then the obtained product was transformed into hydrochloride salt with using HCl/AcOEt (3.6 M; 0.2 mL). The obtained solid of the hydrochloride salt was then lyophilized. The title compound (72) was obtained as a hydrochloride salt as a single isomer in 67% yield (22 mg; 0.037 mmol).


ESI-MS m/z for C30H40ClN6O2 found 551.2/553.2 [M+H]+; Rt=4.73 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.42-7.34 (m, 4H), 6.48 (s, 1H), 4.65-4.47 (m, 1H), 4.09-3.73 (m, 8H), 3.72-3.51 (m, 1H), 3.47-3.32 (m, 1H), 3.27-3.14 (m, 3H), 2.94-2.83 (m, 1H), 2.58-2.45 (m, 2H), 2.43 (s, 3H), 2.34-2.20 (m, 3H), 2.10-2.00 (m, 2H), 1.95-1.81 (m, 2H), 1.79-1.58 (m, 6H).


Example 73
Synthesis of 2-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methoxy)acetic acid hydrochloride (73)



embedded image


Step 1.
Synthesis of tert-butyl (2R,5S)-5-(4-chlorobenzyl)-2-((2-ethoxy-2-oxoethoxy)methyl)morpholine-4-carboxylate (73a)



embedded image


NaH (60% in oil; 88 mg; 2.2 mmol) was suspended in the solvent (2× hexane and 2×THF) and after that the solvent was removed by syringe. Then a solution of 3d (0.5 g; 1.47 mmol) in THF (3 mL) was added at 0° C. After 2 minutes the bath was removed and ethyl bromoacetate (327 μL; 2.93 mmol) was slowly added and then stirred at room temperature overnight. The reaction progress was monitored by LC-MS analysis of small aliquots of the crude reaction mixture. When analysis indicated completion of the reaction, the mixture was quenched with EtOH. The volatiles were removed in vacuo and the crude product was dissolved in DCM/water. Then an aqueous phase was extracted with DCM (2×). The combined organic phases were dried over anhydrous Na2SO4, concentrated in vacuo. The crude product was used to the next step without additional purification. Compound 73a was obtained in 99% yield (619 mg; 1.45 mmol).


ESI-MS m/z for C21H30ClNO6Na found 450.0/452.0 [M+Na]+; Rt=4.00 min (Method D)


Step 2.
Synthesis of methyl 2-(((2R,5S)-5-(4-chlorobenzyl)morpholin-2-yl)methoxy)acetate (73b)



embedded image


The title compound (73b) was obtained as a free amine in 64% yield (172 mg; 0.55 mmol) from 73a (366 mg; 0.86 mmol) according to the General Procedure IVa and then after basic (5% NaHCO3) extraction with DCM.


ESI-MS m/z for C15H21ClNO4 found 314.0/316.0 [M+H]+; Rt=1.25 min (Method D)


Step 3.
Synthesis of methyl 2-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methoxy)acetate (73c)



embedded image


The title compound (73c) was obtained from 73b (173 mg; 0.55 mmol) and ketone 40c (116 mg; 0.55 mmol) according to the General Procedure VII. The crude product mixture was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 0:100, v/v) and give two separated isomers in 49% yield (for the first isomer)(138 mg; 0.27 mmol) and in 22% (for the second isomer)(60 mg; 0.12 mmol).


For the first isomer: ESI-MS m/z for C26H36ClN2O4S found 507.0/509.0 [M+H]+; Rt=2.38 min (Method D); For the second isomer: ESI-MS m/z for C26H36ClN2O4S found 507.0/509.0 [M+H]+; Rt=2.62 min (Method D)


Step 4.
Synthesis of 2-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methoxy)acetic acid hydrochloride (73)



embedded image


To the solution of 73c (the second isomer)(60 mg; 0.12 mmol) in MeOH (5 mL) 1 M NaOH (2 mL) was added and whole was stirred at room temperature for 1.5 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the solvent was evaporated in vacuo and the residue was dissolved in MeCN/water and to this solution 5% TFA was added to pH 3. The crude product was then purified by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 50 min, 12 mL/min) and then the obtained product was transformed into hydrochloride salt with using of 1 M HCl (2 mL). The title compound (73) was obtained as a single isomer as a hydrochloride salt in 50% yield (31 mg; 0.06 mmol).


ESI-MS m/z for C25H34ClN2O4S found 493.0/495.0 [M+H]+; Rt=2.08 min (Method D); 1H NMR (400 MHz, DMSO-d6) δ 7.47-7.37 (m, 2H), 7.36-7.27 (m, 2H), 4.08 (s, 2H), 4.05-3.97 (m, 1H), 3.89-3.84 (m, 1H), 3.64-3.62 (m, 2H), 3.58-3.42 (m, 3H), 3.17-3.03 (m, 2H), 2.95-2.80 (m, 1H), 2.34-2.25 (m, 5H), 2.20 (s, 3H), 2.16-1.95 (m, 2H), 1.75-1.43 (m, 4H), 1.25-1.18 (m, 2H).


Example 74
Synthesis of 2-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)propan-2-ol (74)



embedded image


Step 1.
Synthesis of tert-butyl (2R,5S)-5-(4-chlorobenzyl)-2-(2-hydroxypropan-2-yl)morpholine-4-carboxylate (74a)



embedded image


The title compound (74a) was obtained from 22a (0.50 g; 1.41 mmol) according to the General Procedure VI in 64% yield (332 mg; 0.90 mmol).


ESI-MS C19H28ClNO4Na found 393.2/395.2 [M+Na]+, 270.0/272.0 [M+H−Boc]+; Rt=4.20 min (Method C)


Step 2.
Synthesis of 2-((2R,5S)-5-(4-chlorobenzyl)morpholin-2-yl)propan-2-ol (74b)



embedded image


The title compound (74b) was obtained as a free amine in 92% yield (223 mg; 0.83 mmol) from 74a (332 mg; 0.90 mmol) according to the General Procedure IVb and then after basic (0.5 M NaOH) extraction with DCM.


ESI-MS m/z for C14H21ClNO2 found 270.0/272.0 [M+H]+; Rt=3.40 min (Method C)


Step 3.
Synthesis of 2-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)propan-2-ol (74)



embedded image


The title compound (74) was obtained as a free amine as a single isomer from 74b (223 mg; 0.83 mmol) and ketone 2d (179 mg; 0.91 mmol) according to the General Procedure VII in 12% yield (46 mg; 0.10 mmol). The crude product was purified by flash column chromatography on silica (AcOEt/MeOH, 100:4 to 70:30, v/v, 24 min).


ESI-MS m/z for C25H37ClN3O2 found 446.2/448.2 [M+H]+; Rt=7.50 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.30-7.26 (m, 2H), 7.23-7.18 (m, 2H), 5.89-5.85 (m, 1H), 3.68 (s, 3H), 3.67-3.62 (m, 1H), 3.51-3.45 (m, 1H), 3.06-2.93 (m, 3H), 2.83-2.76 (m, 1H), 2.63-2.48 (m, 3H), 2.33-2.17 (m, 5H), 2.08-1.98 (m, 2H), 1.58-1.43 (m, 2H), 1.39-1.27 (m, 3H), 1.27-1.22 (m, 6H).


Example 75
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(5-methyl-1H-pyrazol-3-yl)morpholine hydrochloride (75)



embedded image


Step 1.
Synthesis of tert-butyl (2R,5S)-2-(but-2-ynoyl)-5-(4-chlorobenzyl)morpholine-4-carboxylate (75a)



embedded image


The title compound (75a) was obtained from 5b (300 mg; 0.750 mmol) and from prop-1-yn-1-ylmagnesium bromide (0.5 M; 2.25 mL; 1.125 mmol) according to the General Procedure VI and the crude product was used to the next step without additional purification.


ESI-MS m/z for C20H25ClNO4 found 378.0/380.0 [M+H]+; Rt=6.50 min (Method C)


Step 2.
Synthesis of tert-butyl (2R,5S)-5-(4-chlorobenzyl)-2-(5-methyl-1H-pyrazol-3-yl)morpholine-4-carboxylate (75b)



embedded image


To the suspension of 75a (the crude product) in MeCN (2 mL) hydrazine monohydrate (73 μL; 1.50 mmol) was added and the reaction mixture was stirred at room temperature for 2 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the whole was concentrated in vacuo and the crude product was used to the next step without additional purification.


ESI-MS m/z for C20H27ClN3O3 found 392.0/394 [M+H]+; Rt=6.20 min (Method C)


Step 3.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-2-(5-methyl-1H-pyrazol-3-yl)morpholine (75c)



embedded image


The title compound (75c) was obtained as a free amine from 75b (the crude product) according to the General Procedure IVb and then after basic (0.5 M NaOH) extraction with DCM. The crude product was used to the next step without additional purification.


ESI-MS m/z for C14H21ClNO2 found 270.0/272.0 [M+H]+; Rt=3.60 min (Method C)


Step 4.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(5-methyl-1H-pyrazol-3-yl)morpholine hydrochloride (75)



embedded image


The title compound (75) was obtained as a hydrochloride salt as a single isomer from 75c (the crude product) and ketone 2d (144 mg; 0.75 mmol) according to the General Procedure VII in 5% yield (per four steps)(20 mg; 0.04 mmol). The crude product was purified by flash column chromatography on silica (AcOEt/MeOH, 100:0 to 70:30, v/v, 24 min) and then by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water+1‰ HCl (36%)/MeCN, 90:10 to 5:95, 35 min, Rt=26 min, 12 mL/min).


ESI-MS m/z for C26H35ClN5O found 468.2/470.2 [M+H]+; Rt=4.25 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.47-7.30 (m, 4H), 6.45 (s, 2H), 5.21-5.07 (m, 1H), 4.11-4.03 (m, 1H), 4.02-3.84 (m, 6H), 3.75-3.49 (m, 2H), 3.40-3.34 (m, 1H), 3.27-3.18 (m, 1H), 2.94-2.82 (m, 1H), 2.58-2.46 (m, 2H), 2.42 (s, 3H), 2.39 (d, J=0.7 Hz, 3H), 2.31-2.22 (m, 2H), 1.93-1.80 (m, 2H), 1.77-1.64 (m, 2H).


Example 76
Synthesis of (2R,5R)-2-(4-chlorobenzyl)-1-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-5-(methylsulfonyl)piperidine hydrochloride (76)



embedded image


Step 1.
Synthesis of (S)-2-((tert-butoxycarbonyl)amino)-3-(4-chlorophenyl)propanoic acid (76a)



embedded image


To a solution of L-p-chlorophenylalanine (18.0 g, 75 mmol) in acetone-water (150 mL:150 mL) sodium hydroxide (6 g, 150 mmol) was added at 0° C. Then di-tert-butyl dicarbonate (16.4 g, 75 mmol) was added. The reaction mixture was stirred at room temperature overnight. Acetone was evaporated in vacuo. An aqueous layer was acidified to pH 2 with 2 M HCl and extracted with ethyl acetate. An organic layer was dried over anhydrous MgSO4, filtered and concentrated under reduced pressure. The crude product was crystallized from hexane to obtain 76a as a white solid in 80% yield (18.0 g; 60 mmol).


ESI-MS m/z for C14H19ClNO4 found 299.8/301.8 [M+H]+; Rt=7.50 min (Method C); 1H NMR (500 MHz, DMSO-d6) δ 7.29 (d, J=8.3 Hz, 2H), 7.21 (d, J=8.3 Hz, 2H), 7.02 (d, J=7.3 Hz, 1H), 4.08-3.99 (m, 1H), 2.96 (dd, J=4.3, 13.7 Hz, 1H), 2.76 (dd, J=10.5, 13.6 Hz, 1H).


Step 2.
Synthesis of (S)-tert-butyl (3-(4-chlorophenyl)-1-(2,2-dimethyl-4,6-dioxo-1,3-dioxan-5-yl)-1-oxopropan-2-yl)carbamate (76b)



embedded image


To a solution of 76a (4 g, 13.34 mmol), DMAP (2.45 g; 20.02 mmol) and Meldrum's acid (2.31 g; 16.01 mmol) in anhydrous DCM (27 mL) DCC (3.3 g; 16.01 mmol) was added portionwise at 0° C. over 10 minutes and the mixture was allowed to room temperature and stirred for 22 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the mixture was filtered through the Celite pad in order to remove insoluble solid which was washed with DCM and discarded. The filtrate was washed with 1 M HCl (30 mL) and brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was used to the next step without additional purification. Compound 76b was obtained in 99% yield (5.61 g; 13.21 mmol).


ESI-MS m/z for C20H24ClNO7Na found 448.1/450.1 [M+Na]+; Rt=4.40 min (Method D)


Step 3.
Synthesis of (R)-tert-butyl (1-(4-chlorophenyl)-3-(2,2-dimethyl-4,6-dioxo-1,3-dioxan-5-yl)propan-2-yl)carbamate (76c)



embedded image


To a cooled to 0° C. solution of 76b (9.35 g, 21.96 mmol) and glacial acetic acid (12.6 mL; 219.56 mmol) in DCM (110 mL) NaBH4 (2.08 g; 54.89 mmol) was added portionwise over 20 minutes and the mixture was allowed to room temperature and stirred for 18 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, phases were separated and an organic phase was washed with brine (100 mL). An aqueous phase was washed additionally with DCM (3×40 mL) and discarded. The combined organic solutions were washed with brine (2×50 mL), then with saturated aqueous NaHCO3 diluted with water (100 mL; 1:1 v/v) until pH 6-7 and with brine (50 mL). Due to emulsion CHCl3 was added to the organic phase during a final washing. The combined organic solutions were dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was used to the next step without additional purification. Compound 76c was obtained in 99% yield (8.94 g; 21.74 mmol).


ESI-MS m/z for C20H26ClNO6Na found 434.1/436.1 [M+Na]+; Rt=3.80 min (Method D)


Step 4.
Synthesis of (R)-tert-butyl 2-(4-chlorobenzyl)-5-oxopyrrolidine-1-carboxylate (76d)



embedded image


The solution of 76c (8.94 g; 21.74 mmol) in toluene (110 mL) was heated to reflux for 15 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the mixture was concentrated in vacuo and the remaining oily residue was dissolved in AcOEt and then coevaporated with silica gel. The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 80:20 to 70:30, v/v, 24 min). Compound 76d was obtained as a dense orange oil in 80% yield (5.35 g; 17.31 mmol).


ESI-MS m/z for C16H20ClNO3Na found 332.1/334.1 [M+Na]+; Rt=3.60 min (Method D)


Step 5.
Synthesis of ((R)-1-(4-chlorobenzyl)-4-oxo-5-dimethylsulfoxonium-pentyl)-carbamic acid tert-butyl ester (76e)



embedded image


To a suspension of trimethylsulfoxonium iodide (4.56 g; 20.72 mmol) in anhydrous DMSO (18 mL) under nitrogen atmosphere, tBuOK (2.13 g; 19 mmol) was added and the mixture was stirred at room temperature for 2 hours until the mixture became clear. In an another flask 76d (5.35 g, 17.31 mmol) was dissolved in DMSO (17 mL) and to this solution the previously prepared ylide was transferred via syringe and the mixture was stirred at room temperature for 17 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, water (200 mL) was added and the product was extracted with AcOEt (200 mL, then 2×50 mL). The combined organic solutions were washed with water (50 mL) and brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was used to the next step without additional purification. Compound 76e was obtained as a white solid in 93% yield (6.46 g; 16.1 mmol).


ESI-MS m/z for C19H29ClNO4S found 402.1/404.1 [M+Na]+; Rt=2.30 min (Method D)


Step 6.
Synthesis of (R)-tert-butyl 2-(4-chlorobenzyl)-5-oxopiperidine-1-carboxylate (76f)



embedded image


To a solution of iridium catalyst (42 mg; 0.06 mmol) in a degassed DCE (20 mL) under nitrogen atmosphere, a solution of 76e (2.5 g; 6.22 mmol) in a degassed DCE (105 mL) was added dropwise via syringe pump (20 mL/hour) over 5 hours at 70° C. After this time the mixture was stirred at 70° C. for 30 minutes. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the mixture was concentrated in vacuo and the oily residue was dissolved in DCM and co-evaporated with silica gel. The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 70:30, v/v, 24 min). Compound 76f was obtained as a thick orange oil in 86% yield (1.73 g; 5.35 mmol).


ESI-MS m/z for C17H23ClNO3 found 325.0/327.0 [M+Na]+; Rt=3.70 min (Method D)


Step 7.
Synthesis of (2R,5R)-tert-butyl 2-(4-chlorobenzyl)-5-hydroxypiperidine-1-carboxylate (76g)



embedded image


To a solution of 76f (136 mg; 0.42 mmol) in EtOH (8.4 mL) NaBH4 (24 mg; 0.63 mmol) was added at −5° C. and the mixture was stirred at this temperature for 4 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the mixture was concentrated in vacuo and saturated aqueous solution of NH4Cl (20 mL) was added and the product was extracted with DCM (4×15 mL). The combined organic solutions were dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was used to the next step without additional purification. Compound 76g was obtained in 99% yield (137 mg; 0.42 mmol).


ESI-MS m/z for C12H17ClNO found 226.1/228.1 [M+H−Boc]+; Rt=6.00 min (Method C)


Step 8.
Synthesis of tert-butyl (2R,5S)-5-(benzoyloxy)-2-(4-chlorobenzyl)piperidine-1-carboxylate (76h)



embedded image


The solution of 76g (2 g; 6.20 mmol), PPh3 (2.1 g; 8.06 mmol) and benzoic acid (9.84 g; 8.06 mmol) in THF (25 mL) was cooled to 0° C. and then DIAD (1.83 mL; 9.30 mmol) was added dropwise. The reaction mixture was stirred under argon atmosphere for 30 minutes below 0° C., then at room temperature. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the solvent was removed in vacuo and the residue was dissolved in AcOEt and washed with brine (2×30 mL). An organic phase was dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 0:100, v/v, 24 min). Compound 76h was obtained in 99% yield (2.63 g; 6.14 mmol).


ESI-MS m/z for C24H29ClNO4 found 430.1/432.1 [M+H]+; Rt=6.80 min (Method C)


Step 9.
Synthesis of tert-butyl (2R,5S)-2-(4-chlorobenzyl)-5-hydroxypiperidine-1-carboxylate (761)



embedded image


To the solution of 76h (2.63 g; 6.14 mmol) in MeOH (15 mL) KOH (382 mg; 6.82 mmol) was added and whole was stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the solvent was evaporated in vacuo and the residue was dissolved in AcOEt and washed with 0.5 M NaOH. An organic phase was dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was used to the next step without additional purification. Compound 761 was obtained in 65% yield (1.3 g; 4.00 mmol).


ESI-MS m/z for C12H17ClNO found 226.1/228.1 [M+H−Boc]+; Rt=6.00 min (Method C)


Step 10.
Synthesis of tert-butyl (2R,5S)-2-(4-chlorobenzyl)-5-((methylsulfonyl)oxy)piperidine-1-carboxylate (76j)



embedded image


The title compound (76j) was obtained from 761 (1.3 g; 4.00 mmol) according to the General Procedure XI. The crude product was used to the next step without additional purification.


ESI-MS m/z for C18H27ClNO5S found 404.1/406.1 [M+H]+; Rt=6.40 min (Method C)


Step 11.
Synthesis of tert-butyl (2R,5R)-2-(4-chlorobenzyl)-5-(methylthio)piperidine-1-carboxylate (76k)



embedded image


To a solution of 76j (the crude product) in acetonitrile (15 mL) MeSNa (561 mg; 8.00 mmol) was added and the reaction mixture was stirred at 80° C. overnight. The reaction progress was monitored by LC-MS analysis. After analytical control indicated completion of the reaction, the mixture was dissolved in DCM and then washed with 2 M NaOH and brine, dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was used to the next step without additional purification.


ESI-MS m/z for C18H27ClNO2S found 356.1/358.1 [M+H]+; Rt=6.80 min (Method C)


Step 12.
Synthesis of tert-butyl (2R,5R)-2-(4-chlorobenzyl)-5-(methylsulfonyl)piperidine-1-carboxylate (761)



embedded image


The reaction of 76k (the crude product) with peracetic acid (35% in AcOH; 2.3 mL; 12.0 mmol) was stirred at room temperature overnight. Then the reaction was concentrated in vacuo and the residue was diluted with AcOEt, washed with 1 M NaOH and Na2S2O3. Phases were separated and the organic one was washed with brine, dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 0:100, v/v, 24 min). Compound 761 was obtained in 60% yield (per three steps)(0.93 g; 2.4 mmol).


ESI-MS m/z for C18H27ClNO4S found 388.1/390.1 [M+H]+; Rt=6.30 min (Method C)


Step 13.
Synthesis of (2R,5R)-2-(4-chlorobenzyl)-5-(methylsulfonyl)piperidine (76m)



embedded image


The title compound (76m) was obtained as a free amine from 761 (0.45 g; 1.12 mmol) according to the General Procedure IVb and then after basic (5% NaHCO3) extraction with DCM. The crude product was used to the next step without additional purification.


ESI-MS m/z for C13H19ClNO2S found 288.0/290.0 [M+H]+; Rt=3.30 min (Method C)


Step 14.
Synthesis of (2R,5R)-2-(4-chlorobenzyl)-1-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-5-(methylsulfonyl)piperidine hydrochloride (76)



embedded image


The title compound (76) was obtained as a hydrochloride salt as a single isomer from 76m (the crude product) and ketone 2d (236 mg; 1.23 mmol) according to the General Procedure VII in 7% yield (per two steps)(40 mg; 0.08 mmol). The crude product was purified by flash column chromatography on silica (AcOEt/MeOH, 100:0 to 70:30, v/v, 24 min) and then by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water+1‰ HCl (36%)/MeCN, 90:10 to 5:95, 35 min, Rt=26 min, 12 mL/min).


ESI-MS m/z for C24H35ClN3O2S found 464.1/466.1 [M+H]+; Rt=4.20 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.43-7.30 (m, 4H), 6.43 (s, 1H), 4.21-3.60 (m, 8H), 3.17-3.12 (m, 3H), 3.12-3.04 (m, 1H), 2.93-2.79 (m, 1H), 2.61-2.45 (m, 1H), 2.42 (s, 3H), 2.37-2.19 (m, 4H), 2.19-2.00 (m, 2H), 2.00-1.79 (m, 4H), 1.74-1.60 (m, 2H).


Example 77
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamide (77)



embedded image


Step 1.
Synthesis of tert-butyl (2R,5S)-2-carbamoyl-5-(4-chlorobenzyl)morpholine-4-carboxylate (77a)



embedded image


The solution of 5a (0.80 g; 2.25 mmol), Boc2O (0.74 g; 3.37 mmol) and pyridine (1 mL) in acetonitrile (16 mL) was stirred at room temperature for 30 minutes. Then to this mixture NH3× H2O (4.8 mL) was added dropwise over 30 minutes and the reaction mixture was stirred at room temperature. The reaction progress was monitored by LC-MS analysis. After analytical control indicated completion of the reaction, the mixture was concentrated in vacuo and the crude product was used to the next step without additional purification.


ESI-MS m/z for C17H24ClN2O4 found 355.1/357.1 [M+H]+; Rt=5.70 (Method C)


Step 2.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)morpholine-2-carboxamide (77b)



embedded image


The title compound (77b) was obtained in 80% yield (per two steps)(457 mg; 1.8 mmol) as a free amine from 77a (the crude product) according to the General Procedure IVb and then after basic (5% NaHCO3) extraction with DCM.


ESI-MS m/z for C12H16ClN2O2 found 255.0/257.0 [M+H]+; Rt=3.00 min (Method C)


Step 3.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-morpholine-2-carboxamide (77)



embedded image


The title compound (77) was obtained as a free amine as a single isomer from 77b (457 mg; 1.80 mmol) and ketone 2d (380 mg; 1.98 mmol) according to the General Procedure VII in 14% yield (110 mg; 0.25 mmol). The crude product was purified by flash column chromatography on silica (AcOEt/MeOH, 100:0 to 70:30, v/v, 24 min).


ESI-MS m/z for C23H32ClN4O2 found 431.1/433.2 [M+H]+; Rt=3.90 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.32-7.26 (m, 2H), 7.26-7.21 (m, 2H), 5.87 (s, 1H), 4.02-3.94 (m, 1H), 3.74-3.64 (m, 4H), 3.59-3.48 (m, 1H), 3.24-3.17 (m, 1H), 3.12-3.03 (m, 1H), 3.02-2.95 (m, 1H), 2.86-2.77 (m, 1H), 2.69-2.58 (m, 2H), 2.56-2.44 (m, 1H), 2.25-2.12 (m, 5H), 2.09-1.97 (m, 2H), 1.58-1.43 (m, 2H), 1.39-1.28 (m, 2H).


Example 78
Synthesis of 2-(4-((2R,5R)-2-(4-chlorobenzyl)-5-(methylsulfonyl)piperidin-1-yl)cyclohexyl)-5-methyloxazole hydrochloride (78)



embedded image


The title compound (78) was obtained as a hydrochloride salt as a single isomer from 76m (333 mg; 1.16 mmol) and ketone 1g (229 mg; 1.28 mmol) according to the General Procedure VII in 3% yield (15 mg; 0.03 mmol). The crude product was purified by flash column chromatography on silica (AcOEt/MeOH, 100:0 to 70:30, v/v, 24 min) and then by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water+1‰ HCl (36%)/MeCN, 90:10 to 5:95, 35 min, Rt=26 min, 12 mL/min).


ESI-MS m/z for C23H32ClN2O3S found 451.1/453.1 [M+H]+; Rt=4.20 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.45-7.29 (m, 4H), 7.20-7.05 (m, 1H), 4.21-3.61 (m, 4H), 3.47-3.34 (m, 1H), 3.28-3.04 (m, 6H), 2.60-2.50 (m, 1H), 2.46-2.24 (m, 6H), 2.17-1.70 (m, 8H).


Example 79
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfinyl)methyl)morpholine (79)



embedded image


Step 1.
Synthesis of tert-butyl (2R,5S)-5-(4-chlorobenzyl)-2-((methylsulfinyl)methyl)morpholine-4-carboxylate (79a)



embedded image


To a cooled to 0° C. solution of 3f (1.75 g; 4.70 mmol) in AcOH (2 mL) H2O2 (140 mL; 14.10 mmol) was added dropwise and stirred at room temperature overnight. Then to this reaction 0.5 M NaOH/Na2S2O3 were added and extracted with AcOEt. The organic one was dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was used to the next step without additional purification. Compound 79a was obtained in 99% yield (1.80 g; 4.65 mmol).


ESI-MS m/z for C18H27ClNO4S found 388.1/390.1 [M+H]+; Rt=5.60 min (Method C)


Step 2.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-2-((methylsulfinyl)methyl)morpholine (79b)



embedded image


The title compound (79b) was obtained in 80% yield (1.44 g; 3.72 mmol) as a free amine from 79a (1.80 g; 4.65 mmol) according to the General Procedure IVb and then after basic (5% NaHCO3) extraction with DCM.


ESI-MS m/z for C13H19ClNO2S found 288.0/290.0 [M+H]+; Rt=3.10 min (Method C)


Step 3.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfinyl)methyl)morpholine (79)



embedded image


The title compound (79) was obtained as a free amine as a single isomer from 79b (1.47 g; 3.80 mmol) and ketone 2d (803 mg; 4.18 mmol) according to the General Procedure VII in 1% yield (22 mg; 0.05 mmol). The crude product was purified by flash column chromatography on silica (AcOEt/MeOH, 100:0 to 70:30, v/v, 24 min) and then by chiral preparative reversed-phase column chromatography (Lux Amylose-1 5 μm, 250×21.2 mm, hexane/iPrOH, 70:30, v/v; 10 mL/min, Rt=22.5 min).


ESI-MS m/z for C24H35ClN3O2S found 464.1/466.1 [M+H]+; Rt=1.80 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.31-7.26 (m, 2H), 7.22-7.16 (m, 2H), 5.89-5.83 (m, 1H), 4.09-3.97 (m, 1H), 3.68 (s, 3H), 3.64-3.58 (m, 1H), 3.55-3.46 (m, 1H), 3.10-2.91 (m, 5H), 2.86-2.78 (m, 1H), 2.77-2.67 (m, 4H), 2.67-2.57 (m, 1H), 2.58-2.46 (m, 1H), 2.25-2.12 (m, 5H), 2.08-1.97 (m, 2H), 1.57-1.42 (m, 2H), 1.38-1.28 (m, 2H).


Example 80
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfinyl)methyl)morpholine (80)



embedded image


The title compound (80) was obtained during the synthesis of compound 79 as a free amine as a second single isomer from 79b (1.47 g; 3.80 mmol) and ketone 2d (803 mg; 4.18 mmol) according to the General Procedure VII in 3% yield (62 mg; 0.13 mmol). The crude product was purified by flash column chromatography on silica (AcOEt/MeOH, 100:0 to 70:30, v/v, 24 min) and then by chiral preparative reversed-phase column chromatography (Lux Amylose-1 5 μm, 250×21.2 mm, hexane/iPrOH, 70:30, v/v; 10 mL/min, Rt=25.0 min).


ESI-MS m/z for C24H35ClN3O2S found 464.0/466.0 [M+H]+; Rt=1.80 min (Method C); 1H NMR (400 MHz, Methanol-d4) δ 7.30-7.25 (m, 2H), 7.22-7.16 (m, 2H), 5.89-5.85 (m, 1H), 4.02-3.93 (m, 1H), 3.68 (s, 3H), 3.65-3.59 (m, 1H), 3.56-3.47 (m, 1H), 3.10-3.03 (m, 2H), 3.03-2.99 (m, 1H), 2.97-2.89 (m, 2H), 2.85-2.80 (m, 1H), 2.69 (s, 3H), 2.67-2.57 (m, 2H), 2.55-2.46 (m, 1H), 2.27-2.10 (m, 5H), 2.10-1.97 (m, 2H), 1.56-1.42 (m, 2H), 1.39-1.25 (m, 2H).


Example 81
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylthio)methyl)morpholine hydrochloride (81)



embedded image


Step 1.
Synthesis of 2R,5S)-5-(4-chlorobenzyl)-2-((methylthio)methyl)morpholine (81a)



embedded image


The title compound (81a) was obtained as a free amine from 3f (148 mg; 0.40 mmol) according to the General Procedure IVb and then after basic (5% NaHCO3) extraction with DCM. The crude product was used to the next step without additional purification.


ESI-MS m/z for C13H19ClNOS found 272.0/274.0 [M+H]+; Rt=1.40 min (Method D)


Step 2.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylthio)methyl)morpholine hydrochloride (81)



embedded image


The title compound (81) was obtained as a hydrochloride salt as a single isomer from 81a (the crude product) and ketone 2d (86 mg; 0.45 mmol) according to the General Procedure VII in 5% yield (8 mg; 0.02 mmol). The crude product was purified by flash column chromatography on silica (AcOEt/MeOH, 100:0 to 70:30, v/v, 24 min) and then by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water+1‰ HCl (36%)/MeCN, 90:10 to 5:95, 35 min, Rt=27 min, 12 mL/min).


ESI-MS m/z for C24H35ClN3OS found 448.1/450.1 [M+H]+; Rt=2.19 min (Method D); 1H NMR (400 MHz, Methanol-d4) δ 7.43-7.37 (m, 2H), 7.36-7.29 (m, 2H), 6.39 (s, 1H), 4.13-4.02 (m, 1H), 3.92 (s, 3H), 3.89-3.66 (m, 4H), 3.62-3.50 (m, 1H), 3.38-3.32 (m, 1H), 3.29-3.23 (m, 1H), 3.24-3.11 (m, 1H), 2.89-2.72 (m, 3H), 2.60-2.44 (m, 2H), 2.41 (s, 3H), 2.31-2.18 (m, 5H), 1.82-1.59 (m, 4H).


Example 82
Synthesis of 1-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)pyrrolidine-2,5-dione 2,2,2-trifluoroacetate (82)



embedded image


Step 1.
Synthesis of tert-butyl (2S,5S)-5-(4-chlorobenzyl)-2-((2,5-dioxopyrrolidin-1-yl)methyl)morpholine-4-carboxylate (82a)



embedded image


To the solution of an alcohol 3d (1 g; 3.00 mmol) and succinimide (330 mg; 3.30 mmol) in THF (10 mL) PPh3 (860 mg; 3.30 mmol) was added. Then a whole was cooled to −10° C. and DIAD (650 μL; 3.30 mmol) was added dropwise and stirred at room temperature for 2 days. The reaction progress was monitored by LC-MS. Then a solid was filtered off and the filtrate was concentrated in vacuo. The residue was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 0:100, v/v, 24 min). The title compound (82a) was obtained in 19% yield (240 mg; 0.57 mmol).


ESI-MS m/z for C21H28ClN2O5 found 423.0/425.0 [M+H]+; Rt=3.50 min (Method D)


Step 2.
Synthesis of 1-(((2R,5S)-5-(4-chlorobenzyl)morpholin-2-yl)methyl)pyrrolidine-2,5-dione 2,2,2-trifluoroacetate (82b)



embedded image


The title compound (82b) was obtained as a TFA salt from 82a (240 mg; 0.57 mmol) according to the General Procedure IVb. The crude product was used to the next step without additional purification.


ESI-MS m/z for C16H20ClN2O3 found 323.0/325.0 [M+H]+; Rt=1.10 min (Method D)


Step 3.
Synthesis of 1-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)pyrrolidine-2,5-dione 2,2,2-trifluoroacetate (82)



embedded image


The title compound (82) was obtained as a TFA salt as a single isomer from 82b (the crude product) and ketone 2d (160 mg; 0.82 mmol) according to the General Procedure VII in 16% yield (per two steps)(58 mg; 0.09 mmol). The crude product was purified by flash column chromatography on silica (AcOEt/MeOH, 100:0 to 70:30, v/v, 24 min) and then by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water+1‰ HCl (36%)/MeCN, 90:10 to 5:95, 35 min, Rt=27.5 min, 12 mL/min).


ESI-MS m/z for C27H36ClN4O3 found 499.2/501.2 [M+H]+; Rt=2.00 min (Method D); 1H NMR (400 MHz, Methanol-d4) δ 7.40-7.35 (m, 2H), 7.34-7.29 (m, 2H), 5.99-5.94 (m, 1H), 4.06-3.96 (m, 1H), 3.90-3.48 (m, 11H), 3.28-3.20 (m, 1H), 3.17-3.11 (m, 1H), 2.80-2.73 (m, 4H), 2.65-2.55 (m, 1H), 2.49-2.40 (m, 2H), 2.26 (s, 3H), 2.22-2.13 (m, 2H), 1.69-1.53 (m, 4H).


Example 83
Synthesis of (2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-2-ethynylmorpholine hydrochloride (83)



embedded image


The title compound (83) was obtained as a single isomer as a free amine from 28a (495 mg; 2.10 mmol) and ketone 40c (462 mg; 2.20 mmol) according to the General Procedure VII in 28% yield (250 mg; 0.58 mmol). The crude product was purified by flash column chromatography on silica (hexane/MeCN, 100:0 to 40:60, v/v, 40 min) and then a small fraction of the product was purified by preparative reversed-phase column chromatography (C-18, water+0.3‰ HCl (36%)/MeCN, 95:5 to 10:90, 30 min, 20 mL/min) to obtain the product 83 a s a hydrochloride salt.


ESI-MS m/z for C24H30ClN2OS found 429.2/431.2 [M+H]+; Rt=1.38 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.5 Hz, 2H), 7.31 (d, JBB′=8.5 Hz, 2H), 4.72-4.70 (m, 1H), 3.74-3.68 (m, 3H), 3.54 (d, J=2.2 Hz, 1H), 3.52-3.47 (m, 2H), 3.35 (dd, J=13.1 Hz, J=8.9 Hz, 1H), 3.18-3.15 (m, 1H), 3.05-3.02 (m, 1H), 2.85 (tt, J=12.3 Hz, J=3.7 Hz, 1H), 2.26 (brs, 3H), 2.23-2.11 (m, 7H), 1.65-1.44 (m, 4H).


Example 84
Synthesis of 2-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)acetic acid hydrochloride (84)



embedded image


Step 1.
Synthesis of ethyl 2-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)acetate (84a)



embedded image


To a solution of 83 (75 mg; 0.175 mmol) in MeOH (4 mL) CuSO4×5 H2O (22 mg; 0.087 mmol) was added and after 5 minutes sodium ascorbate (35 mg; 0.175 mmol) was added in one portion and the mixture was stirred at room temperature for 25 minutes. Then to this mixture a solution of ethyl 2-azidoacetate (45 mg; 0.350 mmol) in MeOH (5 mL) was added and the mixture was stirred at room temperature for 2 days. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the all crude reaction mixture was used to the next step without additional purification.


ESI-MS m/z for C28H37ClN5O3S found 558.1/560.1 [M+H]+; Rt=1.40 min (Method A)


Step 2.
Synthesis of 2-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)acetic acid hydrochloride (84)



embedded image


To the 84a (the crude reaction mixture) 1 M NaOH (3 mL) was added and whole was stirred at room temperature for 3 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the solvent was evaporated in vacuo and the residue was dissolved in MeCN/water with HClaq. An insoluble acid was filtered off and the filtrate was taken for the preparative reversed-phase column chromatography (C-18, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min). The title compound (84) was obtained as a single isomer as a hydrochloride salt in 33% yield (per two steps)(33 mg; 0.058 mmol).


ESI-MS m/z for C26H33ClN5O3S found 530.2/532.2 [M+H]+; Rt=1.16 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 8.21 (s, 1H), 7.38 (d, JAA′=8.5 Hz, 2H), 7.32 (d, JBB′=8.5 Hz, 2H), 5.26 (s, 2H), 5.07-5.05 (m, 1H), 3.92-3.90 (m, 1H), 3.81-3.79 (m, 1H), 3.71 (dd, J=13.1 Hz, J=1.3 Hz, 1H), 3.67-3.65 (m, 1H), 3.60-3.57 (m, 1H), 3.54-3.50 (m, 1H), 3.23 (dd, J=13.8 Hz, J=10.6 Hz, 1H), 3.17 (dd, J=13.8 Hz, J=4.0 Hz, 1H), 2.87 (tt, J=11.8 Hz, J=3.5 Hz, 1H), 2.33-2.28 (m, 2H), 2.26 (s, 3H), 2.18-2.16 (m, 5H), 1.65-1.51 (m, 4H).


Example 85
Synthesis of 3-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)propanoic acid hydrochloride (85)



embedded image


To a solution of 83 (50 mg; 0.116 mmol) in MeOH (4 mL) CuSO4×5 H2O (15 mg; 0.058 mmol) was added and after 5 minutes sodium ascorbate (23 mg; 0.116 mmol) was added in one portion and the mixture was stirred at room temperature for 25 minutes. Then to this mixture a solution of 3-azidopropanoic acid (27 mg; 0.233 mmol) in MeOH (5 mL) was added and the mixture was stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the solvent was evaporated in vacuo and the residue was dissolved in MeCN/water with HClaq. An insoluble acid was filtered off and the filtrate was taken for the preparative reversed-phase column chromatography (C-18, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min). The title compound (85) was obtained as a single isomer as a hydrochloride salt in 49% yield (33 mg; 0.057 mmol).


ESI-MS m/z for C26H35ClN5O3S found 544.2/546.2 [M+H]+; Rt=1.06 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 8.18 (s, 1H), 7.38 (d, JAA′=8.5 Hz, 2H), 7.33 (d, JBB′=8.5 Hz, 2H), 5.07-5.05 (m, 1H), 4.59 (t, J=6.6 Hz, 2H), 3.93-3.92 (m, 1H), 3.85-3.83 (m, 1H), 3.71-3.66 (m, 2H), 3.61-3.56 (m, 2H), 3.24 (dd, J=13.9 Hz, J=10.6 Hz, 1H), 3.20-3.17 (m, 1H), 2.92-2.85 (m, 3H), 2.34-2.30 (m, 2H), 2.26 (s, 3H), 2.19-2.16 (m, 5H), 1.67-1.51 (m, 4H).


Example 86
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohex-3-en-1-yl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (86)



embedded image


Step 1.
Synthesis of 1,5-dimethyl-3-(1,4-dioxaspiro[4.5]dec-7-en-8-yl)-1H-pyrazole (86a)



embedded image


The solution of 4,4,5,5-tetramethyl-2-(1,4-dioxaspiro[4.5]dec-7-en-8-yl)-1,3,2-dioxaborolane (980 mg; 3.68 mmol), 3-bromo-1,5-dimethyl-1H-pyrazole (613 mg, 3.50 mmol), Pd(dppf) (143 mg; 0.175 mmol) in mixture of dioxane and 2 M aqueous solution of K2CO3 (20 mL; 4:1 v/v) was heated at 100° C. for 3 hours under argon atmosphere. LC-MS was showed complete consumption of the starting material. Then a whole was concentrated in vacuo and the residue was purified by flash column chromatography on silica (hexane/AcOEt, 90:10 to 0:100, v/v, 30 min, 30 mL/min). Compound 86a was obtained in 79% yield (650 mg; 2.78 mmol).


ESI-MS m/z for C13H19N2O2 found 235.1 [M+H]+; Rt=0.99 min (Method A)


Step 2.
Synthesis of 4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohex-3-en-1-one (86b)



embedded image


The solution of 86a (200 mg; 0.855 mmol) in 1 M KHSO4aq (15 mL) was stirred at 30° C. for 3 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, to this mixture water was added and a whole was taken for lyophilization. Then all crude product was used to the next step without additional purification.


ESI-MS m/z for C13H19N2O2 found 235.1 [M+H]+; Rt=0.98 min (Method A)


Step 3.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohex-3-en-1-yl)-2-((methylsulfonyl)meth yl)morpholine hydrochloride (86)



embedded image


The title compound (86) was obtained as a hydrochloride salt from 3h (288 mg; 0.85 mmol) and ketone 86b (the crude product) according to the General Procedure VII in % yield (4 mg; 0.09 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 30:70, v/v) and then purified three times by preparative reversed-phase column chromatography (twice: C-18, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min; third: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C24H33ClN3O3S found 478.0/480.0 [M+H]+; Rt=1.00 min (Method A); 1H NMR (700 MHz, D2O, 333 K) δ 7.83-7.81 (m, 2H), 7.73-7.70 (m, 2H), 6.67-6.66 (m, 1H), 6.60-6.59 (m, 1H), 4.41-4.39 (m, 1H), 4.35-4.10 (m, 8H), 4.07-4.05 (m, 1H), 3.94-3.91 (m, 1H), 3.88-3.84 (m, 1H), 3.71-3.66 (m, 1H), 3.62-3.59 (m, 4H), 3.26-3.20 (m, 1H), 3.11-3.05 (m, 1H), 2.91-2.79 (m, 3H), 2.65 (s, 3H), 2.26-2.20 (m, 1H).


Example 87
Synthesis of N-(35-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxapentatriacontyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide hydrochloride (87)



embedded image


To the solution of 83 (20 mg; 0.047 mmol) in MeOH (02 mL) CuSO4×5 H2O (6 mg; 0.023 mmol) was added and after 5 minutes to a light green solution sodium L-ascorbate (10 mg; 0.047 mmol) was added in one portion. A whole was stirred for 25 minutes at room temperature and then a solution of an azide (37 mg; 0.047 mmol) in MeOH (0.5 mL) was added and the reaction mixture was stirred at room temperature overnight. After analytical control (LC-MS) indicated completion of the reaction, the reaction mixture was concentrated in vacuo and the residue was mixed with water/MeCN and then this mixture was filtered and purified twice by preparative reversed-phase column chromatography (first: C-18, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min, second: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min). Compound 87 was obtained as a single isomer as a hydrochloride salt in 36% yield (21 mg; 0.017 mmol).


ESI-MS m/z for C58H94ClN8O4S2 found 1225.4/1227.4 [M+H]+; Rt=1.24 min (Method A)


Example 88
Synthesis of 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)acetic acid hydrochloride (88)



embedded image


Step 1.
Synthesis of methyl 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)acetate (88a)



embedded image


The title compound (88a) was obtained from 59c (150 mg; 0.48 mmol) and ketone 40c (100 mg; 0.48 mmol) according to the General Procedure VII in 27% yield (62 mg; 0.13 mmol).


ESI-MS m/z for C25H34ClN2O3S found 477.1/479.1 [M+H]+; Rt=2.27 min (Method D)


Step 2.
Synthesis of 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)acetic acid hydrochloride (88)



embedded image


To the solution of 88a (62 mg; 0.13 mmol) in MeOH (3 mL) 1 M NaOH (2 mL) was added and a whole was stirred at room temperature for 4 hours. The reaction progress was monitored by LC-MS. Then to this mixture 1M HCl was added to pH=2. The crude product was purified by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water/MeCN, 95:5 to 5:95, 50 min, 12 mL/min). The title compound (88) was obtained as a single isomer as a hydrochloride salt in 61% yield (42 mg; 0.08 mmol).


ESI-MS m/z for C24H32ClN2O3S found 463.1/465.1 [M+H]+; Rt=2.16 min (Method D); 1H NMR (400 MHz, DMSO-d6) δ 7.46-7.11 (m, 4H), 4.30-4.16 (m, 1H), 3.96-3.84 (m, 1H), 3.49-3.33 (m, 2H), 3.17-2.98 (m, 2H), 2.92-2.78 (m, 1H), 2.73-2.57 (m, 2H), 2.29-2.06 (m, 8H), 1.95-1.66 (m, 5H), 1.62-1.33 (m, 2H), 1.32-1.15 (m, 2H).


Example 89
Synthesis of 3-((((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methyl)sulfonyl)propanoic acid hydrochloride (89)



embedded image


Step 1.
Synthesis of 3-((((2R,5S)-4-(tert-butoxycarbonyl)-5-(4-chlorobenzyl)morpholin-2-yl)methyl)thio)propanoic acid (89a)



embedded image


To a DMSO (5 mL) DBU (1.91 g; 11.93 mmol) was added and degassed. Then to this solution 3-mercaptopropanoic acid (666 mg; 5.97 mmol) was added. Then the compound 3e (1.0 g; 2.39 mmol) was dissolved in DMSO (9 mL) and degassed and added to the mixture with DBU and all mixture was stirred at room temperature. The reaction progress was monitored by LC-MS analysis. After analytical control indicated completion of the reaction, the mixture was extracted with DCM and then an organic solution was washed with HCl and dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was used to the next step without additional purification.


ESI-MS m/z for C15H21ClNO3S found 330.0/332.0 [M+H−Boc]+; Rt=3.59 min (Method D)


Step 2.
Synthesis of 3-((((2R,5S)-4-(tert-butoxycarbonyl)-5-(4-chlorobenzyl)morpholin-2-yl)methyl)sulfonyl)propanoic acid (89b)



embedded image


To a solution of 89a (the crude product) in DCM (15 mL) mCPBA (1.65 g; 9.54 mmol) was added portionwise at 0° C. Then the cooling bath was removed and the mixture was stirred at room temperature overnight. The reaction progress was monitored by LC-MS analysis. When analysis indicated completion of the reaction, the reaction was quenched with 10% Na2SO3 and stirred for 30 minutes at room temperature. Then phases were separated and the organic one was washed with 1 M NaOH (4×) and brine, dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was used to the next step without additional purification. Compound 89b was obtained in 54% yield (per two steps)(0.60 g; 1.30 mmol).


ESI-MS m/z for C20H28ClNO7SNa found 484.1/486.1 [M+Na]+; Rt=3.32 min (Method D)


Step 3.
Synthesis of methyl 3-((((2R,5S)-5-(4-chlorobenzyl)morpholin-2-yl)methyl)sulfonyl)propanoate (89c)



embedded image


The title compound (89c) was obtained as a free amine in 99% yield (484 mg; 1.29 mmol) from 89b (0.60 g; 1.30 mmol) according to the General Procedure IVa and then after basic (saturated solution of NaHCO3) extraction with DCM.


ESI-MS m/z for C16H23ClNO5S found 376.0/378.0 [M+H]+; Rt=2.21 min (Method D)


Step 4.
Synthesis of 3-((((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methyl)sulfonyl)propanoic acid hydrochloride (89)



embedded image


To the solution of 89c (256 mg; 0.68 mmol) in MeOH (2 mL) a ketone 40c (150 mg; 0.72 mmol) and ZnCl2 (107 mg; 0.78 mmol) were added. After everything was dissolved NaBH3CN (50 mg; 0.78 mmol) was added and the mixture was stirred at room temperature overnight. The reaction progress was monitored by LC-MS analysis. After analytical control indicated completion of the reaction, the mixture was extracted with DCM and then an organic solution was washed with water and dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was purified by flash column chromatography on silica (hexane/AcOEt, 6:4 to 0:100, v/v) and then the obtained product (25% yield; 96 mg; 0.17 mmol) was dissolved in 20% HCl (5 mL) and all was stirred at room temperature for 2.5 hours. After analytical control indicated completion of the reaction, the solvents were removed in vacuo and the residue was purified by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water/MeCN, 95:5 to 5:95, 50 min, 12 mL/min). The title compound (89) was obtained as a single isomer as a hydrochloride salt in 45% yield (45 mg; 0.076 mmol).


ESI-MS m/z for C26H36ClN2O5S2 found 555.1/557.1 [M+H]+; Rt=2.20 min (Method D); 1H NMR (400 MHz, DMSO-d6) δ 7.43-7.37 (m, 2H), 7.36-7.31 (m, 2H), 4.38-4.28 (m, 1H), 3.96-3.88 (m, 1H), 3.83-3.70 (m, 2H), 3.47-3.40 (m, 3H), 3.37-3.28 (m, 1H), 3.21-3.07 (m, 2H), 2.94-2.63 (m, 3H), 2.37-2.30 (m, 2H), 2.28 (s, 3H), 2.24-2.11 (m, 6H), 2.04-1.93 (m, 1H), 1.80-1.41 (m, 5H).


Example 90
Synthesis of 3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2,4-dione hydrochloride (90)



embedded image


The title compound (90) was obtained as a hydrochloride salt as a single isomer in 4% overall yield in a similar way to Example 82 with the exception that, in the first step of the synthesis, 6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2,4-dione was used instead of a succinimide and in the second step of the synthesis, the obtained TFA salt was transformed into a free amine with the basic (0.5 M NaOH) extraction with DCM. The crude product was purified by flash column chromatography on silica (AcOEt/MeOH, 100:0 to 70:30, v/v, 24 min) and then by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water+1‰ HCl (36%)/MeCN, 90:10 to 5:95, 35 min, Rt=25 min, 12 mL/min).


ESI-MS m/z for C30H40ClN4O3 found 539.2/541.2 [M+H]+; Rt=2.39 min (Method D); 1H NMR (400 MHz, D2O) δ 7.27-7.14 (m, 2H), 7.13-7.01 (m, 2H), 6.09 (s, 1H), 3.85-3.74 (m, 1H), 3.73-3.65 (m, 1H), 3.63 (s, 3H), 3.59-3.44 (m, 3H), 3.43-3.27 (m, 3H), 3.03-2.90 (m, 3H), 2.67-2.53 (m, 1H), 2.50-2.34 (m, 2H), 2.34-2.17 (m, 2H), 2.12 (s, 3H), 2.06-1.92 (m, 2H), 1.45-1.26 (m, 4H), 1.04 (s, 3H), 1.00 (s, 3H).


Example 91
Synthesis of 3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2,4-dione hydrochloride (91)



embedded image


The title compound (91) was obtained as a hydrochloride salt as a single isomer in 5% overall yield in a similar way to Example 82 with the exception that, in the first step of the synthesis, 6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2,4-dione was used instead of a succinimide and in the second step of the synthesis, the obtained TFA salt was transformed into a free amine with the basic (0.5 M NaOH) extraction with DCM and in the third step of the synthesis, the ketone 40c was used instead of the ketone 2d. The crude product was purified by flash column chromatography on silica (AcOEt/MeOH, 100:0 to 70:30, v/v, 24 min) and then by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water+1‰ HCl (36%)/MeCN, 90:10 to 5:95, 35 min, Rt=24 min, 12 mL/min).


ESI-MS m/z for C30H39ClN3O3S found 556.1/558.1 [M+H]+; Rt=2.54 min (Method D); 1H NMR (400 MHz, D2O) δ 7.26-7.19 (m, 2H), 7.13-7.05 (m, 2H), 3.90-3.77 (m, 1H), 3.77-3.67 (m, 1H), 3.66-3.32 (m, 6H), 3.19-3.10 (m, 1H), 3.07-2.93 (m, 3H), 2.51-2.40 (m, 2H), 2.34-2.25 (m, 2H), 2.23-2.10 (m, 8H), 1.66-1.36 (m, 4H), 1.07 (s, 3H), 1.02 (s, 3H).


Example 92
Synthesis of 3-((((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)amino)-4-hydroxycyclobut-3-ene-1,2-dione hydrochloride (92)



embedded image


To the solution of 46c (120 mg; 0.288 mmol) in MeOH (2 mL) 3,4-dimethoxycyclobut-3-ene-1,2-dione (41 mg; 0.288 mmol) was added and the reaction mixture was stirred at room temperature for 30 minutes. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the mixture was concentrated in vacuo. The crude product was treated with water and evaporated in vacuo. Then to this residue a solution of NH4Cl was added and stirred at room temperature for 30 minutes. Then the mixture was concentrated in vacuo and the crude product was purified by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water+1‰ HCl (36%)/MeCN, 90:10 to 5:95, 35 min, Rt=22.5 min, 12 mL/min). The title compound (92) was obtained as a hydrochloride salt as a single isomer in 49% yield (78 mg; 0.142 mmol).


ESI-MS m/z for C2H34ClN4O4 found 513.1/515.1 [M+H]+; Rt=2.14 min (Method D); 1H NMR (400 MHz, Methanol-d4) δ 7.40-7.35 (m, 2H), 7.35-7.30 (m, 2H), 6.39 (s, 1H), 4.15-3.99 (m, 1H), 3.92 (s, 3H), 3.89-3.73 (m, 5H), 3.73-3.65 (m, 1H), 3.65-3.54 (m, 1H), 3.27-3.13 (m, 3H), 2.89-2.77 (m, 1H), 2.55-2.43 (m, 2H), 2.40 (s, 3H), 2.31-2.20 (m, 2H), 1.86-1.60 (m, 4H).


Example 93
Synthesis of 3-amino-4-((((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)amino)cyclobut-3-ene-1,2-dione hydrochloride (93)



embedded image


To the solution of 46c (120 mg; 0.288 mmol) in MeOH (2 mL) 3,4-dimethoxycyclobut-3-ene-1,2-dione (41 mg; 0.288 mmol) was added and the reaction mixture was stirred at room temperature overnight. Then to this mixture NH3 (7 N solution in MeOH; 0.1 mL) was added and stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the mixture was concentrated in vacuo. The crude product was purified by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water+1‰ HCl (36%)/MeCN, 90:10 to 5:95, 35 min, Rt=21.5 min, 12 mL/min). The title compound (93) was obtained as a hydrochloride salt as a single isomer in 18% yield (29 mg; 0.053 mmol).


ESI-MS m/z for C27H35ClN5O3 found 512.1/514.1 [M+H]+; Rt=1.79 min (Method D); 1H NMR (400 MHz, Methanol-d4) δ 7.39-7.26 (m, 4H), 6.38 (s, 1H), 4.09-3.99 (m, 1H), 3.95-3.85 (m, 5H), 3.86-3.70 (m, 4H), 3.64-3.52 (m, 1H), 3.28-3.10 (m, 3H), 2.89-2.80 (m, 1H), 2.55-2.45 (m, 2H), 2.40 (s, 3H), 2.33-2.22 (m, 2H), 1.86-1.60 (m, 4H).


Example 94
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-2-((ethylsulfonyl)methyl)morpholine hydrochloride (94)



embedded image


Step 1.
Synthesis of tert-butyl (2R,5S)-5-(4-chlorobenzyl)-2-((ethylthio)methyl)morpholine-4-carboxylate (94a)



embedded image


To a solution of 3e (700 mg; 1.66 mmol) in acetonitrile (3 mL) EtSNa (460 mg; 3.33 mmol) was added and the reaction mixture was stirred at 82° C. overnight. The reaction progress was monitored by LC-MS analysis. After analytical control indicated completion of the reaction, the mixture was diluted with water and the MeCN was evaporated off. An aqueous phase was extracted with DCM (3×). The combined organic phases were washed with 1 M NaOH and brine, dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was used to the next step without additional purification. Compound 94a was obtained in 99% yield (632 mg; 1.64 mmol).


ESI-MS m/z for C19H29ClNO3S found 386.1/388.1 [M+H]+; Rt=4.59 min (Method D).


Step 2.
Synthesis of tert-butyl (2R,5S)-5-(4-chlorobenzyl)-2-((ethylsulfonyl)methyl)morpholine-4-carboxylate (94b)



embedded image


To a solution of 94a (632 mg; 1.64 mmol) in DCM (6.6 mL) mCPBA (719 mg; 4.16 mmol) was added portionwise at 0° C. Then the cooling bath was removed and the mixture was stirred at room temperature overnight. The reaction progress was monitored by LC-MS analysis. Then the reaction mixture was cooled to 0° C. and the another portion of mCPBA (719 mg; 4.16 mmol) was added and stirred at room temperature for 4 hours. When analysis indicated completion of the reaction, the reaction was quenched with 10% Na2SO3 (20 mL) and stirred for 30 minutes at room temperature. Then phases were separated and the organic one was washed with 1 M NaOH (4×) and brine, dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was purified by flash column chromatography on silica (dry load, DCM/AcOEt, 100:0 to 0:100, v/v). Compound 94b was obtained in 99% yield (677 mg; 1.62 mmol).


ESI-MS m/z for C19H29ClNO5S found 418.1/420.1 [M+H]+; Rt=3.67 min (Method D).


Step 3.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-2-((ethylsulfonyl)methyl)morpholine 2,2,2-trifluoroacetate (94c)



embedded image


The title compound (94c) was obtained as a TFA salt in 99% yield (207 mg; 0.48 mmol) from 94b (200 mg; 0.48 mmol) according to the General Procedure IVb.


ESI-MS m/z for C14H21ClNO3S found 318.0/320.0 [M+H]+; Rt=1.38 min (Method D).


Step 4.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-2-((ethylsulfonyl)methyl)morpholine hydrochloride (94)



embedded image


The title compound (94) was obtained as a hydrochloride salt as a single isomer from 94c (207 mg; 0.48 mmol) and ketone 40c (100 mg; 0.48 mmol) according to the General Procedure VII in 19% yield (48 mg; 0.09 mmol). The crude product was purified by flash column chromatography on silica (DCM/AcOEt, 100:0 to 0:100, v/v, then: AcOEt/MeOH, 92:8, v/v) and then by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water+1‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 15 mL/min).


ESI-MS m/z for C25H36ClN2O3S2 found 511.0/513.0 [M+H]+; Rt=2.15 min (Method D); 1H NMR (400 MHz, D2O) δ 7.20-7.16 (m, 2H), 7.09-7.04 (m, 2H), 4.22-4.14 (m, 1H), 3.77-3.69 (m, 1H), 3.68-3.62 (m, 1H), 3.58-3.37 (m, 4H), 3.28-3.15 (m, 2H), 3.15-3.01 (m, 4H), 2.99-2.93 (m, 1H), 2.35-2.20 (m, 2H), 2.16 (s, 3H), 2.13 (s, 3H), 1.78-1.72 (m, 2H), 1.59-1.38 (m, 4H), 1.13 (t, J=7.4 Hz, 3H).


Example 95
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((ethylsulfonyl)methyl)morpholine hydrochloride (95)



embedded image


The title compound (95) was obtained as a hydrochloride salt as a single isomer from 94c (207 mg; 0.48 mmol) and ketone 2d (192 mg; 1.00 mmol) according to the General Procedure VII in 14% yield (35 mg; 0.07 mmol). The crude product was purified by flash column chromatography on silica (DCM/AcOEt, 100:0 to 40:60, v/v) and then by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water+1‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 15 mL/min).


ESI-MS m/z for C25H37ClN3O3S found 494.1/496.1 [M+H]+; Rt=2.35 min (Method D); 1H NMR (400 MHz, D2O) δ 7.19-7.10 (m, 2H), 7.08-6.99 (m, 2H), 6.06 (s, 1H), 4.17-4.10 (m, 1H), 3.72-3.65 (m, 1H), 3.64-3.61 (m, 1H), 3.59 (s, 3H), 3.51-3.40 (m, 3H), 3.35-3.27 (m, 1H), 3.25-3.14 (m, 2H), 3.09-2.99 (m, 3H), 2.96-2.88 (m, 1H), 2.62-2.51 (m, 1H), 2.27-2.12 (m, 2H), 2.07 (s, 3H), 2.01-1.89 (m, 2H), 1.48-1.23 (m, 4H), 1.09 (t, J=7.2 Hz, 3H).


Example 96
Synthesis of 3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1-methylimidazolidine-2,4-dione hydrochloride (96)



embedded image


To the solution of an alcohol 17 (133 mg; 0.25 mmol) and 1-methylhydantoin (69 mg; 0.60 mmol) in THF (3 mL) PPh3 (158 mg; 0.60 mmol) was added. Then a whole was cooled to −10° C. and DIAD (130 μL; 0.65 mmol) was added dropwise and stirred at room temperature overnight. The reaction progress was monitored by LC-MS. Then a solid was filtered off and a light yellow filtrate was concentrated in vacuo. The residue was dissolved in water/MeCN/HClaq, filtered off and taken for preparative reversed-phase column chromatography (first: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 98:2 to 30:70.30 min, 20 mL/min, second: C-18, water+0.3‰ HCl (36%)/MeCN, 98:2 to 20:80, 30 min, 18 mL/min). The title compound (96) was obtained as a hydrochloride salt as a single isomer in 36% yield (51 mg; 0.09 mmol).


ESI-MS m/z for C27H37ClN5O3 found 514.4/516.4 [M+H]+; Rt=0.99 min (Method D); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.41-7.36 (m, 2H), 7.32-7.28 (m, 2H), 5.83 (s, 1H), 4.11-4.03 (m, 1H), 4.01-3.94 (m, 2H), 3.79-3.74 (m, 1H), 3.73-3.68 (m, 2H), 3.66-3.62 (m, 1H), 3.61 (s, 3H), 3.56-3.51 (m, 2H), 3.47-3.44 (m, 1H), 3.19-3.09 (m, 3H), 2.88 (s, 3H), 2.48-2.44 (m, 1H), 2.28-2.21 (m, 2H), 2.17 (s, 3H), 2.07-2.01 (m, 2H), 1.59-1.52 (m, 2H), 1.46-1.40 (m, 2H).


Example 97
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-(1-methyl-1H-imidazol-4-yl)morpholine-2-carboxamide hydrochloride (97)



embedded image


The title compound (97) was obtained as a hydrochloride salt as a single isomer from 40 (43 mg; 0.089 mmol) and from 1-methyl-1H-imidazol-4-amine hydrochloride (15 mg; 0.107 mmol) according to the General Procedure V in 30% yield (15 mg; 0.027 mmol). The crude product was purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 98:2 to 20:80, 30 min, 18 mL/min).


ESI-MS m/z for C27H35ClN5O2S found 528.4/530.4 [M+H]+; Rt=1.02 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 8.31 (s, 1H), 7.43 (d, J=1.7 Hz, 1H), 7.41 (d, JAA′=8.5 Hz, 2H), 7.36 (d, JBB′=8.5 Hz, 2H), 4.63-4.61 (m, 1H), 3.88-3.86 (m, 2H), 3.75-3.73 (m, 4H), 3.66-3.64 (m, 1H), 3.57-3.53 (m, 1H), 3.51-3.47 (m, 1H), 3.16-3.09 (m, 2H), 2.89 (tt, J=12.1 Hz, J=3.5 Hz, 1H), 2.27-2.24 (m, 5H), 2.18-2.13 (m, 5H), 1.68-1.58 (m, 2H), 1.56-1.50 (m, 2H).


Example 98
Synthesis of 4-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)butanoic acid hydrochloride (98)



embedded image


To a solution of 83 (35 mg; 0.082 mmol) in MeOH (1.5 mL) CuSO4×5 H2O (11 mg; 0.041 mmol) was added and after 5 minutes sodium L-ascorbate (17 mg; 0.082 mmol) was added in one portion and the mixture was stirred at room temperature for 25 minutes. Then to this mixture a solution of 4-azidobutanoic acid (21 mg; 0.164 mmol) in MeOH (0.5 mL) was added and the mixture was stirred at room temperature for 2.5 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the solvent was evaporated in vacuo and the residue was dissolved in MeCN/water with HClaq. An insoluble acid was filtered off and the filtrate was taken for the preparative reversed-phase column chromatography (C-18, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 19 mL/min). The title compound (98) was obtained as a single isomer as a hydrochloride salt in 49% yield (24 mg; 0.040 mmol).


ESI-MS m/z for C28H37ClN5O3S found 558.4/560.4 [M+H]+; Rt=1.03 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 8.21 (s, 1H), 7.38 (d, JAA′=8.6 Hz, 2H), 7.34 (d, JBB′=8.6 Hz, 2H), 5.08-5.06 (m, 1H), 4.41 (t, J=7.1 Hz, 2H), 3.95-3.93 (m, 1H), 3.86-3.84 (m, 1H), 3.72-3.68 (m, 2H), 3.62-3.56 (m, 2H), 3.25 (dd, J=13.9 Hz, J=10.7 Hz, 1H), 3.19 (dd, J=13.9 Hz, J=4.3 Hz, 1H), 2.88 (tt, J=12.1 Hz, J=3.6 Hz, 1H), 2.35-2.26 (m, 7H), 2.19-2.17 (m, 5H), 2.10-2.06 (m, 2H), 1.68-1.52 (m, 4H).


Example 99
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-hydroxymorpholine-2-carboxamide hydrochloride (99)



embedded image


The title compound (99) was obtained as a hydrochloride salt as a single isomer from 40 (43 mg; 0.089 mmol) and from hydroxylamine hydrochloride (8 mg; 0.107 mmol) according to the General Procedure V in 13% yield (6 mg; 0.012 mmol). The crude product was purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 98:2 to 30:70, 30 min, 18 mL/min).


ESI-MS m/z for C23H31ClN3O3S found 464.3/466.3 [M+H]+; Rt=0.96 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.6 Hz, 2H), 7.34 (d, JBB′=8.6 Hz, 2H), 4.43-4.42 (m, 1H), 3.86-3.84 (m, 1H), 3.79-3.77 (m, 1H), 3.68 (dd, J=13.3 Hz, J=2.3 Hz, 1H), 3.57-3.54 (m, 2H), 3.40-3.37 (m, 1H), 3.16-3.10 (m, 2H), 2.94 (tt, J=12.2 Hz, J=3.6 Hz, 1H), 2.29-2.16 (m, 10H), 1.75-1.65 (m, 2H), 1.59-1.52 (m, 2H).


Example 100
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-N-(1,5-dimethyl-1H-pyrazol-3-yl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carboxamide hydrochloride (100)



embedded image


The title compound (100) was obtained as a hydrochloride salt as a single isomer from 40 (43 mg; 0.089 mmol) and from 1,5-dimethyl-1H-pyrazol-3-amine (12 mg; 0.110 mmol) according to the General Procedure V in 45% yield (23 mg; 0.040 mmol). The crude product was purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 98:2 to 10:90, 30 min, 18 mL/min).


ESI-MS m/z for C28H37ClN5O2S found 542.5/544.5 [M+H]+; Rt=1.17 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.6 Hz, 2H), 7.35 (d, JBB′=8.6 Hz, 2H), 6.25 (s, 1H), 4.51-4.49 (m, 1H), 3.83 (dd, J=13.1 Hz, J=2.1 Hz, 1H), 3.79-3.74 (m, 2H), 3.61-3.60 (m, 4H), 3.54-3.50 (m, 2H), 3.17-3.12 (m, 2H), 2.87 (tt, J=12.2 Hz, J=3.7 Hz, 1H), 2.28-2.14 (m, 13H), 1.67-1.59 (m, 2H), 1.58-1.50 (m, 2H).


Example 101
Synthesis of 5-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)pentanoic acid hydrochloride (101)



embedded image


To a solution of 83 (35 mg; 0.082 mmol) in MeOH (3 mL) CuSO4×5 H2O (11 mg; 0.041 mmol) was added and after 5 minutes sodium L-ascorbate (17 mg; 0.082 mmol) was added in one portion and the mixture was stirred at room temperature for 25 minutes. Then to this mixture 5-azidopentanoic acid (24 mg; 0.164 mmol) was added and the mixture was stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the solvent was evaporated in vacuo and the residue was dissolved in MeCN/water with HClaq. An insoluble acid was filtered off and the filtrate was taken for the preparative reversed-phase column chromatography (C-18, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min). The title compound (101) was obtained as a single isomer as a hydrochloride salt in 30% yield (15 mg; 0.025 mmol).


ESI-MS m/z for C29H39ClN5O3S found 572.4/574.4 [M+H]+; Rt=1.07 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 8.19 (s, 1H), 7.38 (d, JAA′=8.6 Hz, 2H), 7.33 (d, JBB′=8.6 Hz, 2H), 5.07-5.05 (m, 1H), 4.38 (t, J=7.0 Hz, 2H), 3.94-3.92 (m, 1H), 3.85-3.83 (m, 1H), 3.72-3.67 (m, 2H), 3.62-3.56 (m, 2H), 3.24 (dd, J=13.9 Hz, J=10.7 Hz, 1H), 3.20-3.18 (m, 1H), 2.88 (tt, J=12.0 Hz, J=3.7 Hz, 1H), 2.35-2.30 (m, 2H), 2.26-2.24 (m, 5H), 2.19-2.17 (m, 5H), 1.89-1.85 (m, 2H), 1.67-1.49 (m, 6H).


Example 102
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-N-(1,5-dimethyl-1H-pyrazol-3-yl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamide hydrochloride (102)



embedded image


The title compound (102) was obtained as a hydrochloride salt as a single isomer from 11 (35 mg; 0.064 mmol) and from 1,5-dimethyl-1H-pyrazol-3-amine (9 mg; 0.080 mmol) according to the General Procedure V in 39% yield (16 mg; 0.025 mmol). The crude product was purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 98:2 to 30:70, 30 min, 18 mL/min).


ESI-MS m/z for C28H38ClN6O2S found 525.5/527.5 [M+H]+; Rt=1.04 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.5 Hz, 2H), 7.35 (d, JBB′=8.5 Hz, 2H), 6.24 (s, 1H), 5.82 (s, 1H), 4.51-4.50 (m, 1H), 3.85-3.82 (m, 1H), 3.80-3.74 (m, 2H), 3.62-3.60 (m, 7H), 3.51-3.46 (m, 2H), 3.17-3.11 (m, 2H), 2.47 (tt, J=12.0 Hz, J=3.7 Hz, 1H), 2.25-2.16 (m, 8H), 2.06-2.01 (m, 2H), 1.63-1.54 (m, 2H), 1.47-1.38 (m, 2H).


Example 103
Synthesis of N-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carbonyl)-N-methylglycine hydrochloride (103)



embedded image


Step 1.
Synthesis of methyl N-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carbonyl)-N-methylglycinate hydrochloride (103a)



embedded image


The title compound (103a) was obtained as a hydrochloride salt from 40 (35 mg; 0.072 mmol) and from methyl methylglycinate hydrochloride (13 mg; 0.094 mmol) according to the General Procedure V. The crude product was purified by preparative reversed-phase column chromatography (C-18, water+0.3‰ HCl (36%)/MeCN, 98:2 to 10:90, 30 min, 18 mL/min) and was taken to the next step.


ESI-MS m/z for C2H37ClN3O4S found 534.2/536.1 [M+H]+; Rt=1.10 min (Method A)


Step 2.
Synthesis of N-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carbonyl)-N-methylglycine hydrochloride (103)



embedded image


To the aqueous solution of 103a (the crude product) in MeOH (2 mL) 4 M NaOH (0.5 mL) was added and whole was stirred at room temperature for 2 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the solvent was evaporated in vacuo and the residue was purified by preparative reversed-phase column chromatography (C-18, water+0.3‰ HCl (36%)/MeCN, 98:2 to 10:90, 30 min, 18 mL/min). The title compound (103) was obtained as a single isomer as a hydrochloride salt in 19% yield (per two steps)(8 mg; 0.014 mmol).


ESI-MS m/z for C26H35ClN3O4S found 520.4/522.4 [M+H]+; Rt=1.04 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 7.38 (d, JAA′=8.2 Hz, 2H), 7.31-7.29 (m, 2H), 4.81-4.74 (m, 1H), 4.20-4.16 (m, 1H), 4.10-3.99 (m, 1H), 3.87-3.80 (m, 2H), 3.74-3.54 (m, 4H), 3.23-3.21 (m, 1H), [3.09 (s, 1.8H), 2.92 (s, 1.2H), rotamers, 3H], 3.02-2.97 (m, 1H), 2.87 (tt, J=12.5 Hz, J=3.3 Hz, 1H), 2.26-2.11 (m, 10H), 1.72-1.68 (m, 1H), 1.64-1.52 (m, 2H), 1.47-1.44 (m, 1H).


Example 104
Synthesis of (2R,5S)-4-(4-(1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)-5-(4-(trifluoromethyl)benzyl)morpholine 2,2,2-trifluoroacetate (104)



embedded image


Step 1.
Synthesis of (S)-2-amino-3-(4-(trifluoromethyl)phenyl)propan-1-ol (104a)



embedded image


The title compound (104a) was obtained from optically pure (S)-2-amino-3-(4-(trifluoromethyl)phenyl)propanoic acid (7 g; 30 mmol) according to the General Procedure Ia in 99% yield (6.5 g; 29.7 mmol).


ESI-MS m/z for C10H13F3NO found 220.1 [M+H]+; Rt=1.17 min (Method A)


Step 2.
Synthesis of 2-bromo-3-(tert-butoxy)-N—((S)-1-hydroxy-3-(4-(trifluoromethyl)phenyl)propan-2-yl)propanamide (104b)



embedded image


The title compound (104b) was obtained from 104a (7.3 g; 33.3 mmol) according to the General Procedure V in 89% yield (12.66 g; 29.78 mmol).


ESI-MS m/z for C17H24BrF3NO3 found 426.1 [M+H]+; Rt=1.43 min (Method A)


Step 3.
Synthesis of (2S,5S)-2-(tert-butoxymethyl)-5-(4-(trifluoromethyl)benzyl)morpholin-3-one (104c)



embedded image


The title compound (104c) was obtained from 104b (12.66 g; 29.78 mmol) according to the General Procedure II in 99% yield (10.18 g; 29.48 mmol).


ESI-MS m/z for C17H23F3NO3 found 346.2 [M+H]+; Rt=1.43 min (Method A)


Step 4.
Synthesis of ((2R,5S)-5-(4-(trifluoromethyl)benzyl)morpholin-2-yl)methanol (104d)



embedded image


The title compound (104d) was obtained from 104c (10.18 g; 29.48 mmol) according to the General Procedure Ib in 99% yield (8.03 g; 29.19 mmol).


ESI-MS m/z for C13H17F3NO2 found 276.1 [M+H]+; Rt=0.55 min (Method A)


Step 5.
Synthesis of tert-butyl (2R,5S)-2-(hydroxymethyl)-5-(4-(trifluoromethyl)benzyl)morpholine-4-carboxylate (104e)



embedded image


To a solution of an amino alcohol 104d (8.03 g; 29.19 mmol) in 4 N NaOH (110 mL) the solution of di-tert-butyl dicarbonate (Boc2O) (9.45 g; 43.3 mmol) in THF (50 mL) was added and the reaction mixture was stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the phases were separated and an aqueous one was extracted with Et2O (3×100 mL). The combined organic solutions were dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the residue was purified by silica-gel column chromatography (hexane/AcOEt, 1:5 to 2.5:1, v/v). Compound 104e was obtained as a colorless oil in 99% yield (10.84 g; 28.9 mmol).


ESI-MS m/z for C18H25F3NO4 found 376.2 [M+H]+; Rt=1.45 min (Method A)


Step 6.
Synthesis of tert-butyl (2R,5S)-2-(((methylsulfonyl)oxy)methyl)-5-(4-(trifluoromethyl)benzyl)-morpholine-4-carboxylate (104f)



embedded image


The title compound (104f) was obtained from 104e (2.1 g; 5.59 mmol) according to the General Procedure VIII in 99% yield (2.51 g; 5.53 mmol).


ESI-MS m/z for C19H27F3NO6S found 454.1 [M+H]+; Rt=1.70 min (Method A)


Step 7.
Synthesis of tert-butyl (2R,5S)-2-((methylthio)methyl)-5-(4-(trifluoromethyl)benzyl)morpholine-4-carboxylate (104g)



embedded image


To a solution of 104f (2.51 g; 5.53 mmol) in acetonitrile (20 mL) MeSNa (1 g; 14 mmol) was added and the reaction mixture was stirred at 80° C. overnight. The reaction progress was monitored by LC-MS analysis. After analytical control indicated completion of the reaction, the mixture was dissolved in Et2O and then washed with 1 M NaOH and brine, extracted with Et2O (3×70 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was used to the next step without additional purification. Compound 104g was obtained in 99% yield (2.22 g; 5.47 mmol).


ESI-MS m/z for C19H27F3NO3S found 406.2 [M+H]+; Rt=1.48 min (Method A)


Step 8.
Synthesis of tert-butyl (2R,5S)-2-((methylsulfonyl)methyl)-5-(4-(trifluoromethyl)benzyl)-morpholine-4-carboxylate (104h)



embedded image


To the pre-cooled (0° C.) solution of 104g (2.22 g; 5.47 mmol) in DCM (40 mL) mCPBA (4.13 g; 16.77 mmol) was added in several portions. The reaction was stirred at room temperature for 3 hours. The reaction progress was monitored by LC-MS analysis. After analytical control indicated completion of the reaction, the reaction was washed with 10% Na2S2O3, saturated NaHCO3, brine, dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was used to the next step without additional purification. Compound 104h was obtained in 99% yield (2.37 g; 5.42 mmol).


ESI-MS m/z for C19H27F3NOS found 438.2 [M+H]+; Rt=1.55 min (Method A)


Step 9.
Synthesis of (2R,5S)-2-((methylsulfonyl)methyl)-5-(4-(trifluoromethyl)benzyl)morpholine (1041)



embedded image


The title compound (1041) was obtained as a free base in 86% yield (1.57 g; 4.66 mmol) from 104h (2.37 g; 5.42 mmol) according to the General Procedure IVb and then after basic (4 M NaOH) extraction with DCM.


Step 10.
Synthesis of 1-methyl-3-(1,4-dioxaspiro[4.5]dec-7-en-8-yl)-1H-1,2,4-triazole (104j)



embedded image


The solution of 4,4,5,5-tetramethyl-2-(1,4-dioxaspiro[4.5]dec-7-en-8-yl)-1,3,2-dioxaborolane (1.02 g; 3.80 mmol), 3-bromo-1-methyl-1H-1,2,4-triazole (650 mg, 4.00 mmol), [PPh3]4Pd (130 mg; 0.11 mmol) in mixture of dioxane (16 mL) and 2 M aqueous solution of K2CO3 (4 mL) was heated at 100° C. for 24 hours under an argon atmosphere. LC-MS showed complete consumption of the starting material. Then AcOEt (40 mL) was added and this mixture was washed with water and brine. An aqueous layer was extracted with chloroform. The combined organic layers were concentrated in vacuo and the residue was purified by flash column chromatography on silica (hexane/AcOEt, 1:1, v/v, then: AcOEt/MeOH, 140:1 to 15:1, v/v). Compound 104j was obtained as pale yellow oil in 69% yield (580 mg; 2.62 mmol).


ESI-MS m/z for C11H16N3O2 found 222.0 [M+H]+; Rt=0.56 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 8.27-8.23 (m, 1H), 6.52-6.48 (m, 1H), 3.95-3.92 (m, 4H), 3.83 (s, 3H), 2.62-2.58 (m, 2H), 2.39-2.36 (m, 2H), 1.82-1.76 (m, 2H).


Step 11.
Synthesis of 1-methyl-3-(1,4-dioxaspiro[4.5]decan-8-yl)-1H-1,2,4-triazole (104k)



embedded image


To the solution of 104j (570 mg; 2.57 mmol) in AcOEt (15 mL) 10% Pd/C (5 mol %; 137 mg, 0.13 mmol) was added and the reaction mixture was conducted under hydrogen atmosphere at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, Pd/C was filtered off through the Celite pad and the solvents were evaporated in vacuo. The crude product was used to the next step without additional purification.


ESI-MS m/z for C11H18N3O2 found 224.2 [M+H]+; Rt=0.45 min (Method A)


Step 12.
Synthesis of 4-(1-methyl-1H-1,2,4-triazol-3-yl)cyclohexan-1-one (1041)



embedded image


The solution of 104k (the crude product) in 1 M HCl (15 mL) was stirred at room temperature for overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, to this mixture an aqueous solution of NaOH (4 M) was added to pH 13 and then extracted with CHCl3 (2×). The combined organic solutions were dried over anhydrous MgSO4, filtered and concentrated in vacuo. The crude product was used to the next step without additional purification. Compound 1041 was obtained in 92% yield (per two steps)(424 mg; 2.37 mmol).


ESI-MS m/z for C9H14N3O found 180.2 [M+H]+; Rt=0.27 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 300 K) δ 8.33 (s, 1H), 3.78 (s, 3H), 3.19-3.12 (m, 1H), 2.49-2.45 (m, 2H), 2.31-2.26 (m, 2H), 2.24-2.16 (m, 2H), 1.92-1.84 (m, 2H).


Step 13.
Synthesis of (2R,5S)-4-(4-(1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)-5-(4-(trifluoromethyl)benzyl)morpholine 2,2,2-trifluoroacetate (104)



embedded image


The title compound (104) was obtained as a TFA salt as a single isomer from 1041 (135 mg; 0.40 mmol) and ketone 104l (72 mg; 0.40 mmol) according to the General Procedure VII in 15% yield (35 mg; 0.06 mmol). The crude product was purified by silica-gel column chromatography (DCM/MeOH, 200:1 to 20:1, v/v) and then by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 99:1 to 50:50, 30 min, 18 mL/min).


ESI-MS m/z for C23H32F3N4O3S found 501.3 [M+H]+; Rt=0.83 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 8.28-8.24 (m, 1H), 7.70-7.66 (m, 2H), 7.55-7.52 (m, 2H), 4.34-4.27 (m, 1H), 3.90-3.85 (m, 1H), 3.84-3.80 (m, 1H), 3.78 (s, 3H), 3.71-3.66 (m, 1H), 3.63-3.57 (m, 2H), 3.54-3.48 (m, 1H), 3.45-3.44 (m, 1H), 3.36-3.29 (m, 2H), 3.26-3.21 (m, 1H), 3.04 (s, 3H), 2.70-2.64 (m, 1H), 2.33-2.25 (m, 2H), 2.20-2.11 (m, 2H), 1.62-1.50 (m, 4H).


Example 105
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-imidazol-4-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine 2,2,2-trifluoroacetate (105)



embedded image


The title compound (105) was obtained as a TFA salt as a single isomer in 1% overall yield in a similar way to Example 15 with the exception that, in the first step of the synthesis, 4-bromo-1-methyl-1H-imidazole was used instead of 3-bromo-1,5-dimethyl-1H-1,2,4-triazole.


ESI-MS m/z for C23H33ClN3O3S found 466.2/468.2 [M+H]+; Rt=0.96 min (Method A); 1H NMR (700 MHz, Methanol-d4) δ 8.82-8.80 (m, 1H), 7.41-7.38 (m, 2H), 7.38-7.36 (m, 1H), 7.35-7.31 (m, 2H), 4.41-4.32 (m, 1H), 3.95-3.91 (m, 1H), 3.90 (d, J=0.4 Hz, 3H), 3.87-3.80 (m, 2H), 3.77-3.71 (m, 1H), 3.66-3.55 (m, 2H), 3.50-3.40 (m, 2H), 3.37-3.32 (m, 1H), 3.20-3.16 (m, 1H), 3.10 (s, 3H), 2.87-2.77 (m, 1H), 2.54-2.45 (m, 2H), 2.36-2.26 (m, 2H), 1.76-1.55 (m, 4H).


Example 106
Synthesis of (2R,5S)-4-(4-(1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-5-(4-(trifluoromethyl)-benzyl)morpholine-2-carboxylic acid 2,2,2-trifluoroacetate (106)



embedded image


The title compound (106) was obtained as a TFA salt as a single isomer in 2% overall yield in a similar way to Example 65 with the exception that, in the first step of the synthesis, the compound 104e was used instead of the compound 63e and in the third step of the synthesis, ketone 1041 was used instead of ketone 2d. The crude product was purified by preparative reversed-phase column chromatography (C-18, water/MeCN+1‰ TFA, 99:1 to 40:60, 30 min, 20 mL/min).


ESI-MS m/z for C22H28F3N4O3 found 453.3 [M+H]+; Rt=0.90 min (Method A); 1H NMR (700 MHz, D2O) δ 9.22 (s, 1H), 7.82-7.78 (m, 2H), 7.62-7.58 (m, 2H), 4.69-4.55 (m, 1H), 4.12-4.08 (m, 1H), 4.07 (s, 3H), 3.99-3.81 (m, 3H), 3.79-3.51 (m, 2H), 3.43-3.31 (m, 2H), 3.09-3.02 (m, 1H), 2.62-2.30 (m, 4H), 1.84-1.61 (m, 4H).


Example 107
Synthesis of (2S,5S)-5-(4-chlorobenzyl)-2-methyl-4-(4-(5-methyl-1H-pyrazol-3-yl)cyclohexyl)-morpholine 2,2,2-trifluoroacetate (107)



embedded image


The title compound (107) was obtained as a TFA salt as a single isomer in 9% overall yield in a similar way to Example 2 with the exception that, in the third step of the synthesis, methylhydrazine was used instead of hydrazine monohydrate. The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 1:1 to 0:1, v/v, then: AcOEt/MeOH, 10:1, v/v) and then purified by preparative reversed-phase column chromatography (C-18, water/MeCN+1‰ TFA, 90:10 to 10:90, 30 min, 20 mL/min).


ESI-MS m/z for C22H31ClN3O found 388.1/390.1 [M+H]+; Rt=0.95 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.4 Hz, 2H), 7.31 (d, JBB′=8.4 Hz, 2H), 5.84 (s, 1H), 3.85-3.83 (m, 1H), 3.73-3.69 (m, 2H), 3.60-3.59 (m, 1H), 3.47-3.44 (m, 2H), 3.17-3.14 (m, 1H), 3.08 (dd, J=13.7 Hz, J=3.8 Hz, 1H), 3.03-3.01 (m, 1H), 2.57-2.53 (m, 1H), 2.32-2.29 (m, 1H), 2.27-2.24 (m, 1H), 2.16 (s, 3H), 2.09-2.05 (m, 2H), 1.56-1.42 (m, 4H), 1.23 (d, J=6.2 Hz, 3H).


Example 108
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine 2,2,2-trifluoroacetate (108)



embedded image


The title compound (108) was obtained as a TFA salt as a single isomer from 3h (288 mg; 0.95 mmol) and ketone 1041 (170 mg; 0.95 mmol) according to the General Procedure VII in 14% yield (75 mg; 0.13 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 0:100, v/v, then: AcOEt/MeOH, 50:1 to 2:1, v/v) and then transferred into TFA salt with using TFA/DCM and then purified by preparative reversed-phase column chromatography (C-18, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C22H32ClN4O3S found 467.1/469.1 [M+H]+; Rt=0.78 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 8.22 (s, 1H), 7.37-7.38 (m, 2H), 7.32-7.31 (m, 2H), 4.29-4.25 (m, 1H), 3.79-3.77 (m, 5H), 3.68 (d, J=12.1 Hz, 1H), 3.59-3.54 (m, 2H), 3.48-3.45 (m, 1H), 3.41-3.39 (m, 1H), 3.29 (t, J=12.0 Hz, 1H), 3.21-3.17 (m, 1H), 3.11 (dd, J=13.8 Hz, J=4.3 Hz, 1H), 3.03 (s, 3H), 3.68-3.63 (m, 1H), 2.28-2.26 (m, 2H), 2.16-2.12 (m, 2H), 1.60-1.51 (m, 4H).


Example 109
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-morpholine-2-carboxylic acid 2,2,2-trifluoroacetate (109)



embedded image


The title compound (109) was obtained as a TFA salt as a single isomer in 37% overall yield in a similar way to Example 65 with the exception that, in the first step of the synthesis, the compound 3d was used instead of the compound 63e and in the third step of the synthesis, ketone 1041 was used instead of ketone 2d. The crude product was purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 98:2 to 30:70, 30 min, 20 mL/min).


ESI-MS m/z for C21H28ClN4O3 found 419.0/421.0 [M+H]+; Rt=0.75 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 8.21 (s, 1H), 7.37 (d, JAA′=8.4 Hz, 2H), 7.31 (d, JBB′=8.4 Hz, 2H), 4.46-4.45 (m, 1H), 3.77 (s, 3H), 3.75-3.70 (m, 3H), 3.53-3.44 (m, 3H), 3.15-3.12 (m, 1H), 3.02 (dd, J=13.9 Hz, J=9.8 Hz, 1H), 2.65 (tt, J=11.7 Hz, J=3.6 Hz, 1H), 2.21-2.09 (m, 4H), 1.64-1.48 (m, 4H).


Example 110
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-1,2,3-triazol-4-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine 2,2,2-trifluoroacetate (110)



embedded image


Step 1.
Synthesis of 4-(1,4-dioxaspiro[4.5]decan-8-yl)-1-((trimethylsilyl)methyl)-1H-1,2,3-triazole (110a)



embedded image


To a solution of 8-ethynyl-1,4-dioxaspiro[4.5]decane (458 mg; 2.76 mmol) in MeOH (23 mL) CuSO4×5 H2O (350 mg; 1.40 mmol) was added and after 5 minutes sodium L-ascorbate (555 mg; 2.80 mmol) was added in one portion and the mixture was stirred at room temperature for 15 minutes. Then to an yellow mixture trimethylsilylmethyl azide (413 μL; 2.80 mmol) was added and the mixture was stirred at room temperature for 2 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the green suspension was concentrated in vacuo and the residue was taken between water/DCM. An organic layer was washed with brine, dried over anhydrous MgSO4, filtered and concentrated in vacuo and the crude product mixture was purified by flash column chromatography on silica (AcOEt/MeOH, 100:1, v/v). Compound 110a was obtained in 92% yield (750 mg; 2.54 mmol).


ESI-MS m/z for C14H26N3O2Si found 296.2 [M+H]+; Rt=1.28 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 7.64 (s, 1H), 3.90 (s, 2H), 3.87-3.82 (m, 4H), 2.71-2.66 (m, 1H), 1.93-1.88 (m, 2H), 1.71-1.66 (m, 2H), 1.64-1.54 (m, 4H).


Step 2.
Synthesis of 4-(1-((trimethylsilyl)methyl)-1H-1,2,3-triazol-4-yl)cyclohexan-1-one (110b)



embedded image


The solution of 110a (750 mg; 2.54 mmol) and 1 M KHSO4 (10 mL) was stirred at room temperature overnight. The reaction progress was monitored by LC-MS analysis. After analytical control indicated completion of the reaction, to this mixture 4 M NaOH was added to pH 12. Then an aqueous suspension was extracted with DCM (2×). The combined organic solutions were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude ketone was used to the next step without additional purification. Compound 110b was obtained in 68% yield (435 mg; 1.73 mmol).


ESI-MS m/z for C12H22N3OSi found 252.1 [M+H]+; Rt=1.12 min (Method A)


Step 3.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)-4-(4-(1-((trimethylsilyl)-methyl)-1H-1,2,3-triazol-4-yl)cyclohexyl)morpholine (110c)



embedded image


The title compound (110c) was obtained from 3h (299 mg; 0.88 mmol) and ketone 110b (200 mg; 0.79 mmol) according to the General Procedure VII in 30% yield (130 mg; 0.24 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 5:4 to 0:100, v/v, then: AcOEt/MeOH, 50:1, v/v).


ESI-MS m/z for C25H40ClN4O3SSi found 539.1/541.1 [M+H]+; Rt=1.11 min (Method A)


Step 4.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-1,2,3-triazol-4-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine 2,2,2-trifluoroacetate (110)



embedded image


To a solution of 110c (130 mg; 0.240 mmol) in THF (3 mL) at −70° C. TBAF (1 M in THF; 25 μL; 0.025 mmol) was added and the cooling bath was removed and the mixture was stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the reaction mixture was evaporated in vacuo and the crude product was purified by preparative reversed-phase column chromatography (C-18, water/MeCN+1‰ TFA, 98:2 to 30:70, 30 min, 20 mL/min). The title compound 110 was obtained as a TFA salt as a single isomer in 80% yield (112 mg; 0.193 mmol).


ESI-MS m/z for C22H32ClN4O3S found 467.0/469.0 [M+H]+; Rt=0.81 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.68 (s, 1H), 7.37 (d, JAA′=8.4 Hz, 2H), 7.31 (d, JBB′=8.4 Hz, 2H), 4.28-4.24 (m, 1H), 3.97 (s, 3H), 3.78-3.74 (m, 2H), 3.67 (d, J=12.5 Hz, 2H), 3.57 (dd, J=15.0 Hz, J=7.7 Hz, 1H), 3.54-3.52 (m, 1H), 3.44-3.38 (m, 2H), 3.28-2.24 (m, 1H), 3.18 (dd, J=13.7 Hz, J=10.7 Hz, 1H), 3.09 (dd, J=13.7 Hz, J=4.2 Hz, 1H), 3.02 (s, 3H), 2.67 (tt, J=11.6 Hz, J=3.5 Hz, 1H), 2.27-2.25 (m, 2H), 2.15-2.12 (m, 2H), 1.58-1.46 (m, 4H).


Example 111
Synthesis of methyl (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-1,2,3-triazol-4-yl)cyclohexyl)morpholine-2-carboxylate 2,2,2-trifluoroacetate (111)



embedded image


The title compound (111) was obtained as a TFA salt as a single isomer in 9% overall yield in a similar way to Example 110 with the exception that, in the third step of the synthesis, the compound 9b was used instead of the compound 3h and in this reaction AcOH was used instead of Et3N. The crude product was purified by flash column chromatography on silica (AcOEt/MeOH, 100:0 to 10:1, v/v) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 90:10 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C22H30ClN4O3 found 433.2/435.2 [M+H]+; Rt=0.83 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.66 (s, 1H), 7.36-7.35 (m, 2H), 7.29-7.28 (m, 2H), 4.49-4.47 (m, 1H), 3.96 (s, 3H), 3.74 (s, 3H), 3.68 (d, J=3.3 Hz, 2H), 3.60-3.58 (m, 1H), 3.39-3.36 (m, 3H), 3.07 (dd, J=13.8 Hz, J=4.2 Hz, 1H), 2.94 (dd, J=13.8 Hz, J=9.9 Hz, 1H), 2.65 (tt, J=12.0 Hz, J=3.4 Hz, 1H), 2.13-2.07 (m, 4H), 1.61-1.38 (m, 4H).


Example 112
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-1,2,3-triazol-4-yl)cyclohexyl)-morpholine-2-carboxylic acid 2,2,2-trifluoroacetate (112)



embedded image


To a solution of 111 (73 mg; 0.134 mmol) in MeOH (2 mL) 1 M NaOH (1 mL) was added and the mixture was stirred at room temperature for 3 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, methanol was evaporated off and the residue was dissolved in MeCN/water and this mixture was acidified to pH 2 by addition of TFA. The crude product was purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 98:2 to 30:70, 30 min, 20 mL/min). The title compound 112 was obtained as a TFA salt as a single isomer in 42% yield (30 mg; 0.056 mmol).


ESI-MS m/z for C21H28ClN4O3 found 419.1/421.1 [M+H]+; Rt=0.83 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 7.72 (s, 1H), 7.31 (d, JAA′=8.4 Hz, 2H), 7.22 (d, JBB′=8.4 Hz, 2H), 3.94 (s, 3H), 3.92 (dd, J=8.4 Hz, J=3.3 Hz, 1H), 3.50 (dd, J=11.4 Hz, J=3.9 Hz, 1H), 3.34 (dd, J=11.3 Hz, J=2.5 Hz, 1H), 2.95-2.90 (m, 2H), 2.79-2.71 (m, 3H), 2.60-2.65 (m, 1H), 2.56 (tt, J=12.3 Hz, J=3.6 Hz, 1H), 2.00-1.94 (m, 3H), 1.92-1.89 (m, 1H), 1.42-1.23 (m, 4H).


Example 113
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-4-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine 2,2,2-trifluoroacetate (113)



embedded image


The title compound (113) was obtained as a TFA salt as a single isomer in 9% overall yield in a similar way to Example 15 with the exception that, in the first step of the synthesis, 4-bromo-1,5-dimethyl-1H-pyrazole was used instead of 3-bromo-1,5-dimethyl-1H-1,2,4-triazole and Pd(dppf)2Cl2 was used instead of [PPh3]4Pd. The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 1:1 to 0:100, v/v; then: AcOEt/MeOH, 20:1, v/v) and then by preparative normal-phase column chromatography (Silica-based HPLC column, Hex:IPA, 1.5:1 (v/v), isocratic elution) and then by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C24H35ClN3O3S found 480.2/482.2 [M+H]+; Rt=0.89 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.41-7.37 (m, 2H), 7.34-7.31 (m, 2H), 7.14 (s, 1H), 4.31-4.23 (m, 1H), 3.81-3.76 (m, 2H), 3.70-3.67 (m, 1H), 3.65 (s, 3H), 3.61-3.54 (m, 2H), 3.49-3.45 (m, 1H), 3.40-3.38 (m, 1H), 3.32-3.26 (m, 1H), 3.22-3.18 (m, 1H), 3.14-3.09 (m, 1H), 3.03 (s, 3H), 2.43-2.35 (m, 1H), 2.28-2.22 (m, 2H), 2.16 (s, 3H), 1.95-1.88 (m, 2H), 1.58-1.49 (m, 2H), 1.49-1.41 (m, 2H).


Example 114
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine 2,2,2-trifluoroacetate (114)



embedded image


The title compound (114) was obtained as a TFA salt as a single isomer in 12% overall yield in a similar way to Example 15 with the exception that, in the first step of the synthesis, 3-bromo-1-methyl-1H-pyrazole was used instead of 3-bromo-1,5-dimethyl-1H-1,2,4-triazole. The crude product was purified by flash column chromatography on silica (pentane/AcOEt, 1:1 to 0:100, v/v; then: AcOEt/MeOH, 50:1 to 20:1, v/v) and then by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C23H33ClN3O3S found 466.0/468.0 [M+H]+; Rt=0.95 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.46 (d, J=2.2 Hz, 1H), 7.38 (d, JAA′=8.5 Hz, 2H), 7.32 (d, JBB′=8.5 Hz, 2H), 6.02 (d, J=2.2 Hz, 1H), 4.28-4.25 (m, 1H), 3.80-3.77 (m, 2H), 3.73 (s, 3H), 3.68-3.67 (m, 1H), 3.61-3.56 (m, 2H), 3.46-3.41 (m, 2H), 3.30-3.26 (m, 1H), 3.19 (dd, J=13.7 Hz, J=11.0 Hz, 1H), 3.11 (dd, J=13.8 Hz, J=4.2 Hz, 1H), 3.03 (s, 3H), 2.57-2.53 (m, 1H), 2.27-2.25 (m, 2H), 2.10-2.06 (m, 2H), 1.56-1.44 (m, 4H).


Example 115
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(2-methyl-2H-1,2,3-triazol-4-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine 2,2,2-trifluoroacetate (115)



embedded image


The title compound (115) was obtained as a TFA salt as a single isomer in 14% overall yield in a similar way to Example 15 with the exception that, in the first step of the synthesis, 4-bromo-2-methyl-2H-1,2,3-triazole was used instead of 3-bromo-1,5-dimethyl-1H-1,2,4-triazole. The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 1:1 to 1:7, v/v; then: AcOEt/MeOH, 70:1, v/v) and then by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C22H32ClN4O3S found 467.0/469.0 [M+H]+; Rt=0.95 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.49 (s, 1H), 7.38 (d, JAA′=8.5 Hz, 2H), 7.31 (d, JBB′=8.5 Hz, 2H), 4.26-4.24 (m, 1H), 4.03 (s, 3H), 3.76-3.73 (m, 2H), 3.67-3.66 (m, 1H), 3.58 (dd, J=14.9 Hz, J=7.9 Hz, 1H), 3.54-3.51 (m, 1H), 3.40-3.38 (m, 2H), 3.27-3.24 (m, 1H), 3.17 (dd, J=13.7 Hz, J=11.0 Hz, 1H), 3.09 (dd, J=13.8 Hz, J=4.1 Hz, 1H), 3.02 (s, 3H), 2.69-2.65 (m, 1H), 2.27-2.25 (m, 2H), 2.14-2.10 (m, 2H), 1.57-1.46 (m, 4H).


Example 116
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(2-methyl-2H-tetrazol-5-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine 2,2,2-trifluoroacetate (116)



embedded image


The title compound (116) was obtained as a TFA salt as a single isomer in 6% overall yield in a similar way to Example 15 with the exception that, in the first step of the synthesis, 5-bromo-2-methyl-2H-tetrazole was used instead of 3-bromo-1,5-dimethyl-1H-1,2,4-triazole. The crude product was purified by flash column chromatography on silica (pentane/AcOEt, 5:1 to 0:100, v/v) and then by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C21H31ClN5O3S found 468.2/470.2 [M+H]+; Rt=0.90 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38 (d, JAA′=8.5 Hz, 2H), 7.32 (d, JBB′=8.5 Hz, 2H), 4.27-4.24 (m, 4H), 3.77-3.73 (m, 2H), 3.68-3.66 (m, 1H), 3.58 (dd, J=15.0 Hz, J=7.9 Hz, 1H), 3.54-3.51 (m, 1H), 3.47-3.44 (m, 1H), 3.40-3.38 (m, 1H), 3.28-3.24 (m, 1H), 3.17 (dd, J=13.7 Hz, J=10.9 Hz, 1H), 3.10 (dd, J=13.6 Hz, J=4.0 Hz, 1H), 3.03 (s, 3H), 2.91 (tt J=12.0 Hz, J=3.6 Hz, 1H), 2.30-2.28 (m, 2H), 2.22-2.19 (m, 2H), 1.67-1.54 (m, 4H).


Example 117
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-N,N-dimethyl-4-(4-(5-methyl-1,3,4-thiadiazol-2-yl)cyclohexyl)morpholine-2-carboxamide 2,2,2-trifluoroacetate (117)



embedded image


The title compound (117) was obtained as a TFA salt as a single isomer in 18% overall yield in a similar way to Example 15 with the exception that, in the first step of the synthesis, 2-bromo-5-methyl-1,3,4-thiadiazole was used instead of 3-bromo-1,5-dimethyl-1H-1,2,4-triazole and in the fourth step of the synthesis, the compound 117b (the synthesis of this compound was described below) was used instead of the compound 3h. The crude product was purified by silica-gel column chromatography (pentane/AcOEt, 3:1 to 0:100, v/v; then: AcOEt/MeOH, 140:1 to 10:1, v/v) and then by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 30:70, 30 min, 20 mL/min).


ESI-MS m/z for C23H32ClN4O2S found 463.2/465.2 [M+H]+; Rt=0.95 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.37 (d, JAA′=8.4 Hz, 2H), 7.31 (d, JBB′=8.4 Hz, 2H), 4.74-4.73 (m, 1H), 3.83-3.79 (m, 2H), 3.65-3.60 (m, 2H), 3.54-3.52 (m, 1H), 3.41-3.39 (m, 1H), 3.22-3.19 (m, 1H), 3.11 (tt, J=11.8 Hz, J=3.4 Hz, 1H), 3.04-3.01 (m, 4H), 2.89 (brs, 3H), 2.66 (3H), 2.26-2.17 (m, 4H), 1.72-1.60 (m, 3H), 1.58-1.52 (m, 1H).


Synthesis of the Compound 117b
Step 1.
Synthesis of tert-butyl (2R,5S)-5-(4-chlorobenzyl)-2-(dimethylcarbamoyl)morpholine-4-carboxylate (117a)



embedded image


The title compound (117a) was obtained from 5a (480 mg; 1.35 mmol) according to the General Procedure V in 97% yield (500 mg; 1.31 mmol).


ESI-MS m/z for C19H28ClN2O4 found 383.2/385.2 [M+H]+; Rt=1.47 min (Method A)


Step 2.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-N,N-dimethylmorpholine-2-carboxamide hydrochloride (117b)



embedded image


The title compound (117b) was obtained as a hydrochloride salt from 117a (180 mg; 0.47 mmol) according to the General Procedure IVa in 99% yield (150 mg; 0.47 mmol).


ESI-MS m/z for C14H20ClN2O2 found 283.1/285.1 [M+H]+; Rt=0.67 min (Method A)


Example 118
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyl-1,3,4-thiadiazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (118)



embedded image


The title compound (118) was obtained as a hydrochloride salt as a single isomer in 18% overall yield in a similar way to Example 15 with the exception that, in the first step of the synthesis, 2-bromo-5-methyl-1,3,4-thiadiazole was used instead of 3-bromo-1,5-dimethyl-1H-1,2,4-triazole. The crude product was purified by silica-gel column chromatography (pentane/AcOEt, 4:1 to 0:100, v/v; then: AcOEt/MeOH, 80:1 to 20:1, v/v) and then by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 90:10 to 20:80, 30 min, 20 mL/min). Then a white solid of TFA salt (after lyophilization) was dissolved in water and to this solution 2 N HCl (2 mL) was added, then concentrated in vacuo and lyophilized from water to give the product as a hydrochloride salt.


ESI-MS m/z for C22H31ClN3O3S2 found 484.1/486.1 [M+H]+; Rt=0.91 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.37 (d, JAA′=8.4 Hz, 2H), 7.33 (d, JBB′=8.4 Hz, 2H), 4.38-4.36 (m, 1H), 3.90 (d, JBB′=13.2 Hz, 1H), 3.80-3.78 (m, 1H), 3.67 (d, JBB′=13.3 Hz, 1H), 3.60-3.52 (m, 3H), 3.42 (dd, J=15.1 Hz, J=3.4 Hz, 1H), 3.31-3.30 (m, 1H), 3.20 (dd, J=13.8 Hz, J=10.8 Hz, 1H), 3.16-3.10 (m, 2H), 3.03 (s, 3H), 2.66 (3H), 2.32-2.29 (m, 2H), 2.27-2.22 (m, 2H), 1.75-1.69 (m, 2H), 1.67-1.60 (m, 2H).


Example 119
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methoxy-1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine 2,2,2-trifluoroacetate (119)



embedded image


The title compound (119) was obtained as a TFA salt as a single isomer in 5% overall yield in a similar way to Example 15 with the exception that, in the first step of the synthesis, 3-bromo-5-methoxy-1-methyl-1H-1,2,4-triazole was used instead of 3-bromo-1,5-dimethyl-1H-1,2,4-triazole and Pd(dppf) was used instead of [PPh3]4Pd. The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 1:1 to 0:100, v/v; then: AcOEt/MeOH, 50:1 to 20:1, v/v) and then transferred into HCl salt with using HCl/DCM and then purified twice by preparative reversed-phase column chromatography (first: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min, second: column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C23H34ClN4O4S found 497.4/499.4 [M+H]+; Rt=0.94 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.37 (d, JAA′=8.4 Hz, 2H), 7.31 (d, JBB′=8.4 Hz, 2H), 4.28-4.24 (m, 1H), 3.97 (s, 3H), 3.78-3.73 (m, 2H), 3.68-3.66 (m, 1H), 3.58-3.55 (m, 1H), 3.52-3.51 (m, 1H), 3.45 (s, 3H), 3.42-3.38 (m, 2H), 3.27-3.24 (m, 2H), 3.17 (dd, J=13.8 Hz, J=10.7 Hz, 1H), 3.09 (dd, J=13.8 Hz, J=4.1 Hz, 1H), 3.02 (s, 3H), 2.25-2.24 (m, 2H), 2.11-2.08 (m, 2H), 1.57-1.48 (m, 4H).


Example 120
Synthesis of 3-(4-((2R,5S)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)morpholino)cyclohexyl)-1-methyl-1H-1,2,4-triazol-5-ol 2,2,2-trifluoroacetate (120)



embedded image


The title compound (120) was obtained during the synthesis of the compound 119 as a TFA salt as a single isomer as a by-product in 5% overall yield. The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 1:1 to 0:100, v/v; then: AcOEt/MeOH, 50:1 to 20:1, v/v) and then transferred into HCl salt with using HCl/DCM and then purified twice by preparative reversed-phase column chromatography (first: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80.30 min, 20 mL/min, second: column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C22H32ClN4O4S found 483.3/485.3 [M+H]+; Rt=0.80 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.36 (d, JAA′=8.4 Hz, 2H), 7.29 (d, JBB′=8.4 Hz, 2H), 4.25-4.21 (m, 1H), 3.74-3.72 (m, 1H), 3.68-3.64 (m, 2H), 3.57-3.53 (m, 1H), 3.46-3.44 (m, 1H), 3.34-3.31 (m, 2H), 3.23-3.20 (m, 4H), 3.15 (dd, J=13.6 Hz, J=10.8 Hz, 1H), 3.05 (dd, J=14.1 Hz, J=4.0 Hz, 1H), 3.01 (s, 3H), 2.47-2.44 (m, 1H), 2.23-2.21 (m, 2H), 2.09-2.06 (m, 2H), 1.51-1.45 (m, 4H)


Example 121
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-isopropyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine 2,2,2-trifluoroacetate (121)



embedded image


The title compound (121) was obtained as a TFA salt as a single isomer in 18% overall yield in a similar way to Example 15 with the exception that, in the first step of the synthesis, 3-bromo-1-isopropyl-1H-pyrazole was used instead of 3-bromo-1,5-dimethyl-1H-1,2,4-triazole and Pd(dppf) was used instead of [PPh3]4Pd. The crude product was purified by silica-gel column chromatography (cyclohexane/AcOEt, 12:1 to 0:100, v/v) and then transferred into TFA salt with using TFA/DCM and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 10:90, 30 min, 20 mL/min).


ESI-MS m/z for C25H37ClN3O3S found 494.5/496.5 [M+H]+; Rt=1.08 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.51 (d, J=2.2 Hz, 1H), 7.37 (d, JAA′=8.4 Hz, 2H), 7.31 (d, JBB′=8.4 Hz, 2H), 6.01 (d, J=2.2 Hz, 1H), 4.37 (hept, J=6.7 Hz, 1H), 4.30-4.26 (m, 1H), 3.79-3.77 (m, 2H), 3.69-3.68 (m, 1H), 3.59-3.55 (m, 2H), 2.47-2.41 (m, 2H), 2.30-2.27 (m, 1H), 3.19 (dd, J=13.8 Hz, J=10.7 Hz, 1H), 3.11 (dd, J=13.8 Hz, J=4.4 Hz, 1H), 3.03 (s, 3H), 2.59-2.54 (m, 1H), 2.27-2.25 (m, 2H), 2.10-2.07 (m, 2H), 1.57-1.44 (m, 4H), 1.36 (d, J=6.7 Hz, 6H).


Example 122
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methoxy-1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-N,N-dimethylmorpholine-2-carboxamide 2,2,2-trifluoroacetate (122)



embedded image


The title compound (122) was obtained as a TFA salt as a single isomer in 5% overall yield in a similar way to Example 15 with the exception that, in the first step of the synthesis, 3-bromo-5-methoxy-1-methyl-1H-1,2,4-triazole was used instead of 3-bromo-1,5-dimethyl-1H-1,2,4-triazole and in this reaction, Pd(dppf) was used instead of [PPh3]4Pd and in the fourth step of the synthesis, the compound 117b was used instead of the compound 3h. The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 1:1 to 0:100, v/v; then: AcOEt/MeOH, 40:1 to 5:1, v/v) and then purified twice by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 30:70, 30 min, 20 mL/min).


ESI-MS m/z for C24H35ClN5O3 found 476.2/478.2 [M+H]+; Rt=0.93 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.37 (d, JAA′=8.4 Hz, 2H), 7.30 (d, JBB′=8.4 Hz, 2H), 4.75-4.73 (m, 1H), 3.97 (s, 3H), 3.83-3.80 (m, 2H), 3.65-3.63 (m, 1H), 3.59-3.51 (m, 2H), 3.44-3.40 (m, 4H), 3.21-3.19 (m, 1H), 3.03-3.01 (m, 4H), 2.89 (brs, 3H), 2.50-2.48 (m, 1H), 2.22-2.20 (m, 1H), 2.14-2.05 (m, 3H), 1.63-1.51 (m, 3H), 1.48-1.42 (m, 1H).


Example 123
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-hydroxy-1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-N,N-dimethylmorpholine-2-carboxamide 2,2,2-trifluoroacetate (123)



embedded image


The title compound (123) was obtained during the synthesis of the compound 122 as a TFA salt as a single isomer as a by-product in 1% overall yield. The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 1:1 to 0:100, v/v; then: AcOEt/MeOH, 40:1 to 5:1, v/v) and then purified twice by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water/MeCN+1‰ TFA, 95:5 to 30:70, 30 min, 20 mL/min).


ESI-MS m/z for C23H33ClN5O3 found 462.2/464.2 [M+H]+; Rt=0.82 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.36 (d, JAA′=8.5 Hz, 2H), 7.28 (d, JBB′=8.5 Hz, 2H), 4.70-4.68 (m, 1H), 3.78 (dd, J=13.0 Hz, J=2.2 Hz, 1H), 3.73-3.72 (m, 1H), 3.63-3.61 (m, 1H), 3.48-3.46 (m, 2H), 3.37-3.35 (m, 1H), 3.21 (s, 3H), 3.16-3.14 (m, 1H), 3.03-2.99 (m, 4H), 2.88 (brs, 3H), 2.48-2.44 (m, 1H), 2.21-2.19 (m, 1H), 2.13-2.02 (m, 3H), 1.58-1.47 (m, 3H), 1.44-1.38 (m, 1H).


Example 124
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,2-dimethyl-1H-imidazol-4-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (124)



embedded image


The title compound (124) was obtained as a hydrochloride salt as a single isomer in 1% overall yield in a similar way to Example 15 with the exception that, in the first step of the synthesis, 4-bromo-1,2-dimethyl-1H-imidazole was used instead of 3-bromo-1,5-dimethyl-1H-1,2,4-triazole. The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 4:1 to 0:100, v/v, then: AcOEt/MeOH, 40:1 to 2:1, v/v) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 99:1 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C24H35ClN3O3S found 480.4/482.4 [M+H]+; Rt=0.58 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 7.37 (d, JAA′=8.4 Hz, 2H), 7.30 (d, JBB′=8.4 Hz, 2H), 7.17 (s, 1H), 4.34-4.30 (m, 1H), 3.86-3.84 (m, 1H), 3.71-3.69 (m, 1H), 3.66-3.64 (m, 4H), 3.57 (dd, J=15.0 Hz, J=7.7 Hz, 1H), 3.50-3.48 (m, 1H), 3.41-3.38 (m, 2H), 3.26-3.22 (m, 1H), 3.17 (dd, J=13.7 Hz, J=10.8 Hz, 1H), 3.09-3.07 (m, 1H), 3.02 (s, 3H), 2.63 (tt, J=12.3 Hz, J=3.5 Hz, 1H), 2.51 (s, 3H), 2.27-2.24 (m, 2H), 2.14-2.10 (m, 2H), 1.67-1.60 (m, 2H), 1.49-1.43 (m, 2H).


Example 125
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-5-(trifluoromethyl)-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (125)



embedded image


The title compound (125) was obtained as a hydrochloride salt as a single isomer in 2% overall yield in a similar way to Example 15 with the exception that, in the first step of the synthesis, 3-bromo-1-methyl-5-(trifluoromethyl)-1H-pyrazole was used instead of 3-bromo-1,5-dimethyl-1H-1,2,4-triazole and in this reaction, Pd(dppf) was used instead of [PPh3]4Pd. The crude product was purified by silica-gel column chromatography (hexane/AcOEt, 5:1 to 0:100, v/v) and then purified twice by preparative reversed-phase column chromatography (first: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 10:90, 30 min, 20 mL/min, second: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 30:70, 30 min, 20 mL/min).


ESI-MS m/z for C24H32ClF3N3O3S found 534.2/536.2 [M+H]+; Rt=1.20 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.37 (d, JAA′=8.5 Hz, 2H), 7.31 (d, JBB′=8.5 Hz, 2H), 6.63 (s, 1H), 4.26-4.24 (m, 1H), 3.86 (brd, J=0.7 Hz, 3H), 3.77-3.75 (m, 1H), 3.71-3.70 (m, 1H), 3.66-3.64 (m, 1H), 3.57 (dd, J=15.0 Hz, J=7.9 Hz, 1H), 3.51-3.49 (m, 1H), 3.40-3.37 (m, 2H), 3.23-3.20 (m, 1H), 3.16 (dd, J=13.6 Hz, J=11.0 Hz, 1H), 3.07 (dd, J=13.6 Hz, J=3.8 Hz, 1H), 3.02 (s, 3H), 2.62-2.58 (m, 1H), 2.25-2.23 (m, 2H), 2.10-2.06 (m, 2H), 1.56-1.44 (m, 4H); 19F NMR (250 MHz, DMSO-d6+D2O) δ −59.5 (s).


Example 126
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiophen-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (126)



embedded image


The title compound (126) was obtained as a hydrochloride salt as a single isomer in 3% overall yield in a similar way to Example 15 with the exception that, in the first step of the synthesis, 5-bromo-2,3-dimethylthiophene was used instead of 3-bromo-1,5-dimethyl-1H-1,2,4-triazole and in this reaction Pd(dppf) was used instead of [PPh3]4Pd. The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 45:55, v/v, 35 min) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 10:90, 30 min, 20 mL/min).


ESI-MS m/z for C25H35ClNO3S2 found 496.3/498.3 [M+H]+; Rt=1.43 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.37 (d, JAA′=8.5 Hz, 2H), 7.31 (d, JBB′=8.5 Hz, 2H), 6.50 (s, 1H), 4.29-4.27 (m, 1H), 3.81-3.79 (m, 1H), 3.74-3.72 (m, 1H), 3.66-3.64 (m, 1H), 3.57 (dd, J=15.0 Hz, J=7.9 Hz, 1H), 3.52-3.50 (m, 1H), 3.42-3.37 (m, 2H), 3.26-3.22 (m, 1H), 3.16 (dd, J=13.8 Hz, J=10.8 Hz, 1H), 3.09 (dd, J=13.8 Hz, J=4.2 Hz, 1H), 3.02 (s, 3H), 2.67 (tt, J=12.0 Hz, J=3.4 Hz, 1H), 2.24-2.22 (m, 2H), 2.20 (s, 3H), 2.09-2.04 (m, 2H), 1.99 (s, 3H), 1.59-1.53 (m, 2H), 1.46-1.40 (m, 2H).


Example 127
Synthesis of 4-(4-((2R,5S)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)morpholino)cyclohexyl)-1-methyl-1H-pyrrole-2-carbonitrile hydrochloride (127)



embedded image


Step 1.
Synthesis of 1-methyl-4-(1,4-dioxaspiro[4.5]dec-7-en-8-yl)-1H-pyrrole-2-carbonitrile (127a)



embedded image


The solution of 4,4,5,5-tetramethyl-2-(1,4-dioxaspiro[4.5]dec-7-en-8-yl)-1,3,2-dioxaborolane (420 mg; 1.58 mmol), 4-bromo-1-methyl-1H-pyrrole-2-carbonitrile (278 mg, 1.50 mmol), Pd(dppf) (122 mg; 0.15 mmol) in mixture of dioxane (8 mL) and 2 M aqueous solution of K2CO3 (2 mL) was heated at 100° C. for 2.5 hours under argon atmosphere. LC-MS showed complete consumption of the starting material. Then a whole was concentrated in vacuo and the residue was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 60:40, v/v, 35 min). Compound 127a was obtained in 62% yield (226 mg; 0.93 mmol).


ESI-MS m/z for C14H17N2O2 found 244.9 [M+H]+; Rt=1.25 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 7.18 (d, J=1.8 Hz, 1H), 7.00 (d, J=1.9 Hz, 1H), 5.84-5.79 (m, 1H), 3.88 (s, 4H), 3.68 (s, 3H), 2.39-2.34 (m, 2H), 2.27-2.21 (m, 2H), 1.78-1.68 (m, 2H).


Step 2.
Synthesis of 1-methyl-4-(1,4-dioxaspiro[4.5]decan-8-yl)-1H-pyrrole-2-carbonitrile (127b)



embedded image


To the solution of 127a (224 mg; 0.917 mmol) in THF (5 mL) 10% Pd/C (25 mol %; 24 mg, 0.23 mmol) was added and the reaction mixture was conducted under hydrogen atmosphere at room temperature for 4 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, Pd/C was filtered off through Celite and the filtrate was concentrated in vacuo. The crude product was used to the next step without additional purification. Compound 127b was obtained in 95% yield (215 mg; 0.873 mmol).


ESI-MS m/z for C14H19N2O2 found 247.0 [M+H]+; Rt=1.20 min (Method A)


Step 3.
Synthesis of 1-methyl-4-(4-oxocyclohexyl)-1H-pyrrole-2-carbonitrile (127c)



embedded image


The solution of 127b (215 mg; 0.873 mmol) and 1 M KHSO4 (15 mL) in MeCN (3 mL) was stirred at room temperature overnight. The reaction progress was monitored by LC-MS analysis. After analytical control indicated completion of the reaction, to this mixture 4 M NaOH was added to pH 12 and a whole was extracted with DCM (2×). The combined organic solutions were dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude ketone was used to the next step without additional purification. Compound 127c was obtained in 95% yield (168 mg; 0.831 mmol).


ESI-MS m/z for C12H15N2O found 203.1 [M+H]+; Rt=1.06 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 7.01 (d, J=1.8 Hz, 1H), 6.82 (d, J=1.9 Hz, 1H), 3.66 (s, 3H), 2.96-2.87 (m, 1H), 2.50-2.44 (m, 2H), 2.25-2.17 (m, 2H), 2.13-2.06 (m, 2H), 1.72-1.59 (m, 2H).


Step 4.
Synthesis of 4-(4-((2R,5S)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)morpholino)cyclohexyl)-1-methyl-1H-pyrrole-2-carbonitrile hydrochloride (127)



embedded image


The title compound (127) was obtained as a hydrochloride salt as a single isomer from 3h (102 mg; 0.30 mmol) and from ketone 127c (60 mg; 0.30 mmol) according to the General Procedure VII in 10% yield (17 mg; 0.03 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 35:65, v/v, 36 min, 11 mL/min) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min).


ESI-MS m/z for C25H33ClN3O3S found 490.4/492.4 [M+H]+; Rt=1.04 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.37 (d, JAA′=8.4 Hz, 2H), 7.30 (d, JBB′=8.4 Hz, 2H), 6.91 (d, J=1.8 Hz, 1H), 6.72 (d, J=1.8 Hz, 1H), 4.27-4.24 (m, 1H), 3.78-3.76 (m, 1H), 3.72-3.70 (m, 1H), 3.66-3.64 (m, 4H), 3.57 (dd, J=15.0 Hz, J=7.9 Hz, 1H), 3.51-3.49 (m, 1H), 3.41-3.35 (m, 2H), 3.24-3.20 (m, 1H), 3.18-3.14 (m, 1H), 3.08-3.06 (m, 1H), 3.02 (s, 3H), 2.42-2.38 (m, 1H), 2.22-2.20 (m, 2H), 2.03-1.99 (m, 2H), 1.58-1.54 (m, 2H), 1.39-1.32 (m, 2H).


Example 128
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-cyano-1-methyl-1H-pyrrol-3-yl)cyclohexyl)morpholine-2-carboxylic acid hydrochloride (128)



embedded image


The title compound (128) was obtained as a hydrochloride salt as a single isomer in 7% overall yield in a similar way to Example 69 with the exception that, in the first step of the synthesis, the ketone 127c and the compound 9b were used instead of the ketone 15c and the compound 65b and in the second step of the synthesis, 4 M NaOH was used instead of 1 M NaOH. The crude product was purified twice by preparative reversed-phase column chromatography (first: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 98:2 to 10:90, 30 min, 18 mL/min, second: C-18, water+0.3‰ HCl (36%)/MeCN, 98:2 to 10:90, 30 min, 18 mL/min).


ESI-MS m/z for C24H29ClN3O3 found 442.3/444.3 [M+H]+; Rt=1.09 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.37 (d, JAA′=8.4 Hz, 2H), 7.30 (d, JBB′=8.4 Hz, 2H), 6.90 (d, J=1.8 Hz, 1H), 6.71 (d, J=1.8 Hz, 1H), 4.42-4.40 (m, 1H), 3.72-3.66 (m, 5H), 3.63-3.62 (m, 1H), 3.42-3.31 (m, 3H), 3.08 (dd, J=14.0 Hz, J=4.5 Hz, 1H), 2.97 (dd, J=13.7 Hz, J=10.3 Hz, 1H), 2.39 (tt, J=12.1 Hz, J=3.6 Hz, 1H), 2.11-2.07 (m, 2H), 2.02-1.95 (m, 2H), 1.59-1.46 (m, 2H), 1.38-1.25 (m, 2H).


Example 129
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-ethyl-5-methyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (129)



embedded image


Step 1.
Synthesis of 1-ethyl-5-methyl-3-(1,4-dioxaspiro[4.5]dec-7-en-8-yl)-1H-pyrazole (129a)



embedded image


The solution of 4,4,5,5-tetramethyl-2-(1,4-dioxaspiro[4.5]dec-7-en-8-yl)-1,3,2-dioxaborolane (739 mg; 2.78 mmol), 3-bromo-1-ethyl-5-methyl-1H-pyrazole (500 mg, 2.64 mmol), Pd(dppf) (212 mg; 0.26 mmol) in mixture of dioxane (16 mL) and 2 M aqueous solution of K2CO3 (4 mL) was heated at 100° C. for 5 hours under argon atmosphere. LC-MS showed complete consumption of the starting material. Then a whole was concentrated in vacuo and the residue was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 25:75, v/v, 35 min). Compound 129a was obtained in 86% yield (565 mg; 2.28 mmol).


ESI-MS m/z for C14H21N2O2 found 249.3 [M+H]+; Rt=1.07 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 6.09 (s, 1H), 5.99-5.97 (m, 1H), 4.00-3.92 (m, 2H), 3.88 (s, 4H), 2.48-2.42 (m, 2H), 2.32-2.24 (m, 2H), 2.19 (s, 3H), 1.74-1.65 (m, 2H), 1.22 (t, J=7.2 Hz, 3H).


Step 2.
Synthesis of 1-ethyl-5-methyl-3-(1,4-dioxaspiro[4.5]decan-8-yl)-1H-pyrazole (129b)



embedded image


To the solution of 129a (560 mg; 2.25 mmol) in AcOEt (20 mL) 10% Pd/C (5 mol %; 14 mg, 0.13 mmol) was added and the reaction mixture was conducted under hydrogen atmosphere at room temperature for 4 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, Pd/C was filtered off through the Celite pad and the filtrate was concentrated in vacuo. The crude product was used to the next step without additional purification. Compound 129b was obtained in 99% yield (558 mg; 2.23 mmol).


ESI-MS m/z for C14H23N2O2 found 251.3 [M+H]+; Rt=0.99 min (Method A)


Step 3.
Synthesis of 4-(1-ethyl-5-methyl-1H-pyrazol-3-yl)cyclohexan-1-one (129c)



embedded image


The solution of 129b (558 mg; 2.23 mmol) in 1 N HCl (15 mL) was stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, to this mixture aqueous solution of NaOH (4 M) was added to pH 12 and then extracted with DCM (2×). The combined organic solutions were dried over anhydrous MgSO4, filtered and concentrated in vacuo. The crude product was used to the next step without additional purification. Compound 129c was obtained in 89% yield (409 mg; 1.98 mmol).


ESI-MS m/z for C12H19N2O found 207.1 [M+H]+; Rt=0.85 min (Method A)


Step 4.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-ethyl-5-methyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (129)



embedded image


The solution of free amine of compound 3h (after basic (0.5 M NaOH) extraction with DCM)(91 mg; 0.30 mmol), ketone 129c (65 mg; 0.31 mmol) in MeOH (2 mL) was stirred at room temperature for 30 minutes and then to this mixture ZnCl2 (49 mg; 0.36 mmol) was added and stirred at room temperature for 10 minutes. Then NaBH3CN (23 mg; 0.36 mmol) was added and this mixture was then stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the reaction mixture was diluted with 5% NaHCO3 and after 10 minutes a whole was concentrated in vacuo. The residue was suspended in DCM and washed with water. An organic phase was dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 0:100, v/v, then: AcOEt/MeOH, 100:0 to 80:20, v/v) and then by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 20 mL/min). The title compound 129 was obtained as a hydrochloride salt as a single isomer in 7% yield (13 mg; 0.02 mmol).


ESI-MS m/z for C25H37ClN3O3S found 494.4/496.4 [M+H]+; Rt=1.00 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 7.40 (d, JAA′=8.5 Hz, 2H), 7.32 (d, JBB′=8.5 Hz, 2H), 5.86 (s, 1H), 4.24-4.21 (m, 1H), 3.94 (q, J=7.2 Hz, 2H), 3.81-3.80 (m, 1H), 3.75-3.72 (m, 1H), 3.64-3.61 (m, 3H), 3.40-3.38 (m, 2H), 3.22-3.15 (m, 2H), 3.09-3.07 (m, 1H), 3.03 (s, 3H), 2.48 (tt, J=11.8 Hz, J=3.4 Hz, 1H), 2.27-2.23 (m, 2H), 2.18 (d, J=0.6 Hz, 3H), 2.03-2.01 (m, 2H), 1.53-1.38 (m, 4H), 1.22 (t, J=7.2 Hz, 2H).


Example 130
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-ethyl-5-methyl-1H-pyrazol-3-yl)cyclohexyl)-morpholine-2-carboxylic acid hydrochloride (130)



embedded image


Step 1.
Synthesis of methyl (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-ethyl-5-methyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylate (130a)



embedded image


The solution of 9b (136 mg; 0.50 mmol), ketone 129c (105 mg; 0.50 mmol) in MeOH (2.5 mL) was stirred at room temperature for 30 minutes and then to this mixture ZnCl2 (83 mg; 0.61 mmol) was added and stirred at room temperature for 10 minutes. Then NaBH3CN (38 mg; 0.61 mmol) was added and this mixture was then stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the reaction mixture was diluted with 5% NaHCO3 and after 10 minutes a whole was concentrated in vacuo. The residue was suspended in DCM and washed with water. An organic phase was dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 0:100, v/v, 35 min). The title compound 130a was obtained in 34% yield (78 mg; 0.17 mmol).


ESI-MS m/z for C25H35ClN3O3 found 460.4/462.4 [M+H]+; Rt=1.06 min (Method A)


Step 2.
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-ethyl-5-methyl-1H-pyrazol-3-yl)cyclohexyl)-morpholine-2-carboxylic acid hydrochloride (130)



embedded image


To a solution of 130a (78 mg; 0.17 mmol) in MeOH (10 mL) 4 M NaOH (0.4 mL) was added and the mixture was stirred at room temperature for 4.5 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, methanol was evaporated off and the residue was dissolved in MeCN/water and this mixture was acidified to pH 4 by addition of HClaq. The crude product was purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 98:2 to 10:90, 30 min, 18 mL/min). The title compound 130 was obtained as a hydrochloride salt as a single isomer in 29% yield (24 mg; 0.05 mmol).


ESI-MS m/z for C24H33ClN3O3 found 446.4/448.4 [M+H]+; Rt=1.03 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.37 (d, JAA′=8.5 Hz, 2H), 7.31 (d, JBB′=8.5 Hz, 2H), 5.79 (s, 1H), 4.47 (dd, J=8.9 Hz, J=3.5 Hz, 1H), 3.93 (q, J=7.2 Hz, 2H), 3.77-3.69 (m, 3H), 3.53-3.42 (m, 3H), 3.12 (dd, J=13.9 Hz, J=4.1 Hz, 1H), 3.02 (dd, J=13.9 Hz, J=10.2 Hz, 1H), 2.47 (tt, J=12.2 Hz, J=3.6 Hz, 1H), 2.17-2.12 (m, 5H), 2.05-1.99 (m, 2H), 1.63-1.51 (m, 2H), 1.46-1.33 (m, 2H), 1.23 (t, J=7.2 Hz, 2H).


Example 131
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-N-ethyl-4-(4-(1-ethyl-5-methyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamide hydrochloride (131)



embedded image


To the solution of 130 (15 mg; 0.031 mmol) and ethylamine hydrochloride (4 mg; 0.047 mmol) in DCM (1.5 mL) diisopropylethylamine (DIPEA; 27 μL; 0.155 mmol) was added and after 5 minutes HATU (15 mg; 0.040 mmol) was added in one portion and the reaction mixture was stirred at room temperature overnight. The reaction progress was monitored by TLC and LC-MS. When analyses indicated completion of the reaction, a whole was concentrated in vacuo. The crude product was purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 19 mL/min). The title compound 131 was obtained as a hydrochloride salt as a single isomer in 58% yield (9 mg; 0.018 mmol).


ESI-MS m/z for C26H38ClN4O2 found 473.5/475.5 [M+H]+; Rt=1.03 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.41-7.38 (m, 2H), 7.37-7.32 (m, 2H), 5.87-5.81 (m, 1H), 4.40-4.31 (m, 1H), 3.96 (q, J=7.2 Hz, 2H), 3.85-3.79 (m, 2H), 3.75-3.70 (m, 1H), 3.63-3.57 (m, 1H), 3.53-3.49 (m, 1H), 3.39-3.34 (m, 1H), 3.24-3.09 (m, 5H), 2.26-2.17 (m, 5H), 2.08-2.01 (m, 2H), 1.65-1.55 (m, 2H), 1.47-1.37 (m, 2H), 1.25 (t, J=7.2 Hz, 3H), 1.08 (t, J=7.2 Hz, 3H).


Example 132
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4-methyl-1H-pyrazol-1-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (132)



embedded image


The title compound (132) was obtained as a hydrochloride salt as a single isomer from 3h (220 mg; 0.648 mmol) and from 4-(4-methyl-1H-pyrazol-1-yl)cyclohexan-1-one (110 mg; 0.617 mmol) according to the General Procedure VII in 18% yield (55 mg; 0.110 mmol). The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 10:90, v/v, 45 min) and then purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 18 mL/min).


ESI-MS m/z for C23H33ClN3O3S found 466.4/468.4 [M+H]+; Rt=1.04 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.41 (s, 1H), 7.37 (d, JAA′=8.5 Hz, 2H), 7.31 (d, JBB′=8.5 Hz, 2H), 7.21 (s, 1H), 4.30-4.26 (m, 1H), 4.08 (tt, J=11.9 Hz, J=3.8 Hz, 1H), 3.80-3.78 (m, 1H), 3.76-3.74 (m, 1H), 3.66 (dd, J=13.3 Hz, J=1.1 Hz, 1H), 3.58 (dd, J=15.0 Hz, J=8.0 Hz, 1H), 3.54-3.52 (m, 1H), 3.51-3.47 (m, 1H), 3.40-3.38 (m, 1H), 3.30-3.26 (m, 1H), 3.18 (dd, J=13.7 Hz, J=10.9 Hz, 1H), 3.12-3.09 (m, 1H), 3.02 (s, 3H), 2.30-2.28 (m, 2H), 2.17-2.13 (m, 2H), 1.98 (s, 3H), 1.84-1.76 (m, 2H), 1.66-1.60 (m, 2H).


Example 133
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4-methyl-1H-pyrazol-1-yl)cyclohexyl)morpholine-2-carboxylic acid hydrochloride (133)



embedded image


The title compound (133) was obtained as a hydrochloride salt as a single isomer in 18% overall yield in a similar way to Example 69 with the exception that, in the first step of the synthesis, 4-(4-methyl-1H-pyrazol-1-yl)cyclohexan-1-one and the compound 9b were used instead of the ketone 15c and the compound 65b and, in the second step of the synthesis, 4 M NaOH was used instead of 1 M NaOH. The crude product was purified twice by preparative reversed-phase column chromatography (first: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 98:2 to 30:70, 30 min, 20 mL/min, second: C-18, water+0.3‰ HCl (36%)/MeCN, 98:2 to 30:70, 30 min, 20 mL/min).


ESI-MS m/z for C22H29ClN3O3 found 418.0/420.0 [M+H]+; Rt=1.16 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.40 (s, 1H), 7.37 (d, JAA′=8.4 Hz, 2H), 7.30 (d, JBB′=8.4 Hz, 2H), 7.20 (s, 1H), 4.41 (dd, J=8.5 Hz, J=3.8 Hz, 1H), 4.06 (tt, J=11.8 Hz, J=3.8 Hz, 1H), 3.71-3.66 (m, 2H), 3.63-3.61 (m, 1H), 3.43-3.36 (m, 3H), 3.10 (dd, J=13.9 Hz, J=4.4 Hz, 1H), 2.98 (dd, J=13.9 Hz, J=10.2 Hz, 1H), 2.19-2.09 (m, 4H), 1.98 (s, 3H), 1.82-1.70 (m, 2H), 1.68-1.56 (m, 2H).


Example 134
Synthesis of N-(39-((((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-morpholin-2-yl)methyl)sulfonyl)-37-oxo-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azanonatriacontyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide hydrochloride (134)



embedded image


The title compound (134) was obtained as a single isomer as a hydrochloride salt from 89 (30 mg; 0.051 mmol) according to the General Procedure XI in 53% yield (36 mg; 0.027 mmol). The crude product was purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 10:90, 30 min, 18 mL/min).


ESI-MS m/z for C60H100ClN6O17S3 found 1308.0/1310.0 [M+H]+; Rt=1.12 min (Method A)


Example 135
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-(1H-tetrazol-5-yl)morpholine-2-carboxamide hydrochloride (135)



embedded image


To the solution of 11 (56 mg; 0.102 mmol) in dry DMF (3 mL) N-methylmorpholine (50 μL; 0.459 mmol) and BOP (benzotriazol-1-yloxytris(dimethylamino)phosphonium hexafluorophosphate) (68 mg; 0.153 mmol) were added followed by 1H-tetrazol-5-amine monohydrate (26 mg; 0.255 mmol) and the reaction mixture was stirred at room temperature for 5 days. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the reaction mixture was concentrated in vacuo. The crude product was purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 98:2 to 30:70, 30 min, 18 mL/min, Rt=15.8-16.7 min). The title compound 135 was obtained as a single isomer as a hydrochloride sat in 31% yield (17 mg; 0.032 mmol).


ESI-MS m/z for C24H32ClN8O2 found 499.5/501.5 [M+H]+; Rt=0.99 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.41-7.37 (m, 2H), 7.37-7.34 (m, 2H), 5.82 (s, 1H), 4.72-4.65 (m, 1H), 3.90-3.84 (m, 1H), 3.83-3.75 (m, 2H), 3.71-3.65 (m, 1H), 3.60 (s, 3H), 3.56-3.48 (m, 2H), 3.17-3.08 (m, 2H), 2.48-2.44 (m, 1H), 2.26-2.19 (m, 2H), 2.17 (s, 3H), 2.08-1.98 (m, 2H), 1.66-1.54 (m, 2H), 1.49-1.38 (m, 2H).


Example 136
Synthesis of (2R,5S)-4-(4-(1H-tetrazol-5-yl)cyclohexyl)-5-(4-chlorobenzyl)-2-((methylsulfonyl)-methyl)morpholine hydrochloride (136)



embedded image


Step 1.
Synthesis of 4-((2R,5S)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)morpholino)cyclohexane-1-carbonitrile (136a)



embedded image


The solution of 3h (as a free amine—obtained after basic (0.5 M NaOH) extraction with DCM) (200 mg; 0.66 mmol), 4-oxocyclohexane-1-carbonitrile (90 mg; 0.73 mmol) in MeOH (3 mL) was stirred at room temperature for 30 minutes and then to this mixture ZnCl2 (100 mg; 0.73 mmol) was added and stirred at room temperature for 10 minutes. Then NaBH3CN (48 mg; 0.76 mmol) was added and this mixture was then stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the reaction mixture was diluted with 5% NaHCO3 and extracted with AcOEt. An organic phase was dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 0:100, v/v, 35 min). The title compound 136a was obtained in 41% yield (168 mg; 0.27 mmol).


ESI-MS m/z for C20H28ClN2O3S found 411.1/413.1 [M+H]+; Rt=1.75 min (Method F)


Step 2.
Synthesis of (2R,5S)-4-(4-(1H-tetrazol-5-yl)cyclohexyl)-5-(4-chlorobenzyl)-2-((methylsulfonyl)-methyl)morpholine hydrochloride (136)



embedded image


To a solution of 136a (111 mg; 0.271 mmol) in DMF (2 mL) NaN3 (71 mg; 1.100 mmol) and NH4Cl (60 mg; 1.100 mmol) were added and the mixture was heated at 120° C. overnight. Then another portions of NaN3 (71 mg; 1.100 mmol) and NH4Cl (60 mg; 1.100 mmol) were added and the mixture was heated at 120° C. for 2 days. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the reaction mixture was quenched with saturated aqueous NaHCO3 and extracted with AcOEt. An organic layer was then dried over anhydrous Na2SO4, filtered and concentrated in vacuo and the crude product was purified by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water+1‰ HCl/MeCN, 95:5 to 20:80, 40 min, 12 mL/min, Rt=23 min). The title compound 136 was obtained as a hydrochloride salt as a single diastereoisomer in 3% yield (4 mg; 0.008 mmol).


ESI-MS m/z for C20H29ClN5O3S found 454.1/456.1 [M+H]+; Rt=1.51 min (Method F); 1H NMR (400 MHz, Methanol-d4) δ 7.43-7.36 (m, 2H), 7.36-7.31 (m, 2H), 4.49-4.22 (m, 1H), 4.03-3.38 (m, 8H), 3.37-3.31 (m, 1H), 3.30-3.26 (m, 1H), 3.26-3.16 (m, 1H), 3.10 (s, 3H), 2.66-2.30 (m, 4H), 1.91-1.64 (m, 4H).


Example 137
Synthesis of (2R,5S)-4-(4-(1H-1,2,4-triazol-1-yl)cyclohexyl)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (137)



embedded image


Step 1.
Synthesis of 1-(1,4-dioxaspiro[4.5]decan-8-yl)-1H-1,2,4-triazole (137a)



embedded image


To the solution of 1,4-dioxaspiro[4.5]decan-8-yl 4-methylbenzenesulfonate (250 mg; 0.80 mmol) and 1H-1,2,4-triazole (78 mg, 1.12 mmol) in anhydrous DMF (2 mL) NaH (60% in oil; 45 mg; 1.12 mmol) was added and the reaction mixture was stirred at 75° C. for 24 hours under inert atmosphere. The reaction progress was monitored by LC-MS analysis of small aliquots of the crude reaction mixture. When analysis indicated completion of the reaction, water and NaHCO3 were added and this mixture was extracted with AcOEt. An organic phase was dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was used to the next step without additional purification.


ESI-MS m/z for C10H16N3O2 found 210.0 [M+H]+; Rt=1.00 min (Method F)


Step 2.
Synthesis of 4-(1H-1,2,4-triazol-1-yl)cyclohexan-1-one (137b)



embedded image


The solution of 137a (the crude product) in 1 M HCl/Et2O (3 mL/3 mL) was stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, to this mixture aqueous solution of NaOH (4 M) was added to pH 13 and then extracted with AcOEt (2×). The combined organic solutions were dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was used to the next step without additional purification.


ESI-MS m/z for C8H12N3O found 166.0 [M+H]+; Rt=0.50 min (Method F)


Step 3.
Synthesis of (2R,5S)-4-(4-(1H-1,2,4-triazol-1-yl)cyclohexyl)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (137)



embedded image


The solution of 3h (as a free amine—obtained after basic (0.5 M NaOH) extraction with DCM) (185 mg; 0.61 mmol), 137b (100 mg; 0.61 mmol) in MeOH (3 mL) was stirred at room temperature for 30 minutes and then to this mixture ZnCl2 (92 mg; 0.67 mmol) was added and stirred at room temperature for 10 minutes. Then NaBH3CN (42 mg; 0.67 mmol) was added and this mixture was then stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the reaction mixture was diluted with 5% NaHCO3 and extracted with AcOEt. An organic phase was dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by flash column chromatography on silica (AcOEt/MeOH, 100:0 to 70:30, v/v, 28 min, Rt=11.5 min) and then by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water+1‰ HCl/MeCN, 95:5 to 20:80, 40 min, 12 mL/min, Rt=22 min). The title compound 137 was obtained as a hydrochloride salt as a single isomer in 13% yield (40 mg; 0.08 mmol).


ESI-MS m/z for C21H30ClN4O3S found 453.1/455.1 [M+H]+; Rt=1.61 min (Method F); 1H NMR (400 MHz, Methanol-d4) δ 9.72 (s, 1H), 8.81 (s, 1H), 7.43-7.27 (m, 4H), 4.72-4.63 (m, 1H), 4.04-3.60 (m, 6H), 3.56-3.39 (m, 2H), 3.34 (s, 2H), 3.09 (s, 3H), 2.64-2.37 (m, 4H), 2.20-2.00 (m, 2H), 2.00-1.83 (m, 2H).


Example 138
Synthesis of (2R,5S)-4-(4-(1H-pyrazol-1-yl)cyclohexyl)-5-(4-chlorobenzyl)-2-((methylsulfonyl)-methyl)morpholine hydrochloride (138)



embedded image


Step 1.
Synthesis of 1-(1,4-dioxaspiro[4.5]decan-8-yl)-1H-pyrazole (138a)



embedded image


NaH (60% in mineral oil; 147 mg; 3.688 mmol) was added to DMF (1.6 mL) and stirred at room temperature for 5 minutes. Then to this suspension 1H-pyrazole (251 mg; 3.688 mmol) was added in several portions within 10 minutes and the mixture was stirred at room temperature for 1 hour. Then to this mixture 1,4-dioxaspiro[4.5]decan-8-yl 4-methylbenzenesulfonate (960 mg; 3.073 mmol) was added and the reaction mixture was vigorously stirred at 60° C. for 16 hours. The reaction progress was monitored by LC-MS and TLC analyses of small aliquots of the crude reaction mixture. When analyses indicated completion of the reaction, the reaction was cooling down to room temperature and then to this mixture water (30 mL) was added and then extracted with AcOEt (3×20 mL) and Et2O (20 mL). The combined organic solutions were washed with water (5 mL) and brine (5 mL), dried over anhydrous Na2SO4, and concentrated in vacuo. The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 50:50, v/v, 25 min). The title compound 138a was obtained as a colorless oil in 72% yield (459 mg; 2.205 mmol).


ESI-MS m/z for C11H17N2O2 found 209.0 [M+H]+; Rt=1.67 min (Method F)


Step 2.
Synthesis of 4-(1H-pyrazol-1-yl)cyclohexan-1-one (138b)



embedded image


To the solution of 138a (459 mg; 2.205 mmol) in THF (8.8 mL) an aqueous solution of HCl (6 M; 4.4 mL) was added and a whole was stirred at room temperature for 40 hours. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, THF was evaporated under reduced pressure and the residue was alkalized with saturated aqueous solution of K2CO3 and then extracted with AcOEt (3×20 mL). The combined organic solutions were dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was used to the next step without additional purification.


ESI-MS m/z for C9H13N2O found 165.0 [M+H]+; Rt=0.87 min (Method F)


Step 3.
Synthesis of (2R,5S)-4-(4-(1H-pyrazol-1-yl)cyclohexyl)-5-(4-chlorobenzyl)-2-((methylsulfonyl)-methyl)morpholine hydrochloride (138)



embedded image


The solution of 3h (as a free amine—obtained after basic (4 M NaOH) extraction with chloroform) (160 mg; 0.527 mmol), 138b (104 mg; 0.632 mmol) in MeOH (5.3 mL) was stirred at room temperature for 30 minutes and then to this mixture ZnCl2 (86 mg; 0.632 mmol) was added and stirred at room temperature for 10 minutes. Then NaBH3CN (40 mg; 0.632 mmol) was added and this mixture was then stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, to the reaction mixture water (10 mL) and an aqueous solution of NaOH (4 M; 15 mL) were added followed by MTBE (25 mL), and the mixture was vigorously stirred at room temperature for 1 hour. Then the phases were separated and an aqueous one was washed with MTBE (2×25 mL) and discarded. The combined organic solutions were washed with 2 M NaOH (5 mL) and brine (5 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 0:100, v/v, 25 min) and then by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water+1‰ HCl (36%)/MeCN, 90:10 to 20:80, 50 min, 12 mL/min, Rt=32 min). The title compound 138 was obtained as a hydrochloride salt as a single isomer in 18% yield (48 mg; 0.098 mmol).


ESI-MS m/z for C22H31ClN3O3S found 452.1/454.1 [M+H]+; Rt=1.89 min (Method F); 1H NMR (400 MHz, Methanol-d4) δ 7.90-7.86 (m, 1H), 7.74-7.71 (m, 1H), 7.42-7.31 (m, 4H), 6.45-6.42 (m, 1H), 4.48-4.34 (m, 2H), 3.96-3.59 (m, 6H), 3.51-3.33 (m, 2H), 3.27-3.17 (m, 1H), 3.10 (s, 3H), 2.59-2.48 (m, 2H), 2.39-2.32 (m, 2H), 2.11-1.94 (m, 2H), 1.93-1.79 (m, 2H), 1.40-1.25 (m, 1H).


Example 139
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(4-fluoro-1H-pyrazol-1-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine hydrochloride (139)



embedded image


The title compound (139) was obtained as a hydrochloride salt as a single isomer in 10% overall yield in a similar way to Example 138 with the exception that, in the first step of the synthesis, 4-fluoro-1H-pyrazole was used instead of 1H-pyrazole and in the last step of the synthesis, the crude product was purified by flash column chromatography on silica (hexane/AcOEt, 100:0 to 0:100, v/v, 25 min) and then by preparative reversed-phase column chromatography (Luna® 5 μm Phenyl-Hexyl, LC Column 250×21.2 mm, water+1‰ HCl (36%)/MeCN, 90:10 to 10:90, 40 min, 12 mL/min, Rt=25.9 min).


ESI-MS m/z for C22H30ClFN3O3S found 470.0/472.0 [M+H]+; Rt=2.09 min (Method F); 1H NMR (400 MHz, Methanol-d4) δ 7.71-7.67 (m, 1H), 7.42-7.32 (m, 5H), 4.49-4.39 (m, 1H), 4.26-4.14 (m, 1H), 3.96-3.53 (m, 5H), 3.51-3.37 (m, 2H), 3.37-3.31 (m, 1H), 3.30-3.28 (m, 1H), 3.25-3.17 (m, 1H), 3.10 (s, 3H), 2.58-2.43 (m, 2H), 2.35-2.23 (m, 2H), 2.04-1.91 (m, 2H), 1.91-1.75 (m, 2H).


Example 140
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-fluoro-1-methyl-1H-pyrazol-3-yl)cyclohexyl)-morpholine-2-carboxylic acid hydrochloride (140)



embedded image


The title compound (140) was obtained as a hydrochloride salt as a single isomer in 7% overall yield in a similar way to Example 69 with the exception that, in the first step of the synthesis, the ketone 4-(5-fluoro-1-methyl-1H-pyrazol-3-yl)cyclohexan-1-one (the synthesis of this ketone was the same as for the ketone 15c with the exception that, in the first step of the synthesis, 3-bromo-5-fluoro-1-methyl-1H-pyrazole and Pd(dppf) x DCM were used instead of 3-bromo-1,5-dimethyl-1H-1,2,4-triazole and [PPh3]4Pd) and the compound 9b were used instead of the ketone 15c and the compound 65b and in the last step of the synthesis, the crude product was purified by preparative reversed-phase column chromatography (column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 98:2 to 10:90, 30 min, 20 mL/min, Rt=14.65-15.66 min).


ESI-MS m/z for C22H28ClFN3O3 found 436.4/438.4 [M+H]+; Rt=1.03 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O, 348 K) δ 7.38-7.34 (m, 2H), 7.32-7.28 (m, 2H), 5.69 (d, J=5.6 Hz, 1H), 4.45-4.41 (m, 1H), 3.75-3.72 (m, 1H), 3.72-3.68 (m, 1H), 3.67-3.63 (m, 1H), 3.57 (s, 3H), 3.47-3.42 (m, 1H), 3.41-3.36 (m, 2H), 3.12-3.06 (m, 1H), 3.03-2.95 (m, 1H), 2.48-2.42 (m, 1H), 2.17-2.09 (m, 2H), 2.06-1.97 (m, 2H), 1.62-1.48 (m, 2H), 1.47-1.32 (m, 2H).


Example 141
Synthesis of N1-(1,31-bis(((2R,3R,4R,5R,6R)-3-acetamido-4,5-dihydroxy-6-(hydroxymethyl)-tetrahydro-2H-pyran-2-yl)oxy)-16-((3-((3-(5-(((2R,3R,4R,5R,6R)-3-acetamido-4,5-dihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl)oxy)pentanamido)propyl)amino)-3-oxopropoxy)methyl)-5,11,21,27-tetraoxo-14,18-dioxa-6,10,22,26-tetraazahentriacontan-16-yl)-N2-(3-(4-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamido)methyl)-1H-1,2,3-triazol-1-yl)propyl)dodecanediamide hydrochloride (141)



embedded image


Step 1.
Synthesis of N1-(1,31-bis(((2R,3R,4R,5R,6R)-3-acetamido-4,5-dihydroxy-6-(hydroxymethyl)-tetrahydro-2H-pyran-2-yl)oxy)-16-((3-((3-(5-(((2R,3R,4R,5R,6R)-3-acetamido-4,5-diacetoxy-6-(acetoxymethyl)tetrahydro-2H-pyran-2-yl)oxy)pentanamido)propyl)amino)-3-oxopropoxy)methyl)-5,11,21,27-tetraoxo-14,18-dioxa-6,10,22,26-tetraazahentriacontan-16-yl)-N12-(3-(4-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamido)methyl)-1H-1,2,3-triazol-1-yl)propyl)dodecanediamide hydrochloride (141a)



embedded image


To the solution of 45 (8 mg; 0.0160 mmol) and an azide (3-Azido-propylamino-C12-tris-β-GalNAc-PerAc)(20 mg; 0.0096 mmol) in MeOH (0.5 mL) CuSO4×5 H2O (1.3 mg; 0.0050 mmol) was added and after 10 minutes sodium L-ascorbate (2 mg; 0.0100 mmol) was added in one portion. Then the reaction mixture was stirred at room temperature over week. After analytical control (LC-MS) indicated completion of the reaction, the reaction mixture was concentrated in vacuo and the residue was dissolved in water/MeCN/HCly, filtered off and taken for preparative reversed-phase column chromatography (first: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 19 mL/min, second: C-18, water+0.3‰ HCl (36%)/MeCN, 95:5 to 20:80, 30 min, 19 mL/min). The title compound (141a) was obtained as a hydrochloride salt as a single isomer in 52% yield (13 mg; 0.0050 mmol).


ESI-MS m/z for C120H188ClN18O40 found 1279.4 [(M+2)/2]+; Rt=1.34 min (Method A)


Step 2.
Synthesis of N1-(1,31-bis(((2R,3R,4R,5R,6R)-3-acetamido-4,5-dihydroxy-6-(hydroxymethyl)-tetrahydro-2H-pyran-2-yl)oxy)-16-((3-((3-(5-(((2R,3R,4R,5R,6R)-3-acetamido-4,5-dihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl)oxy)pentanamido)propyl)amino)-3-oxopropoxy)methyl)-5,11,21,27-tetraoxo-14,18-dioxa-6,10,22,26-tetraazahentriacontan-16-yl)-N12-(3-(4-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamido)methyl)-1H-1,2,3-triazol-1-yl)propyl)dodecanediamide hydrochloride (141)



embedded image


To the cooled to 0° C. solution of 141a (13 mg; 0.0050 mmol) in the MeOH (0.9 mL) sodium methoxide (2.5 mg; 0.0460 mmol) was added in one portion. Then the cooling bath was removed and another portion of) sodium methoxide (3 mg; 0.0550 mmol) was added and stirred at room temperature for 1 hour. After analytical control (LC-MS) indicated completion of the reaction, to the reaction mixture Dowex 50WX8 was added until the solution was weakly acidic. The resin was filtered off and washed with MeOH. The filtrate was concentrated in vacuo and the pink residue was taken for preparative reversed-phase column chromatography (C-18, water+0.3‰ HCl (36%)/MeCN, 95:5 to 10:90, 30 min, 20 mL/min). The title compound (141) was obtained as a hydrochloride salt as a single isomer in 32% yield (3.5 mg; 0.0016 mmol).


ESI-MS m/z for C102H170ClN8O31 found 1090.2 [(M+2)/2]+; Rt=1.03 min (Method A)


Example 142
Synthesis of (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-(5-(2-(2,6-dioxopiperidin-3-yl)-3-oxoisoindolin-4-yl)pentyl)morpholine-2-carboxamide hydrochloride (142)



embedded image


To the solution of 11 (47 mg; 0.086 mmol) and Lenalidomide-C5-NH2 hydrochloride (37 mg; 0.100 mmol) in dry DMF/DCM (0.05 mL/3 mL) diisopropylethylamine (DIPEA; 70 μL; 0.400 mmol) was added and after 5 minutes HATU (46 mg; 0.120 mmol) was added and the reaction mixture was stirred at room temperature overnight. The reaction progress was monitored by LC-MS. When analysis indicated completion of the reaction, the reaction mixture was diluted with DCM, washed with 0.5 M HClaq and then concentrated in vacuo. The crude product was purified twice by preparative reversed-phase column chromatography (first: C-18, water+0.3‰ HCl (36%)/MeCN, 95:5 to 10:90, 30 min, 20 mL/min, second: column: Cosmosil Cholester 20×250 mm, water+0.3‰ HCl (36%)/MeCN, 95:5 to 10:90, 30 min, 19 mL/min, Rt=12.5-14.9 min). The obtained solid of the hydrochloride salt was then lyophilized. The title compound (142) was obtained as a hydrochloride salt as a single isomer in 37% yield (25 mg; 0.032 mmol).


ESI-MS m/z for C41H52ClN6O5 found 743.6/45.5 [M+H]+; Rt=1.35 min (Method A); 1H NMR (700 MHz, DMSO-d6+D2O) δ 7.56-7.55 (m, 1H), 7.45-7.44 (m, 2H), 7.40 (d, JAA′=8.3 Hz, 2H), 7.33 (d, JBB′=8.3 Hz, 2H), 5.86 (s, 1H), 5.05 (dd, J=13.3 Hz, J=5.1 Hz, 1H), 4.45 (d, J=17.1 Hz, 1H), 4.30 (d, J=17.1 Hz, 1H), 4.27-4.23 (m, 1H), 3.75-3.67 (m, 2H), 3.60 (s, 3H), 3.59-3.44 (m, 2H), 3.17-3.03 (m, 5H), 2.88-2.83 (m, 1H), 2.63-2.60 (m, 3H), 2.47-2.37 (m, 2H), 2.25-2.19 (m, 2H), 2.16 (s, 3H), 2.03-1.99 (m, 3H), 1.61-1.38 (m, 8H), 1.30-1.27 (m, 2H).

Claims
  • 1. A compound represented by structural formula (I):
  • 2. The compound according to claim 1, represented by structural formula (I):
  • 3. The compound according to claim 1, having the structural formula (Ia):
  • 4. The compound according to claim 1, wherein:X represents —CH(—R1h)—;Y represents —CH2—; andR1h represents methanesulfonyl.
  • 5. The compound according to claim 1, wherein:X represents —CH(—R1h)—;Y represents —N(—R1k)—.and R1k and R1h taken along with nitrogen and carbon atoms bearing them, respectively, represent a 6-membered heterocyclic ring of structural formula (IIb):
  • 6. The compound according to claim 1, wherein Y represents —O—.
  • 7. The compound according to claim 1, wherein R1d and R1h are in cis relationship.
  • 8. The compound according to claim 1, wherein the absolute stereochemistry at the carbon atom bearing R1d is S.
  • 9. The compound according to claim 1, having the structural formula Ib):
  • 10. The compound according to claim 1, wherein R10 is bromo or chloro.
  • 11. (canceled)
  • 12. The compound according to claim 1, wherein R3f is 5-methyloxazol-2-yl, 1,5-dimethyl-1H-pyrazol-3-yl, 1,5-dimethyl-1H-1,2,4-triazol-3-yl, 4,5-dimethyloxazol-2-yl, 5-fluoro-1-methyl-1H-pyrazol-3-yl, 4,5-dimethylthiazol-2-yl, or 5-methylthiazol-2-yl.
  • 13-18. (canceled)
  • 19. The compound according to claim 1, whereinR3a represents H;R3g represents H; andthe R3f substituent and the heterocyclic ring attached by its nitrogen ring member atom to the cyclohexane ring in 1,4 substitution pattern are in trans relationship.
  • 20. The compound according to claim 1, selected from the group consisting of the following compounds: (2S,5S)-5-(4-chlorobenzyl)-2-methyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine;(2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-methylmorpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-((1r,4S)-4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(3-methylisoxazol-5-yl)morpholine;(7S,9aR)-7-(4-chlorobenzyl)-8-(4-(5-methyloxazol-2-yl)cyclohexyl)octahydropyrazino[2,1-c][1,4]oxazine;(7S,9aR)-7-(4-chlorobenzyl)-8-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)octahydropyrazino[2,1-c][1,4]oxazine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;methyl (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylate;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(1-(2,2,2-trifluoroethyl)-1H-1,2,3-triazol-4-yl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-(1-(2,2,2-trifluoroethyl)-1H-1,2,3-triazol-4-yl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)morpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(fluoromethyl)morpholine;((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methanol;(2R,5S)-5-(4-chlorobenzyl)-N-ethyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N,N-dimethylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-N,N-dimethyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(5-methyl-1,3,4-oxadiazol-2-yl)morpholine;(2R,5S)—N′-acetyl-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carbohydrazide;(2R,5S)-5-(4-chlorobenzyl)-2-(5-methyl-1,3,4-oxadiazol-2-yl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine;(2R,5S)—N′-acetyl-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carbohydrazide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1,5-dimethyl-1H-pyrazole-3-carboxamide;(2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-ethynylmorpholine;N-(35-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxapentatriacontyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-isopropylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-methylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-N,N-dimethylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-N-isopropylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-N-methyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-fluoro-1-methyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)-4-(4-(5-methylthiazol-2-yl)cyclohexyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((prop-2-yn-1-yloxy)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;N-(35-(4-((((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methoxy)methyl)-1H-1,2,3-triazol-1-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxapentatriacontyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide;(2R,5S)-2-(azidomethyl)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-(prop-2-yn-1-yl)morpholine-2-carboxamide;N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1H-pyrazole-3-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-((1-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)-1H-1,2,3-triazol-4-yl)methyl)morpholine-2-carboxamide;N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-2H-1,2,3-triazole-4-carboxamide;3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)imidazolidine-2,4-dione;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1H-imidazole-4-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohex-3-en-1-yl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohex-3-en-1-yl)-N-ethylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)morpholine-2-carboxamide;(2R,5S)-2-(azidomethyl)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-((1-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)-1H-1,2,3-triazol-4-yl)methyl)morpholine-2-carboxamide;N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methyl)-1H-1,2,3-triazole-5-carboxamide;N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1-methyl-1H-imidazole-4-carboxamide;methyl 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)acetate;2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)acetic acid;2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)-N-ethylacetamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;N-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)methanesulfonamide;(2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-bromobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-bromobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;N-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1,1,1-trifluoromethanesulfonamide;(2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;(2R,5S)—N-(2-(3-(but-3-yn-1-yl)-3H-diazirin-3-yl)ethyl)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)morpholine-2-carboxamide;2-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methoxy)acetic acid;2-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)propan-2-ol;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(5-methyl-1H-pyrazol-3-yl)morpholine;(2R,5R)-2-(4-chlorobenzyl)-1-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-5-(methylsulfonyl)piperidine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamide;2-(4-((2R,5R)-2-(4-chlorobenzyl)-5-(methylsulfonyl)piperidin-1-yl)cyclohexyl)-5-methyloxazole;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfinyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylthio)methyl)morpholine;1-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)pyrrolidine-2,5-dione;(2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-2-ethynylmorpholine;2-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)acetic acid;3-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)propanoic acid;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohex-3-en-1-yl)-2-((methylsulfonyl)methyl)morpholine;N-(35-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxapentatriacontyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide;2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)acetic acid;3-((((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methyl)sulfonyl)propanoic acid;3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2,4-dione;3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2,4-dione;3-((((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)amino)-4-hydroxycyclobut-3-ene-1,2-dione;3-amino-4-((((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)amino)cyclobut-3-ene-1,2-dione;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-2-((ethylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((ethylsulfonyl)methyl)morpholine;3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1-methylimidazolidine-2,4-dione;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-(1-methyl-1H-imidazol-4-yl)morpholine-2-carboxamide;4-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)butanoic acid;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-hydroxymorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-N-(1,5-dimethyl-1H-pyrazol-3-yl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carboxamide;5-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)pentanoic acid;(2R,5S)-5-(4-chlorobenzyl)-N-(1,5-dimethyl-1H-pyrazol-3-yl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamide;N-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carbonyl)-N-methylglycine;(2R,5S)-4-(4-(1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)-5-(4-(trifluoromethyl)benzyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-imidazol-4-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-4-(4-(1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-5-(4-(trifluoromethyl)benzyl)morpholine-2-carboxylic acid;(2S,5S)-5-(4-chlorobenzyl)-2-methyl-4-(4-(5-methyl-1H-pyrazol-3-yl)cyclohexyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-1,2,3-triazol-4-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;methyl (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-1,2,3-triazol-4-yl)cyclohexyl)morpholine-2-carboxylate;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-1,2,3-triazol-4-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-4-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(2-methyl-2H-1,2,3-triazol-4-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(2-methyl-2H-tetrazol-5-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-N,N-dimethyl-4-(4-(5-methyl-1,3,4-thiadiazol-2-yl)cyclohexyl)morpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyl-1,3,4-thiadiazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methoxy-1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;3-(4-((2R,5S)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)morpholino)cyclohexyl)-1-methyl-1H-1,2,4-triazol-5-ol;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-isopropyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methoxy-1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-N,N-dimethylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-hydroxy-1-methyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-N,N-dimethylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,2-dimethyl-1H-imidazol-4-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-methyl-5-(trifluoromethyl)-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiophen-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;4-(4-((2R,5S)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)morpholino)cyclohexyl)-1-methyl-1H-pyrrole-2-carbonitrile;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-cyano-1-methyl-1H-pyrrol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-ethyl-5-methyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1-ethyl-5-methyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-chlorobenzyl)-N-ethyl-4-(4-(1-ethyl-5-methyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4-methyl-1H-pyrazol-1-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4-methyl-1H-pyrazol-1-yl)cyclohexyl)morpholine-2-carboxylic acid;N-(39-((((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methyl)sulfonyl)-37-oxo-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azanonatriacontyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-(1H-tetrazol-5-yl)morpholine-2-carboxamide;(2R,5S)-4-(4-(1H-tetrazol-5-yl)cyclohexyl)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)-morpholine;(2R,5S)-4-(4-(1H-1,2,4-triazol-1-yl)cyclohexyl)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)-morpholine;(2R,5S)-4-(4-(1H-pyrazol-1-yl)cyclohexyl)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)-morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4-fluoro-1H-pyrazol-1-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-fluoro-1-methyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;N1-(1,31-bis(((2R,3R,4R,5R,6R)-3-acetamido-4,5-dihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl)oxy)-16-((3-((3-(5-(((2R,3R,4R,5R,6R)-3-acetamido-4,5-dihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl)oxy)pentanamido)propyl)amino)-3-oxopropoxy)methyl)-5,11,21,27-tetraoxo-14,18-dioxa-6,10,22,26-tetraazahentriacontan-16-yl)-N2-(3-(4-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamido)methyl)-1H-1,2,3-triazol-1-yl)propyl)dodecanediamide; and(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-(5-(2-(2,6-dioxopiperidin-3-yl)-3-oxoisoindolin-4-yl)pentyl)morpholine-2-carboxamide;or is a tautomer, stereoisomer, pharmaceutically acceptable salt, solvate, or polymorph thereof.
  • 21. The compound according to claim 2, selected from the group consisting of the following compounds: (2S,5S)-5-(4-chlorobenzyl)-2-methyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine;(2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-methylmorpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-((1r,4S)-4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(3-methylisoxazol-5-yl)morpholine;(7S,9aR)-7-(4-chlorobenzyl)-8-(4-(5-methyloxazol-2-yl)cyclohexyl)octahydropyrazino[2,1-c][1,4]oxazine;(7S,9aR)-7-(4-chlorobenzyl)-8-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)octahydropyrazino[2,1-c][1,4]oxazine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;methyl (2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylate;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(1-(2,2,2-trifluoroethyl)-1H-1,2,3-triazol-4-yl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-(1-(2,2,2-trifluoroethyl)-1H-1,2,3-triazol-4-yl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)morpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(fluoromethyl)morpholine;((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methanol;(2R,5S)-5-(4-chlorobenzyl)-N-ethyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N,N-dimethylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-N,N-dimethyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(5-methyl-1,3,4-oxadiazol-2-yl)morpholine;(2R,5S)—N′-acetyl-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carbohydrazide;(2R,5S)-5-(4-chlorobenzyl)-2-(5-methyl-1,3,4-oxadiazol-2-yl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine;(2R,5S)—N′-acetyl-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carbohydrazide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1,5-dimethyl-1H-pyrazole-3-carboxamide;(2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-ethynylmorpholine;N-(35-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxapentatriacontyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-isopropylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-methylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-N,N-dimethylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-N-isopropylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-N-methyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-fluoro-1-methyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-2-((methylsulfonyl)methyl)-4-(4-(5-methylthiazol-2-yl)cyclohexyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((prop-2-yn-1-yloxy)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;N-(35-(4-((((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methoxy)methyl)-1H-1,2,3-triazol-1-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxapentatriacontyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide;(2R,5S)-2-(azidomethyl)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-(prop-2-yn-1-yl)morpholine-2-carboxamide;N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1H-pyrazole-3-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-((1-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)-1H-1,2,3-triazol-4-yl)methyl)morpholine-2-carboxamide;N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-2H-1,2,3-triazole-4-carboxamide;3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)imidazolidine-2,4-dione;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1H-imidazole-4-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohex-3-en-1-yl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohex-3-en-1-yl)-N-ethylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)morpholine-2-carboxamide;(2R,5S)-2-(azidomethyl)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-((1-(37-oxo-41-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxa-36-azahentetracontyl)-1H-1,2,3-triazol-4-yl)methyl)morpholine-2-carboxamide;N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methyl)-1H-1,2,3-triazole-5-carboxamide;N-(((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1-methyl-1H-imidazole-4-carboxamide;methyl 2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)acetate;2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)acetic acid;2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)-N-ethylacetamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;N-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)methanesulfonamide;(2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-bromobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-bromobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;N-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1,1,1-trifluoromethanesulfonamide;(2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-ethylmorpholine-2-carboxamide;(2R,5S)—N-(2-(3-(but-3-yn-1-yl)-3H-diazirin-3-yl)ethyl)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol- 3-yl)cyclohexyl)morpholine-2-carboxamide;2-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methoxy)acetic acid;2-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)propan-2-ol;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(5-methyl-1H-pyrazol-3-yl)morpholine;(2R,5R)-2-(4-chlorobenzyl)-1-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-5-(methylsulfonyl)piperidine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamide;2-(4-((2R,5R)-2-(4-chlorobenzyl)-5-(methylsulfonyl)piperidin-1-yl)cyclohexyl)-5-methyloxazole;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfinyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylthio)methyl)morpholine;1-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)pyrrolidine-2,5-dione;(2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-2-ethynylmorpholine;2-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)acetic acid;3-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)propanoic acid;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohex-3-en-1-yl)-2-((methylsulfonyl)methyl)morpholine;N-(35-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)-3,6,9,12,15,18,21,24,27,30,33-undecaoxapentatriacontyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide;2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)acetic acid;3-((((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methyl)sulfonyl)propanoic acid;3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2,4-dione;3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)methyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2,4-dione;3-((((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)amino)-4-hydroxycyclobut-3-ene-1,2-dione;3-amino-4-((((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)amino)cyclobut-3-ene-1,2-dione;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-2-((ethylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((ethylsulfonyl)methyl)morpholine;3-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)-1-methylimidazolidine-2,4-dione;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-(1-methyl-1H-imidazol-4-yl)morpholine-2-carboxamide;4-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)butanoic acid;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-N-hydroxymorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-N-(1,5-dimethyl-1H-pyrazol-3-yl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carboxamide;5-(4-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)-1H-1,2,3-triazol-1-yl)pentanoic acid;(2R,5S)-5-(4-chlorobenzyl)-N-(1,5-dimethyl-1H-pyrazol-3-yl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamide; andN-((2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carbonyl)-N-methylglycine;or a tautomer, stereoisomer, pharmaceutically acceptable salt, solvate, or polymorph thereof.
  • 22. The compound according to claim 2, selected from the group consisting of the following compounds: (2S,5S)-5-(4-chlorobenzyl)-2-methyl-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine;(2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-methylmorpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-((1r,4S)-4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(3-methylisoxazol-5-yl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-(fluoromethyl)morpholine;((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methanol;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N,N-dimethylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-N-methylmorpholine-2-carboxamide;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(5-fluoro-1-methyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohex-3-en-1-yl)-2-((methylsulfonyl)methyl)morpholine;2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)acetic acid;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;N-(((2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholin-2-yl)methyl)methanesulfonamide;(2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-bromobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5S)-5-(4-bromobenzyl)-4-(4-(5-methyloxazol-2-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)-2-((methylsulfonyl)methyl)morpholine;(2R,5S)-5-(4-bromobenzyl)-4-(4-(1,5-dimethyl-1H-1,2,4-triazol-3-yl)cyclohexyl)morpholine-2-carboxylic acid;(2R,5R)-2-(4-chlorobenzyl)-1-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-5-(methylsulfonyl)piperidine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)morpholine-2-carboxamide;2-(4-((2R,5R)-2-(4-chlorobenzyl)-5-(methylsulfonyl)piperidin-1-yl)cyclohexyl)-5-methyloxazole;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylsulfinyl)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((methylthio)methyl)morpholine;(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohex-3-en-1-yl)-2-((methylsulfonyl)methyl)morpholine;2-((2S,5S)-5-(4-chlorobenzyl)-4-(4-(4,5-dimethylthiazol-2-yl)cyclohexyl)morpholin-2-yl)acetic acid; and(2R,5S)-5-(4-chlorobenzyl)-4-(4-(1,5-dimethyl-1H-pyrazol-3-yl)cyclohexyl)-2-((ethylsulfonyl)methyl)morpholine;or a tautomer, stereoisomer, pharmaceutically acceptable salt, solvate, or polymorph thereof.
  • 23. A pharmaceutical composition comprising a therapeutically effective amount of at least one compound according to claim 1, and a pharmaceutically acceptable carrier therefor; optionally wherein the pharmaceutical composition further comprises a therapeutically effective amount of a therapeutic agent selected from the group consisting of steroids, membrane stabilizers, 5LO inhibitors, leukotriene synthesis and receptor inhibitors, inhibitors of IgE isotype switching or IgE synthesis, IgG isotype switching or IgG synthesis, β-agonists, tryptase inhibitors, acetylsalicylic acid, COX inhibitors, methotrexate, anti-TNF drugs, rituxin and other B-cell targeting agents, THF-targeting agents, PD4 inhibitors, p38 inhibitors, PDE4 inhibitors, and antihistamines.
  • 24. (canceled)
  • 25. A method for inhibiting the function of the chitinase-like protein YKL-40 or a method of degrading YKL-40 in a cell or a tissue, comprising contacting the cell or the tissue with at least one compound according to claim 1.
  • 26. A method for the treatment or prevention of a disease, disorder, or condition associated with aberrant expression or activity of the chitinase-like protein YKL-40, comprising administering to a subject in need thereof a therapeutically effective amount of at least one compound according to claim 1.
  • 27. The method of claim 26, wherein the disease, disorder, or condition is selected from the group consisting of cancer, allergic diseases, acute and chronic inflammatory diseases, autoimmune diseases, dental diseases, neurologic diseases, metabolic diseases, liver diseases, kidney diseases, polycystic ovary syndrome, endometriosis, asthma, and fibrotic disorders.
  • 28-38. (canceled)
  • 39. A method of identifying compounds that interfere with the chitinase-like protein YKL-40 and asialofetuin interaction by: (1) in a binding competition assay, subjecting (i) a compound according to claim 1 and (ii) biotinylated asialofetuin to YKL-40 or derivative of YKL-40 comprising a purification tag; and(2) assessing the resulting AlphaScreen luminescence signal; wherein an emission signal decline from the high level at lowest inhibitor concentration to the lower level (diminished by the value of at least 3× standard deviation of the noise displayed at plateau values at highest concentrations of inhibitor in the response plots for IC50 determinations) at highest inhibitor concentration indicates that the compound interferes with the chitinase-like protein YKL-40 and asialofetuin interaction.
Priority Claims (1)
Number Date Country Kind
P.441066 Apr 2022 PL national
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of priority to Polish Patent Application number P.441066, filed Apr. 29, 2022; and U.S. Provisional Patent Application No. 63/336,426, filed Apr. 29, 2022.

Divisions (1)
Number Date Country
Parent 63336426 Apr 2022 US
Child 18140705 US