![]() Hepatic X receptor (LXR) modulators for the treatment of diseases, disorders and skin conditions
专利摘要:
公开号:ES2691079T9 申请号:ES13754329T 申请日:2013-02-28 公开日:2019-02-08 发明作者:Raju Mohan 申请人:Ralexar Therapeutics Inc; IPC主号:
专利说明:
[0001] Hepatic X receptor (LXR) modulators for the treatment of diseases, disorders and skin conditions Cross-reference [0002] This application claims the benefit of the U.S. Provisional Application. No. 61 / 606,160 filed on March 2, 2012. [0003] BACKGROUND OF THE INVENTION [0004] Activation of the hepatic X receptor (LXR) is associated with inflammation, differentiation of the hyperproliferative and / or disordered cutaneous barrier. The activation of LXR also modulates multiple pathways underlying the etiology and pathology of skin aging. WO2010 / 096170, WO2007 / 002559 and FINN J et al., BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 13, no. 13, January 1, 2013, pages 2231-2234 may be useful for understanding the invention. [0005] Summary of the invention [0006] The present invention relates to compounds of Formula E, pharmaceutical compositions including such compounds, and methods of using same, to modulate the LXR. Also described herein are compounds of pharmaceutical compositions of Formula V that include such compounds, and methods of use thereof, for modulating the LXR. In one aspect, it is a compound of the invention for use in the treatment of diseases, disorders or dermal conditions in mammals that would benefit from the modulation of LXR. Preferred embodiments of the invention are described in the dependent claims. The associated methods are also described herein to aid understanding of the invention, but these are not part of the claimed invention. The examples or embodiments described herein that do not fall within the definition of the claims do not form part of the present invention. [0007] Diseases, disorders or skin conditions include skin aging, scarring, psoriasis, dermatitis, eczema, hives, rosacea, burns, acne or any other condition described here. Dermal diseases or disorders also refer to pigmentary disorders, including vitiligo. Dermal diseases also refer to skin malignancies and cancer, which include melanoma and the metastatic forms of these diseases. [0008] Accordingly, methods and compositions for the maintenance of the dermal barrier and / or the normalization of the dermal barrier and / or the reduction of the dermal barrier injury and / or the regeneration of the dermal barrier are described herein. [0009] According to the present invention, there is provided a compound having the structure of Formula (E): [0010] [0011] [0012] [0013] [0014] in which: [0015] A and B are each nitrogen, wherein A and B are linked together to form a five-membered heteroaryl ring; [0016] L 1 is a bond, alkyl C 1 -C 6 heteroalkyl or C1 - C6; [0017] L 2 is alkyl of C 1 -C 6 heteroalkyl or C1 - C6; [0018] R 1 is hydrogen, halogen, -CF 3 , -OR 8, -N (R 8 ) 2 , -C (= O) R 8, -C (= O) OR 8, -C (= O) N (R 8 ) 2 , -C (= N-OH) R8, -C (= S) N (R 8) 2, -C (= CH 2) CHa, or -C (= O) OCH2SCH3; [0019] R 2 is -C (= O) ORg, -C (= O) N (Rg) 2 , -NR 1 üC (= O) Rg, -C (= N-OH) Rg, -C (= S) N (Rg) 2 , or -C (= O) OCH 2 SCH 3 ; [0020] R 3 is hydrogen, halogen, C 1 -C 6 alkyl, or C 1 -C 6 haloalkyl; [0021] R 4 is aryl or heteroaryl; wherein aryl or heteroaryl is substituted with at least one R 11 ; [0022] each R 8, each Rg, and each R 10 are each independently hydrogen, alkyl of C 1 -C 6 heteroalkyl C 1 -C 6 -Ci-C6 alkyl-aryl, aryl, or heteroaryl; [0023] Rii is independently halogen, nitro, -OR 10 , -N (Rio) 2 , -CN, -C (= O) Rio, -C (= O) ORio, -C (= O) N (Rio) 2 , - NRioC (= O) Rio, NR 10 SO 2 R 10, -SOR 10, -SO 2 R 10, -SO 2 N (Ri 0) 2, -C (= O) OCH 2 SCH 3, alkyl of C 1 - C 6 , C 3 -C 8 cycloalkyl, Ci-Ca haloalkyl, Ci-Ca heteroalkyl, Ci-Ce-aryl alkyl, optionally substituted aryl or optionally substituted heteroaryl; [0024] or a pharmaceutically acceptable salt, a pharmaceutically acceptable solvate or a pharmaceutically acceptable prodrug thereof. [0025] In one embodiment, it is a compound of Formula E wherein R 4 is aryl. In a further embodiment, it is a compound of Formula E wherein R 2 is -C (= O) ORg, and R 9 is Ci-alkyl. Ca or Ci-Ca heteroalkyl. In a further embodiment, it is a compound of Formula E wherein L 2 is Ci-Ca alkyl. In a further embodiment, it is a compound of Formula E wherein L 2 is -CH 2 -. In a further embodiment, it is a compound of Formula E wherein Li is a bond. In a further embodiment is a compound of Formula E wherein Ri is -CF 3 , -C (= O) R 8, -C (= O) OR 8, -C (= O) N (R 8 ) 2 , or - C (= CH 2 ) CH 3 . In a further embodiment it is a compound of Formula E wherein R 4 is phenyl in which the phenyl is substituted with an R 11 . In a further embodiment, it is a compound of Formula E wherein R 11 is -SO 2 Rio and Rio is Ci-Ca alkyl. [0026] In another aspect, it is a pharmaceutical composition comprising a compound of Formula E, and a pharmaceutically acceptable diluent, excipient, carrier or binder thereof. In another aspect, it is a pharmaceutical composition comprising a compound of Formula V, and a pharmaceutically acceptable diluent, excipient, vehicle or binder thereof. [0027] Brief description of the figures [0028] Figure 1 shows the expression of the ABCG1 gene analyzed by real-time PCR for three compounds of Formula I-VI: Compound A, Compound B and Compound C as described in Example 29. [0029] Figure 2 shows the swelling of the ear and the weight of the ear for Compound A compared to Clobetasol (corticosteroid used to treat various skin disorders) as described in Example 41. Detailed description of the invention [0030] The LXR was first described by Willy, PJ, et al., "LXR, a nuclear receptor that defines a distinct retinoid response pathway," Genes & Development 9: io33-io45 (Cold Spring Harbor Laboratory Press). [0031] The hepatic X receptors (LXR alpha and LXR beta) are highly expressed in the epidermis and the activators of LXR stimulate the proliferation and differentiation of keratinocytes. The activation of LXR also improves the homeostasis of the permeability barrier by several mechanisms, which include stimulating the synthesis of epidermal lipids, increasing the formation and secretion of lamellar bodies and increasing the activity of enzymes required for the extracellular processing of lipids in the stratum. corneal, which leads to the formation of lamellar membranes that mediate the permeability barrier function. The activation of LXR is also anti-inflammatory, reducing inflammation in animal models of allergic dermatitis and irritant contact. (Schmuth et al., 2oo8, Journal of Lipid Research, 49, 499-5o9). [0032] The epidermis serves to form a barrier against the excessive loss of transcutaneous water to the environment. This barrier is formed by the anucleated, cornified and outermost layers of the epidermis, known collectively as the stratum corneum. The stratum corneum regulates a natural rate of water loss in the skin, a process called transepidermal water loss (or TEWL). Normal and healthy hydrated skin loses around 8o to 10o grams of water to the atmosphere each day. The TEWL process is affected by the integrity of the epidermal barrier and the lipid structure and, for a healthy skin, these elements regulate the rate of TEWL and help to maintain the appropriate levels of moisture in the stratum corneum. [0033] In this way, the maintenance of a normal epidermal barrier is a physiological means of inhibiting epidermal hyperproliferation. [0034] Examples of conditions that involve or give rise to an interrupted or dysfunctional epidermal barrier are: inflammation of the mucous membranes, such as cheilitis, chapped lips, nasal irritation and vulvovaginitis; eczematous dermatitis, such as atopic and seborrheic dermatitis, allergic or irritant contact dermatitis, craque eczema, photoallergic dermatitis, phototoxic dermatitis, phytophotodermatitis, radiation dermatitis and stasis dermatitis; ulcers and erosions resulting from trauma, burns, bullous disorders or ischemia of the skin or mucous membranes; various forms of ichthyosis; epidermolysis bullosa; psoriasis; hypertrophic and keloid scars and cutaneous changes of intrinsic aging and photo-aging. [0035] The constituents of the epidermis that play a role in the maintenance of a functional barrier are the sheets of intercellular lamellar bilayer of the lipids of the stratum corneum. The synthesis of lipids of the stratum corneum is relatively autonomous from circulating or dietary influences. The synthetic response is regulated instead by alterations in the permeability barrier functions. Regulation occurs through changes in the activities, the phosphorylation (activation) state, the mass and the mRNA of the enzymes limiting the speed of each of the three key lipids: serine palmitoyl transferase (for ceramides), HMGCoA reductase (for cholesterol) and both acetyl CoA carboxylase and fatty acid synthase (for fatty acids). Other results of alterations in the barrier function are the regulation of key enzymes of extracellular lipid processing. One of these enzymes is beta-glucocerebrosidase, which catalyzes the conversion of precursor glycosylceramides into ceramides. [0036] It has now been discovered that the formation of a mature fully differentiated stratum corneum and a functional epidermal functional barrier are accelerated by the topical administration of certain activators of the hepatic X receptor (LXR) with its two isoforms, LXR alpha and LXR beta. [0037] LXR activators improve barrier function by at least two parallel mechanisms: the stimulation of epidermal differentiation and the production of lipids. Given that the increase in the production of epidermal lipids probably generates additional endogenous activators of these nuclear hormone receptors, this process can be considered as a type of advance mechanism that regulates the generation of both the corneocytes and the extracellular matrix in a coordinated manner. of the stratum corneum. [0038] Hatano et al. have shown that topical application of LXR activators improves multiple parameters of AD-type dermatoses in a hapten-induced mouse model (Hatano et al (2010) The Journal of Allergy and Clinical Immunology 125 (1) 160-169. This model recapitulates virtually all known clinical, structural, functional, lipid biochemical and immunological abnormalities of human AD. [0039] The inherited abnormalities in important proteins for the barrier predispose to the development of atopic dermatitis (AD). On the contrary, the normalization of the barrier function, in turn, would reduce the two main inflammation factors in AD. Here methods are provided for reducing the generation of cytokines, which originate from, for example, disturbed corneocytes. In one embodiment, treatment with topical LXR activators reduces the levels of IL-1a and TNFa. In addition, the improved permeability barrier function simultaneously reduces the transdermal penetration of proinflammatory xenobiotes, including haptens and microbial pathogens. [0040] Chang et al. (Mol Endocrinol 2008, 22, 2407-2419) have demonstrated the efficacy of LXR ligands in normal human epidermal keratinocytes and in a mouse photoageing model. An integral molecular basis for efficacy in the mouse model was established by in vitro studies in normal human epidermal keratinocytes and in skin cell preparations of wild type LXR and LXR knock-out mice. In these studies, LXR activators: [0041] (a) reduced expression of cytokines and metalloproteinases in UV-activated epidermal keratinocytes and dermal fibroblasts activated by TNFa [0042] (b) increased the expression of keratinocyte differentiation markers [0043] (c) increased the expression of the genes required for the synthesis of fatty acids in keratinocytes (d) increased the expression of cholesterol binding proteins and lipid transporters in skin cells [0044] (e) increased the expression of enzymes involved in the synthesis of ceramide in keratinocytes. [0045] Lee et al. (J Invest Dermatol, 2012 Dec 6. doi: 10.1038 / jid.2012.409. [Electronic publication before printing]) have shown that in human primary melanocytes, MNT-1, and B16 melanoma cells, the activation of LXR and the LXR agonists have been shown to inhibit melanogenesis by inhibiting melanogenic enzymes through the degradation of MITF induced by Ras and ERK. This supports the logic that LXRs can be key target proteins in pigment disorders and that LXR agonists can be beneficial in the treatment of skin pigment disorders, including vitiligo. [0046] Pencheva et al. (Cell, 2012 Nov 21; 151 (5): 1068-82) have shown that skin-directed apolipoproteins such as ApoE converge to molecular targets such as LRP1 / LRP8 that are involved in metastasis of melanoma and melanoma. angiogenesis Because ApoE is a target gene for LXR, activation of LXR may be beneficial in the treatment of skin malignancies, including metastatic melanoma. [0047] Accordingly, methods and compositions comprising LXR activators as active ingredients in a formulation that is pharmaceutically acceptable for topical administration are described herein. [0048] The topical formulations containing LXR activators or activators described herein are applied for beneficial effect to the skin and / or mucous membranes. The activators are formulated in the form of lotions, solutions, gels, creams, emollient creams, ointments, aerosols or any other form that allows topical application. The formulation may also contain one or more agents that promote the extension of the formulation on the affected area, but which are otherwise biologically inactive. Examples of these agents are surfactants, humectants, wetting agents, emulsifiers or propellants. [0049] The amounts referred to herein as effective in enhancing barrier growth are any amount that will cause substantial relief of the symptoms of an altered or dysfunctional epidermal permeability barrier when repeatedly applied over time. The optimum amounts in any given case will be apparent to those skilled in the art or are capable of determination by routine experimentation. [0050] Examples of skin conditions that are amenable to topical treatment with LXR activators are: atopic and seborrheic dermatitis; inflammation of the mucous membranes, such as cheilitis, chapped lips, nasal irritation and vulvovaginitis; eczematous dermatitis caused by allergic and irritant contact, craquelee eczema, radiation dermatitis and stasis; ulcers and erosions due to chemical or thermal burns, bullous disorders or vascular compromise or ischemia that include venous, arterial, embolic or diabetic ulcers; ichthyosis, with or without an associated barrier anomaly; epidermolysis bullosa; psoriasis; hypertrophic and keloid scars; intrinsic aging, photoaging and / or dermatoheliosis; Melanoma and non-melanoma skin cancer, including lignin melanoma, basal cell carcinoma, squamous cell carcinoma, actinic keratosis and virus-induced neoplasia (warts and condylomata accuminata). [0051] The optimum methods and frequency of administration will be readily apparent to those skilled in the art or are capable of determination by routine experimentation. The effective results in most cases are achieved by the topical application of a thin layer on the affected area, or the area where you want to achieve the desired effect. Depending on the condition being treated, its stage or degree, and if the application is made for therapeutic or preventive reasons, effective results are obtained with application rates of one application every two or three days to four or more applications per day. [0052] The methods and compositions described herein are generally applicable to the treatment of mammalian skin including, for example, humans, domestic pets and livestock and other farm animals. [0053] Definitions [0054] In the context of this description, several terms will be used. [0055] As used herein, the term "about" or "about" means within 20%, preferably within 10%, and more preferably within 5% of a given value or range. [0056] The term "therapeutically effective amount" as used herein refers to the amount of an LXR modulator that, when administered to a mammal in need thereof, is effective to at least partially improve or at least partially prevent related conditions. with the aging of the skin. [0057] As used herein, the term "expression" includes the process by which the polynucleotides are transcribed into mRNA and translated into peptides, polypeptides or proteins. [0058] The term "modular" encompasses a decrease or an increase in activity or expression depending on the target molecule. For example, it is considered that a TIMP1 modulator modulates the expression of TIMP1 if the presence of said TIMP1 modulator results in an increase or decrease in the expression of TIMP1. The term "activator" is used in this specification to denote any molecular species that produces the activation of the indicated receptor, regardless of whether the species itself binds to the receptor or if a metabolite of the species binds to the receptor when the species is administered. topically Thus, the activator can be a receptor ligand or it can be an activator that is metabolized to the ligand of the receptor, i.e., a metabolite that is formed in the tissue and is the actual ligand. [0059] The terms "induce" or "induction" of the expression of TIMP1, ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1 or decorin refer to an increase, induction, or if not increase expression of TIMP1, ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1 or decorin and / or protein mRNA. The increase, induction or increase can be measured by one of the tests provided here. The induction of expression of TIMP1, ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1 or decorin does not necessarily indicate maximal expression of TIMP1, ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1 or decorin. An increase in the expression of TIMP1, ABCA12 or decorin can be, for example, at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more . In one embodiment, the induction is measured by comparing the mRNA expression levels of TIMP1, ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA12, ABCA13, ABCG1 or decorin of untreated keratinocytes with those of the mRNA expression levels of TIMP1, ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1 or decorin of keratinocytes treated with LXR modulator. [0060] The terms "inhibiting" the expression or "inhibition" of the expression of TNFa, MMP1, MMP3 or IL -8 refer to a reduction, inhibition or otherwise a decrease in the expression of protein and / or TNFa, MMP1, MMP3 or IL- 8 mRNA. The reduction, inhibition or reduction of the joint can be measured by one of the tests provided herein. The inhibition of the expression of TNFa, MMP1, MP3 or IL -8 does not necessarily indicate a complete negation of the expression of TNFa, MMP1, MMP3 or IL- 8 . A reduction in expression may be, for example, at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more. In one embodiment, the inhibition is measured by comparing the expression levels of TNFα, MMP1, MMP3 or IL -8 mRNA of untreated keratinocytes with the expression levels of TNFα, MMP1, MMP3 or IL -8 mRNA of keratinocytes treated with LXR modulator. "Hepatic X-Receptor" or "LXR" refers to both LXRa and LXRp, and variants, isoforms and active fragments thereof. LXRp is expressed ubiquitously, whereas expression of LXRa is limited to the liver, kidney, intestine, spleen, adipose tissue, macrophages, skeletal muscle and, as demonstrated here, to the skin. GenBank® accession numbers representative for the LXRa sequences include the following: human (Homo sapiens, Q 13133), mouse (Mus musculus, Q9Z0Y9), rat (Rattus norvegicus, Q62685), cow (Bos taurus, Q5E9B6) , pork (Sus scrofa, AAY43056), chicken (Gallus gallus, AAM90897). The representative GenBank® access numbers for LXRp include the following: human (Homo sapiens, P55055), mouse (Mus musculus, q60644), rat (Rattus norvegicus, Q62755), cow (Bos taurus, Q5BIS6). [0061] The term "mammal" refers to a human being, a non-human, canine, feline, bovine, ovine, porcine, murine or other veterinary or laboratory mammal. Those skilled in the art recognize that a therapy that reduces the severity of a pathology in a mammalian species is predictive of the effect of the therapy on another mammalian species. [0062] "Proinflammatory cytokine" as used herein refers to any cytokine that can activate cytotoxic, inflammatory or delayed hypersensitivity reactions. Examples of proinflammatory cytokines include colony stimulating factors (CSFs), for example granulocyte-macrophage CSF, granulocyte CSF, erythropoietin; Transforming growth factors (TGFs), for example, TGFp; interferons (IFNs), for example, IFNa, IFNp, IFNy; interleukins (IL), for example IL-1a, IL-ip, IL-3, IL- 6 , IL-7, IL- 8 , IL-9, IL-11, IL-12, IL-15; tumor necrosis factors (TNFs), for example TNFa, TNFp; adhesion proteins, for example intracellular adhesion molecule (ICAM), vascular cell adhesion molecule (VCAM); growth factors, for example leukemia inhibitory factor (LIF), factor that inhibits macrophage migration (MIF), epidermal growth factor (EGF), platelet derived growth factor (PDGF), fibroblast growth factor (FGF) ), insulin-like growth factor (IGF), nerve growth factor (NGF), B-cell growth factor (BCGF); chemokines, for example monocyte chemotactic proteins (MCP-1, MCP-2, MCP-3), macrophage inflammatory protein (MIP), growth-related oncogene, interferon-gamma-inducible protein; leukotrienes, for example, leukotriene B 4 , leukotriene D 4 ; vasoactive factors, for example, histamine, bradykinin, platelet activating factor (PAF); prostaglandins, for example prostaglandin E 2 . [0063] The expression "aging of the skin" includes conditions derived from intrinsic chronological aging (for example, deeper expression lines, reduction of skin thickness, inelasticity and / or smooth surface without defects), those derived from photoaging (for example, deep wrinkles, yellow and tanned surface, hardening of the skin, elastosis, roughness, depigmentation (age spots) and / or stained skin) and those deriving from skin thinning induced by steroids. [0064] Modulators of LXR [0065] The LXR modulators contemplated for use in the compositions and methods described herein are compounds with LXRa and / or LXRp modulating activities. The term "LXR modulator" includes LXRa and / or LXRp agonists, tissue-selective LXR antagonists and modulators, as well as other agents that induce the expression and / or protein levels of LXRs in skin cells. [0066] The preferred compounds will be the LXR modulators with LXRa and / or LXRp modulating activities. Preferred LXR modulators are LXR activators. The term "LXR activator" or "LXR activator" includes LXRa and / or LXRp agonists, partial agonists and tissue-selective LXR modulators, as well as other agents that induce the expression and / or protein levels of LXRs in the skin cells. [0067] According to the invention, it is a compound of Formula (E): [0068] [0069] [0070] in which: [0071] A and B are each nitrogen, wherein A and B are linked together to form a five-membered heteroaryl ring; [0072] L 1 is a bond, alkyl C 1 -C 6 heteroalkyl or C1 - C6; [0073] L 2 is alkyl of C 1 -C 6 heteroalkyl or C1 - C6; [0074] R 1 is hydrogen, halogen, -CF 3 , -OR 8, -N (R 8 ) 2 , -C (= O) R 8, -C (= O) OR 8, -C (= O) N (R 8 ) 2 , -C (= N-OH) R8, -C (= S) N (R 8) 2, -C (= CH 2) CHa, or -C (= O) OCH2SCH3; [0075] R 2 is -C (= O) ORg, -C (= O) N (Rg) 2 , -NR 1 üC (= O) Rg, -C (= N-OH) Rg, -C (= S) N (Rg) 2 , or -C (= O) OCH 2 SCH 3 ; [0076] R 3 is hydrogen, halogen, C 1 -C 6 alkyl, or C 1 -C 6 haloalkyl; [0077] R 4 is aryl or heteroaryl; wherein aryl or heteroaryl is substituted with at least one R 11 ; [0078] each R8, Rg each, and each R 1 Ü are each independently hydrogen, alkyl of C 1 -C 6 heteroalkyl C 1 -C 6 -alkyl of C1 - C6 alkylaryl, aryl, or heteroaryl; [0079] R 11 is independently halogen, nitro, -OR 1 Ü, -N (R 1 ü) 2 , -CN, -C (= O) R 1 ü, -C (= O) OR 10 , -C (= O) N (R 1 u) 2, -NR 1 Uc (= O) R 1 ü, NR 1 2 R 1 ü use, -SOR 1 ü, ü -SO 2 R 1, -SO 2 N (R 1 ii) 2 , -C (= O) OCH 2 SCH 3, alkyl of C 1 -C 6 cycloalkyl C 3 -C 8 alkyl, halo C 1 -C 6 heteroalkyl C 1 -C 6 -alkyl C 1 -Ca -aryl, optionally substituted aryl or optionally substituted heteroaryl; [0080] or a pharmaceutically acceptable salt, a pharmaceutically acceptable solvate or a pharmaceutically acceptable prodrug thereof. [0081] In another aspect, it is a compound of Formula (V): [0082] [0083] [0084] [0085] [0086] in which: [0087] L 1 is a bond, alkyl C 1 -C 6 heteroalkyl or C 1 -Ca; [0088] L 2 is alkyl of C 1 -C 6 heteroalkyl or C 1 -Ca; [0089] R 1 is hydrogen, halogen, -CF 3 , -OR 8, -N (R 8 ) 2 , -C (= O) R 8, -C (= O) OR 8, -C (= O) N (R 8 ) 2 , -C (= N-OH) R 8, -C (= S) N (R 8 ) 2 , -C (= CH 2 ) CH 3 , or -C (= O) OCH 2 SCH 3 ; [0090] R 2 is -C (= O) ORg, -C (= O) N (Rg) 2 , -NR 1 üC (= O) Rg, -C (= N-OH) Rg, -C (= S) N (Rg) 2 , or -C (= O) OCH 2 SCH 3 ; [0091] R 3 is hydrogen, halogen, C 1 -C 6 alkyl, or C 1 -C 6 haloalkyl; [0092] R 4 is aryl or heteroaryl; wherein the aryl or heteroaryl is substituted with at least one R 11 ; [0093] each R8, Rg each, and each R are each independently hydrogen, alkyl of C 1 -C 6 heteroalkyl C 1 -C 6 -alkyl of C1 - C6 alkylaryl, aryl, or heteroaryl; [0094] R 11 is independently halogen, nitro, -OR 1 Ü, -N (R 1 ü) 2 , -CN, -C (= O) R 1 ü, -C (= O) OR 1 ü, -C (= O ) N (R 1 u) 2, -NR 1 Uc (= O) R 1 ü, NR 1 2 R 1 ü use, -SOR 1 ü, ü -SO 2 R 1, -SO 2 N (R 1 ii) 2, -C (= O) OCH 2 SCH 3, alkyl of C 1 -C 6 cycloalkyl C 3 -C 8 alkyl, halo C 1 -C 6 heteroalkyl C 1 -C 6 -alkyl C 1 - C 6 -aryl, optionally substituted aryl or optionally substituted heteroaryl; [0095] In some embodiments, it is a compound of Formula V wherein R 1 is hydrogen, halogen, -CF 3 , -OR 8, -N (R 8 ) 2 , -C (= O) R 8, -C (= O) OR 8 , -C (= O) N (R 8 ) 2 , -C (= N-OH) R 8, -C (= S) N (R 8 ) 2 , -C (= CH 2) CH 3, or -C (= O) OCH2SCH3. In some embodiments, R 1 is hydrogen. In some embodiments, R 1 is halogen. In some embodiments, R 1 is -CF 3 . In some embodiments, R 1 is -OR 8. In some embodiments, R 1 is -N (R 8 ) 2 . In some embodiments, R 1 is -C (= O) R 8. In some embodiments, R 1 is -C (= O) O 8. In some embodiments, R 1 is -C (= O) N (R 8 ) 2 . In some embodiments, R 1 is -C (= N-OH) R 8. In some embodiments, R 1 is -C (= S) N (R 8 ) 2 . In additional embodiments, R8 It is hydrogen, alkyl of C 1 -C 6 heteroalkyl C 1 -C 6 -alkyl Ci-C6-aryl, aryl, or heteroaryl. In some embodiments, R8 is hydrogen. In some embodiments, R8 is C 1 -C 6 alkyl. In some embodiments, R8 is methyl. In some embodiments, R8 is ethyl In some embodiments, R8 is C 1 -C 6 heteroalkyl. In some embodiments, R 8 is-C 1 -C 6 alkyl aryl. In some embodiments, R8 is aryl. In some embodiments, R8 is heteroaryl. In some embodiments, R 1 is -C (= CH 2 ) CH 3 . In some embodiments, R 1 is -C (= O) OCH 2 SCH 3 . [0096] In some embodiments, it is a compound of Formula V wherein R 2 is -C (= O) ORg, -C (= O) N (Rg) 2 , -NR 1 üC (= O) Rg, -C (= N-OH) Rg, -C (= S) N (Rg) 2 , or -C (= O) OCH 2 SCH 3 . In some embodiments, R 2 is -C (= O) Og. In some embodiments, R 2 is -C (= O) N (Rg) 2 . In some embodiments, R 2 is -NR 1 or C (= O) Rg. In some embodiments, R 2 is -C (= N-OH) Rg. In some embodiments, R 2 is -C (= S) N (Rg) 2 . In some embodiments, R 2 is -C (= O) OCH 2 SCH 3 . In further embodiments, Rg is hydrogen, alkyl of C 1 -C 6 heteroalkyl C 1 -C 6 -alkyl of C1 - C6 alkylaryl, aryl, or heteroaryl. In some embodiments, Rg is hydrogen. In some embodiments, Rg is C 1 -C 6 alkyl. In some embodiments, Rg is methyl. In some embodiments, Rg is ethyl In some embodiments, Rg is C 1 -C 6 heteroalkyl. In some embodiments, Rg is -C 1 -C 6 alkyl aryl. In some embodiments, Rg is aryl. In some embodiments, Rg is heteroaryl [0097] [0098] In some embodiments, it is a compound of Formula V wherein L 1 is a bond, alkyl C 1 -C 6 heteroalkyl or C1 - C6. In some embodiments is a compound of Formula V in which L 2 is alkyl of C 1 -C 6 heteroalkyl or C1 - C6. In further embodiments, L 1 is a bond and L 2 is C 1 -C 6 alkyl. In further embodiments, L 1 is a bond and L 2 is C 1 -C 6 heteroalkyl. In further embodiments, L 1 and L 2 are each C 1 -C 6 alkyl. In further embodiments, L 1 is C 1 -C 6 alkyl and L 2 is C 1 -C 6 heteroalkyl. In further embodiments, L 1 and L 2 are each C 1 -C 6 heteroalkyl. In further embodiments, L 1 is heteroalkyl C1 - C6 alkyl and L 2 is C 1 -C 6. [0099] [0100] In some embodiments, it is a compound of Formula V wherein R 4 is aryl or heteroaryl; wherein the aryl or heteroaryl is substituted with at least one R 11 . In some embodiments, R 4 is aryl substituted with an R 11 . In some embodiments, R 4 is aryl substituted with two R 11 . In some embodiments, R 4 is aryl substituted with three R 11 . In further embodiments, R 4 is phenyl substituted with an R 11 . In further embodiments, R 4 is phenyl substituted with two R 11 . In further embodiments, R 4 is phenyl substituted with three R 11 . In some embodiments, R 4 is heteroaryl substituted with an R 11 . In some embodiments, R 4 is heteroaryl substituted with two R 11 . In some embodiments, R 4 is heteroaryl substituted with three R 11 . [0101] [0102] In some embodiments it is a compound of Formula V wherein R 4 is phenyl substituted with at least one R 11 , and each R 11 is independently -OR 10 , -N (R 1 o) 2 , -CN, -C ( = O) R 1 or, -C (= O) OR 1 or, -C (= O) N (R 1 o) 2 , -NR 1 oC (= O) R 1 o, NR 10 SO 2 R 10 , -SoR 1 or, -SO 2 R 1 or, -SO 2 N (R 1 o) 2 , -C (= O) OCH 2 SCH 3 , C 1 -C 6 alkyl, C 3 -C 8 cycloalkyl, haloalkyl of C 1 -C 6 heteroalkyl C 1 -C 6 -alkyl of C1 - C6 alkylaryl, optionally substituted aryl, or optionally substituted heteroaryl. In some embodiments it is a compound of Formula V wherein R 4 is heteroaryl substituted with at least one R 11 , and each R 11 is independently -OR 1 or, -N (R 1 or) 2 , -CN, -C (= O) R 1 or, -C (= O) OR 1 or, -C (= O) N (R 1 o) 2 , -NR 1 oC (= O) R 1 o, NR 1 o SO 2 R 1 or, -SOR 1 or, -SO 2 R 1 or, -SO 2 N (R 1 o) 2 , C 1 -C 6 alkyl, C 3 -C 8 cycloalkyl, C 1 -C 6 haloalkyl, heteroalkyl C 1 -C 6 -alkyl of C1 - C6 alkylaryl, optionally substituted aryl, or optionally substituted heteroaryl. In further embodiments, R 11 is -ORm In further embodiments, R 11 is -N (R 1 or) 2 . In further embodiments, R 11 is -CN. In further embodiments, R 11 is -C (= O) R 1 or. In further embodiments, R 11 is -C (= O) OR 1 or. In further embodiments, R 11 is -C (= O) N (R 1 o) 2 . In further embodiments, R 11 is -NR 1 or C (= O) R 1 or. In further embodiments, R 11 is NR 1 or SO 2 R 1 or. In further embodiments, R 11 is -SOR 1 or. In further embodiments, R 11 is -SO 2 R ™. In further embodiments, R 11 is -SO 2 N (R 1 o) 2 . In further embodiments, R 11 is -C (= O) OCH 2 SCH 3 . In further embodiments, R 11 is C 1 -C 6 alkyl. In further embodiments, R 11 is optionally substituted C 3 -C 8 cycloalkyl. In further embodiments, R 11 is C 1 -C 6 haloalkyl. In further embodiments, R 11 is C 1 -C 6 heteroalkyl. In further embodiments, R 11 is-C 1 -C 6 alkyl aryl. In further embodiments, R 11 is optionally substituted aryl. In further embodiments, R 11 is optionally substituted heteroaryl. In further embodiments, each R 1 is independently hydrogen or alkyl of C 1 -C 6 heteroalkyl C 1 -C 6 -alkyl of C1 - C6 alkylaryl, aryl, or heteroaryl. In some embodiments, R 1 or is hydrogen. In some embodiments, R 1 or is C 1 -C 6 alkyl. In some embodiments, R 1 or is C 1 -C 6 heteroalkyl. In some embodiments, R 1 or is -C 1 -C 6 alkyl aryl. In some embodiments, R 1 or is aryl. In some embodiments, R 1 or is heteroaryl. [0103] [0104] In another embodiment it is a compound of Formula V wherein R 1 is C (= O) OR 8, R 8 is C 1 -C 6 alkyl, and L 2 is C 1 -C 6 alkyl. In a further embodiment, R 2 is -C (= O) OCH 2 SCH 3 . In a further embodiment, R 2 is -C (= O) N (Rg) 2 . In a further embodiment, R 2 is -C (= O) ORg. In a further embodiment, L 1 is a link. In a further embodiment, L 1 is C 1 -C 6 alkyl. In a further embodiment, R 4 is phenyl substituted with an R 11 . In a further embodiment, R 11 is -SO 2 R 1 o and R 1 or is C 1 -C 6 alkyl. In a further embodiment, R 11 is -SO 2 R 1 o and R 1 or is CH 3. [0105] [0106] In another embodiment it is a compound of Formula V wherein L 1 is a bond, R 1 is -CF 3 , L 2 is C 1 -C 6 alkyl, R 2 is C (= O) ORg, and Rg is alkyl of C 1 -C 6 . In a further embodiment, R 4 is phenyl substituted with an R 11 . In a still further embodiment, R 11 is -SO 2 R 1 o and R 1 or is C 1 -C 6 alkyl. In a further embodiment, R 11 is -SO 2 R 1 o and R 1 or is CH 3 . [0107] In another embodiment it is a compound of Formula V in which Li is a bond, Ri is -CF 3 , L 2 is C 1 -C 6 alkyl, R 2 is C (= O) ORg, and R 9 is heteroalkyl C 1 -C 6 . In a further embodiment, R 4 is phenyl substituted with an R 11 . In a still further embodiment, R 11 is -SO 2 R 10 and R 10 is C 1 -C 6 alkyl. In a further embodiment, R 11 is -SO 2 R 10 and R 10 is CH 3 . [0108] [0109] In another embodiment it is a compound of Formula V wherein L 1 is a bond, R 1 is -CF 3 , L 2 is C 1 -C 6 alkyl, R 2 is -C (= O) OCH 2 SCH 3 . In a further embodiment, R 4 is phenyl substituted with an R 11 . In a still further embodiment, R 11 is -SO 2 R 10 and R 10 is C 1 -C 6 alkyl. In a further embodiment, R 11 is -SO 2 R 10 and R 10 is CH 3 . [0110] [0111] In another embodiment of the aforementioned embodiments, R 3 is hydrogen, halogen, alkyl of C 1 -C 6 haloalkyl or C1 - C6. In some embodiments of the aforementioned embodiments, R 3 is hydrogen. In some embodiments of the aforementioned embodiments, R 3 is halogen. In some embodiments of the aforementioned embodiments, R 3 is C 1 -C 6 alkyl. In some embodiments of the embodiments mentioned above, R 3 is C 1 -C 6 haloalkyl [0112] [0113] Any combination of the groups described above for the various variables is contemplated here. Throughout the specification, a person skilled in the art can choose groups and substituents thereof to provide stable moieties and compounds. [0114] [0115] In some embodiments, it is a compound selected from: [0116] [0117] [0118] or a pharmaceutically acceptable salt, a pharmaceutically acceptable solvate or a pharmaceutically acceptable prodrug thereof. [0119] In some embodiments, the therapeutic agent (s) (e.g., the compound of Formula V) is (are) present in the pharmaceutical composition in the form of a pharmaceutically acceptable salt. In some embodiments, any compound described above is suitable for any method or composition described herein. [0120] In certain embodiments, the compounds presented herein possess one or more stereocenters and each center exists independently in the R or S configuration. The compounds presented herein include all diastereomer, enantiomer and epimeric forms, as well as appropriate mixtures thereof. The stereoisomers are obtained, if desired, by methods such as stereoselective synthesis and / or the separation of stereoisomers by chiral chromatographic columns. In some embodiments, a compound of Formula V is used as a single enantiomer. In some embodiments, a compound of Formula V is used as a racemic mixture. [0121] The methods and formulations described herein include the use of N-oxides (if appropriate), crystalline forms (also known as polymorphs), or pharmaceutically acceptable salts of compounds having the structures presented herein, as well as active metabolites of these compounds having the same type of activity. In some situations, the compounds may exist as tautomers. All tautomers are included within the present description. The compounds described herein may exist in solvated forms with pharmaceutically acceptable solvents such as water, ethanol and the like. The compounds described herein may exist in unsolvated form. [0122] In some cases, the compounds of Formula V described herein include solvent addition forms or crystalline forms thereof, particularly solvates or polymorphs. The solvates contain stoichiometric or non-stoichiometric amounts of a solvent, and can be formed during the crystallization process with pharmaceutically acceptable solvents such as water, ethanol and the like. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. [0123] In some cases, the sites in the compounds of Formula IV described herein are susceptible to various metabolic reactions. Therefore, the incorporation of appropriate substituents at the sites of metabolic reactions will reduce, minimize or eliminate the metabolic pathways. In specific cases, the appropriate substituent to decrease or eliminate the susceptibility of the aromatic ring to metabolic reactions is, by way of example only, a halogen, deuterium or an alkyl group. [0124] In some cases, the compounds of Formula V described herein are isotopically labeled, which are identical to those listed in the various formulas and structures presented herein, except for the fact that one or more atoms are replaced by an atom having a mass atomic or mass number different from the atomic mass or mass number that is generally found in nature. In some examples, one or more hydrogen atoms are replaced by deuterium. In some examples, the metabolic sites in the compounds described herein are deuterated. In some examples, deuterium substitution provides certain therapeutic advantages resulting from increased metabolic stability, such as, for example, increased in vivo half-life or reduced dosage requirements. [0125] The compounds described herein, such as the compounds of Formula V, are in various forms, including, but not limited to, amorphous forms, ground forms and forms in nanoparticles. In addition, the compounds described herein include crystalline forms, also known as polymorphs. The polymorphs include the different crystal packing arrangements of the same elemental composition of a compound. Polymorphs usually have different X-ray diffraction patterns, melting points, density, hardness, crystalline shape, optical properties, stability and solubility. Various factors such as the recrystallization solvent, the crystallization rate and the storage temperature can cause a single crystalline form to dominate. [0126] The screening and characterization of pharmaceutically acceptable salts, polymorphs and / or solvates can be performed using a variety of techniques including, but not limited to, thermal analysis, X-ray diffraction, spectroscopy, vapor sorption and microscopy. Thermal analysis methods refer to thermochemical degradation or thermo-physical processes that include, but are not limited to, polymorphic transitions, and such methods are used to analyze the relationships between polymorphic forms, determine the weight loss, find the glass transition temperature or compatibility studies with excipients. Such methods include Differential Scanning Calorimetry (DSC), Differential Scanning Modulated Calorimetry (MDCS), Thermogravimetric Analysis (TGA) and Thermogravimetric and Infrared Analysis (TG / IR). X-ray diffraction methods include, but are not limited to, single crystal and powder diffractometers and synchrotron sources. The various spectroscopic techniques used include, but are not limited to, Raman, FTIR, UV-VIS and NMR (in liquid and solid state). The various microscopy techniques include, but are not limited to, polarized light microscopy, Scanning Electron Microscopy (SEM) with energy dispersive X-ray analysis (EDX), environmental scanning electron microscopy with EDX (in gaseous or water vapor), IR microscopy and Raman microscopy. [0127] Throughout the specification, the groups and their substituents may be chosen to provide stable moieties and compounds. [0128] Synthesis of compounds [0129] The synthesis of the compounds described herein is carried out using the means described in the chemical literature, using the methods described herein, or by a combination thereof. In addition, the solvents, temperatures and other reaction conditions presented herein may vary. [0130] The starting materials and reagents used for the synthesis of the compounds described herein are synthesized or obtained from commercial sources, such as, among others, Sigma-Aldrich, Fischer Scientific (Fischer Chemicals) and AcrosOrganics. [0131] The compounds described herein and other related compounds having different substituents are synthesized using techniques and materials described herein as well as those that are recognized in the art, as described, for example, in Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1- 17 (John Wiley and Sons, 1991); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplementals (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989), March, Advanced Organic Chemistry 4th Ed., (Wiley 1992); Carey and Sundberg, Advanced Organic Chemistry 4th Ed., Vols. A and B (Plenum 2000, 2001) and Green and Wuts, Protective Groups in Organic Synthesis 3rd ed., (Wiley 1999). General methods for the preparation of the compound as described herein can be derived from reactions and the reactions can be modified by the use of reagents and appropriate conditions, for the introduction of the various residues found in the formulas as provided herein. As a guide, the following synthetic methods can be used. [0132] Formation of covalent bonds by reaction of an electrophile with a nucleophile [0133] The compounds described herein can be modified using various electrophiles and / or nucleophiles to form new functional groups or substituents. Table IA entitled "Examples of covalent bonds and precursors thereof" lists selected non-limiting examples of covalent bonds and precursor functional groups that produce the covalent bonds. Table IA can be used as a guide for the variety of combinations of electrophiles and nucleophiles available that provide covalent linkages. The precursor functional groups are shown as electrophilic groups and nucleophilic groups. [0134] Table IA: Examples of covalent bonds and precursors thereof [0135] [0136] [0137] [0138] [0139] [0140] Use of protective groups [0141] In the reactions described, it may be necessary to protect reactive functional groups, for example hydroxy, amino, imino, thio or carboxy groups, when these are desired in the final product, in order to avoid their unwanted participation in the reactions. The protecting groups are used to block some or all of the reactive moieties and prevent said groups from participating in chemical reactions until the protecting group is removed. It is preferred that each protecting group be removable by a different means. Protective groups that are cleaved under totally different reaction conditions meet the requirement of differential withdrawal. [0142] The protecting groups can be removed by acid, base, reducing conditions (such as, for example, hydrogenolysis) and / or oxidizing conditions. Groups such as trityl, dimethoxytrityl, acetal and t-butyldimethylsilyl are acid labile and can be used to protect reactive carboxy and hydroxy moieties in the presence of amino groups protected with Cbz groups, which are removable by hydrogenolysis, and Fmoc groups, which they are labile in bases. Reagent moieties with carboxylic acid and hydroxy can be blocked with base leaving groups such as, but not limited to, methyl, ethyl and acetyl in the presence of blocked amines with acid-labile groups such as t-butyl carbamate or with carbamates which they are stable in both acid and base but hydrolytically removable. [0143] The carboxylic acid and the hydroxy reactive moieties can also be blocked with hydrolytically removable protective groups such as the benzyl group, while the amine groups capable of hydrogen bonding with acids can be blocked with base labile groups such as Fmoc. Reactive carboxylic acid moieties can be protected by conversion to simple ester compounds as exemplified herein, which include conversion to alkyl esters, or can be blocked with protective groups removable by oxidation such as 2,4-dimethoxybenzyl, while Coexisting amino groups can be blocked with fluoro-labile silyl carbamates. [0144] Allyl blocking groups are useful in the presence of acid and base protecting groups since the former are stable and can be subsequently removed by metal or pi acid catalysts. For example, a carboxylic acid blocked with allyl can be deprotected with a Pd0-catalyzed reaction in the presence of labile t-butyl carbamate in acid or base-labile amine acetate protecting groups. Yet another form of protecting group is a resin to which a compound or intermediate can be attached. As long as the residue binds to the resin, that functional group is blocked and can not react. Once released from the resin, the functional group is available to react. [0145] Typically, blocking / protection groups can be selected from: [0146] Other protective groups, plus a detailed description of the techniques applicable to the creation of protective groups and their removal are described in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons, New York, NY, 1999, and Kocienski, Protective Groups, Thieme Verlag, New York, NY, 1994. [0147] Certain terminology [0148] Unless otherwise defined, all technical and scientific terms used here have the same meaning as commonly understood to which the claimed matter belongs. In case there are a plurality of definitions for the terms here, those in this section prevail. When referring to a URL or other identifier or address of this type, it is understood that such identifiers may change and that particular information on the Internet may appear and disappear, but equivalent information can be found by searching the Internet. The reference to it shows the availability and public dissemination of such information. [0149] In this application, the use of the singular includes the plural unless specifically indicated otherwise. It should be noted that, as used in the specification and the appended claims, the singular forms "a", "an" and "the" include plural references unless the context clearly dictates otherwise. In this application, the use of "or" means "and / or" unless otherwise indicated. In addition, the use of the term "including", as well as other forms, such as "include", "include" and "included", is not limiting. [0150] The headings of the sections used here are for organizational purposes only and should not be construed as limiting the subject described. [0151] The definition of standard chemistry terms can be found in reference works, which include, but are not limited to, Carey and Sundberg's "Advanced Organic Chemistry 4th Ed.". Vols. A (2000) and B (2001), Plenum Press, New York. Unless otherwise indicated, conventional methods of mass spectroscopy, NMR, HPLC, protein chemistry, biochemistry, recombinant DNA techniques and pharmacology. [0152] Unless specific definitions are provided, the nomenclature used in relation to, and the laboratory procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medical and pharmaceutical chemistry described herein are those recognized in the field. Standard techniques can be used for chemical synthesis, chemical analysis, pharmaceutical preparation, formulation and administration, and patient treatment. Standard techniques can be used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection). The reactions and purification techniques can be performed, for example, using manufacturer's specification kits or as commonly performed in the art or as described herein. The above techniques and procedures can generally be performed from conventional methods and as described in various general and more specific references that are cited and discussed throughout the present specification. [0153] As used herein, Ci-Cx includes C 1 -C 2 , C 1 -C 3 ... Ci-Cx. Ci-Cx refers to the number of carbon atoms that make up the rest it designates (excluding optional substituents). [0154] An "alkyl" group refers to an aliphatic hydrocarbon group. The alkyl groups may or may not include units of unsaturation. The alkyl moiety can be a "saturated alkyl" group, which means that it does not contain any unit of unsaturation (ie, a carbon-carbon double bond or a carbon-carbon triple bond). The alkyl group may also be an "unsaturated alkyl" moiety, which means that it contains at least one unit of unsaturation. The alkyl moiety, whether saturated or unsaturated, can be branched, straight-chain or cyclic. [0155] The "alkyl" group can have from 1 to 6 carbon atoms (when it appears here, a numerical range such as "from 1 to 6" refers to each whole number in the given range, for example, "from 1 to 6 atoms. "carbon" means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 6 carbon atoms, although the present definition also includes the occurrence of the term "alkyl""in which a numerical range is not designated). The alkyl group of the compounds described herein can be designated as "C 1 -C 6 alkyl" or similar designations. By way of example only, "C 1 -C 6 alkyl" indicates that there are from one to six carbon atoms in the alkyl chain, ie, the alkyl chain is selected from the group consisting of methyl, ethyl, -propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, t-butyl, n-pentyl, iso-pentyl, neopentyl, hexyl, propen-3-yl (allyl), cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclohexylmethyl. The alkyl groups may be substituted or unsubstituted. Depending on the structure, an alkyl group may be a monoradical or a diradical (ie, an alkylene group). [0156] An "alkoxy" refers to a group "-O-alkyl", wherein alkyl is as defined herein. [0157] The term "alkenyl" refers to a type of alkyl group in which the first two atoms of the alkyl group form a double bond that is not part of an aromatic group. That is, an alkenyl group starts with the atoms -C (R) = CR 2 , where R refers to the remaining portions of the alkenyl group, which may be the same or different. Non-limiting examples of an alkenyl group include -CH = c H 2 , -C ( c H 3 ) = CH 2 , -CH = CHCH 3 , -CH = C (CH 3 ) 2 and -C (CH 3 ) = CHCH 3 The alkenyl moiety can be branched, straight chain or cyclic (in which case, it would also be known as the "cycloalkenyl" group). The alkenyl groups can have from 2 to 6 carbons. The alkenyl groups may be substituted or unsubstituted. Depending on the structure, an alkenyl group can be a monoradical or a diradical (ie, an alkenylene group). [0158] The term "alkynyl" refers to a type of alkyl group in which the first two atoms of the alkyl group form a triple bond. That is, an alkynyl group starts with the atoms -C e C r , where R refers to the remaining portions of the alkynyl group. Non-limiting examples of an alkynyl group include -C e CH, -CECCH 3 , -CECCH 2 CH 3 and -CECCH 2 CH 2 CH 3 . The "R" portion of the alkynyl moiety can be branched, straight chain or cyclic. An alkynyl group can have from 2 to 6 carbons. The alkynyl groups may be substituted or unsubstituted. Depending on the structure, an alkynyl group can be a monoradical or a diradical (i.e., an alkynylene group). [0159] "Amino" refers to a group -NH 2 group. [0160] The term "alkylamine" or "alkylamino" refers to the group -N (alkyl) xHy, wherein alkyl is as defined herein and x and y are selected from the group x = 1, y = 1 and x = 2, y = 0. When x = 2, the alkyl groups, taken together with the nitrogen to which they are attached, can optionally form a cyclic ring system. "Dialkylamino" refers to a group -N (alkyl) 2 , wherein alkyl is as defined herein. [0161] The term "aromatic" refers to a flat ring having a delocalized n electron system containing 4n 2 n electrons, where n is an integer. Aromatic rings can be formed from five, six, seven, eight, nine or more than nine atoms. The aromatics may be optionally substituted. The term "aromatic" includes both aryl groups (e.g., phenyl, naphthalenyl) and heteroaryl groups (e.g., pyridinyl, quinolinyl). [0162] As used herein, the term "aryl" refers to an aromatic ring in which each of the atoms forming the ring is a carbon atom. The aryl rings can be formed by five, six, seven, eight, nine or more than nine carbon atoms. The aryl groups may be optionally substituted. Examples of aryl groups include, but are not limited to, phenyl and naphthalenyl. Depending on the structure, an aryl group can be a monoradical or a diradical (i.e., an arylene group). [0163] "Carboxi" refers to -CO 2 H. In some cases, the carboxy moieties can be replaced with a "carboxylic acid bioisostere", which refers to a functional group or moiety that exhibits physical and / or chemical properties similar to the rest carboxylic acid A carboxylic acid bioisostere has similar biological properties to those of a carboxylic acid group A compound with a carboxylic acid moiety can have the carboxylic acid moiety exchanged with a carboxylic acid bioisostere and have similar physical and / or biological properties when it is compared to the carboxylic acid-containing compound, For example, a carboxylic acid bioisostere would ionize at physiological pH to approximately the same point as a carboxylic acid group Examples of bioisosteres of a carboxylic acid include, but are not limited to, [0164] [0165] [0166] [0167] The term "cycloalkyl" refers to a non-aromatic monocyclic or polycyclic radical, wherein each of the ring-forming atoms (ie, atoms of the main chain) is a carbon atom. Cycloalkyls can be saturated, or partially unsaturated. The cycloalkyls may be fused with an aromatic ring (in which case, the cycloalkyl is attached via a non-aromatic ring atom). Cycloalkyl groups include groups having from 3 to 10 ring atoms. Illustrative examples of cycloalkyl groups include, but are not limited to, the following moieties: [0168] [0169] [0170] [0171] and similar. [0172] The terms "heteroaryl" or, alternatively "heteroaromatic" refer to an aryl group that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur. A "heteroaromatic" or "heteroaryl" moiety containing N refers to an aromatic group in which at least one of the atoms of the ring skeleton is a nitrogen atom. The polycyclic heteroaryl groups can be fused or non-condensed. Illustrative examples of heteroaryl groups include the following moieties: [0173] [0174] [0175] [0176] and similar. [0177] A "heterocycloalkyl" group or "heteroalicyclic" group refers to a cycloalkyl group, wherein at least one atom of the ring skeleton is a heteroatom selected from nitrogen, oxygen and sulfur. The radicals can be condensed with an aryl or heteroaryl. Illustrative examples of heterocycloalkyl groups, also referred to as non-aromatic heterocycles, include: [0178] [0179] [0180] [0181] and similar. The term "heteroalicyclic" also includes all ring forms of carbohydrates, including, but not limited to monosaccharides, disaccharides and oligosaccharides. Unless otherwise mentioned, heterocycloalkyls have 2 to 10 carbons in the ring. It is understood that when referring to the number of carbon atoms in a heterocycloalkyl, the number of carbon atoms in the heterocycloalkyl is not the same as the total number of atoms (including the heteroatoms) that make up the heterocycloalkyl (i.e. skeleton of the heterocycloalkyl ring). [0182] The term "halo" or, alternatively, "halogen" means fluoro, chloro, bromo and iodo. [0183] The term "haloalkyl" refers to an alkyl group that is substituted with one or more halogens. The halogens can be the same or they can be different. Non-limiting examples of haloalkyls include -CH 2 Cl, -CF 3 , -CHF 2 , -CH 2 CF 3 , -CF 2 CF 3 , -CF (CH 3) 3, and the like. [0184] The terms "fluoroalkyl" and "fluoroalkoxy" include alkyl and alkoxy groups, respectively, which are substituted with one or more fluorine atoms. Non-limiting examples of fluoroalkyls include -CF 3 , -CHF 2 , -CH 2 F, -CH 2 CF 3 , -CF 2 CF 3 , -CF 2 CF 2 CF 3 , -CF (CH 3 ) 3 , and the like . Nonlimiting examples of fluoroalkoxy groups include -OCF 3, -OCHF 2, -OCH 2 F, -OCH 2 CF 3, -OCF 2 CF 3, -OCF 2 CF 2 CF 3, -OCF (CH3) 2, and the like . [0185] The term "heteroalkyl" refers to an alkyl radical in which one or more atoms of the skeleton chain is selected from an atom other than carbon, for example, oxygen, nitrogen, sulfur, phosphorus, silicon, or combinations thereof . The heteroatom (s) can be placed in any interior position of the heteroalkyl group. Examples include, but are not limited to, -CH 2 -O-CH 3 , -CH 2 -CH 2 -O-CH 3 , -CH 2 -NH-CH 3 , -CH 2 -CH 2 -NH-CH 3 , -CH 2 -N (CH 3) -CH 3, -CH 2 -CH 2 -NH-CH 3 , -CH 2 -CH 2 -N (CH 3) -CH 3, -CH 2 -S-CH 2 -CH 3 , -CH 2 -CH 2, -S (O) -CH 3, -CH 2 -CH 2 -S (O) 2 -CH 3, -CH 2 -NH-OCH 3 , -CH 2 -O-Si (CH 3) 3, -CH 2 -CH = N-OCH3, and -CH = CH-N (CH3) -CH3. In addition, up to two heteroatoms may be consecutive, such as, by way of example, -CH 2 -NH-OCH 3 and -CH 2 -O-Si (CH 3 ) 3 . Excluding the number of heteroatoms, a "heteroalkyl" may have from 1 to 6 carbon atoms. [0186] The term "bond" or "single bond" refers to a chemical bond between two atoms, or two residues when the atoms bound by the bond are considered to be a part of a larger structure. [0187] The term "residue" refers to a specific segment or functional group of a molecule. Chemical moieties are often recognized chemical entities embedded in or added to a molecule. [0188] As used herein, the substituent "R" appearing alone and without a designation number refers to a substituent selected from alkyl, heteroalkyl, alkenyl, cycloalkyl, aryl, heteroaryl (attached via a ring carbon), and heterocycloalkyl. [0189] The term "optionally substituted" or "substituted" means that the reference group may be substituted with one or more additional groups selected individually and independently from alkyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl, -OH, alkoxy, aryloxy, alkylthio, arylthio, alkylsulfoxide, arylsulfoxide, alkylsulphone, arylsulfone, -CN, alkyne, C 1 -C 6 alkylalkyne, halo, acyl, acyloxy, -CO 2 H, -CO 2 -alkyl, nitro, haloalkyl, fluoroalkyl, and amino, which they include mono- and di-substituted amino groups (e.g., -NH 2 , -NHR, -N (R) 2 ), and the protected derivatives thereof. By way of example, optional substituents may be LsRs, in which each Ls is independently selected from a bond, -O-, -C (= O) -, -S-, -S (= O) -, -S (= O) 2 -, -NH-, -NHC (O) -, -C (O) NH-, S (= O) 2 NH-, -NHS (= O) 2 , -OC (O) NH- , -NHC (O) O-, - (C 1 -C 6 alkyl) -, or - (C 2 -C 6 alkenyl) -; and each Rs is independently selected from H, (C 1 -C 6 alkyl), (C 3 -C 8 cycloalkyl), aryl, heteroaryl, heterocycloalkyl, and C 1 -C 6 heteroalkyl. Protecting groups that can form the protective derivatives of the above substituents are found in sources such as Greene and Wuts, supra. [0190] The methods and formulations described herein include the use of crystalline forms (also known as polymorphs), or pharmaceutically acceptable salts of compounds having the structure of Formulas I, II, III, IV, V or VI, as well as active metabolites of these compounds that have the same type of activity. In some situations, the compounds may exist as tautomers. All tautomers are included within the scope of the compounds presented herein. In addition, the compounds described herein can exist in unsolvated and solvated forms with pharmaceutically acceptable solvents such as water, ethanol and the like. The solvated forms of the compounds presented herein are also considered to be described herein. [0191] Methods of treatment and prevention [0192] Methods for stimulating LXR activity in a cell by contacting the cell with an LXR modulator are described herein. Examples of such LXR modulators are described above. Other LXR modulators that can be used to stimulate LXR activity are identified using screening assays that select such compounds, as described in detail herein. [0193] Prophylactic methods [0194] Methods for preventing skin aging in a subject are described herein by administration to the subject of an LXR modulator. The administration of a prophylactic LXR modulator can occur before the manifestation of the aging symptoms of the skin, so that the aging of the skin is prevented or, alternatively, its progression is delayed. [0195] Therapeutic methods [0196] Methods for modulating the activity of LXR for the treatment of skin aging involving the contact of a cell with an LXR modulator that induces the expression of TIMP1, ASAH1, SPTLC 1 , SMPD1, LASS2, TXNRD1, GPX3, are described herein. GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1 and / or decorin and / or inhibits the expression of TNFa, MMP1, MMP3 and / or IL-8. These methods are done in vitro (for example, by cultivating cell with an LXR modulator) or, alternatively, in vivo (eg, by administering an LXR modulator to a subject). As such, the methods are directed to the treatment of a subject affected by skin aging who would benefit from the induction of expression of TIMP1, ASAH1, SPTLC1, SMPD1, LASS 2 , TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1 and / or decorin and / or the inhibition of TNFa expression, MMP1, MMP3 and / or IL-8. [0197] LXR modulators induce the expression of differential genes in keratinocytes. In human keratinocytes, LXR modulators induce the early differentiation marker of keratinocyte involucrin (IVL), as well as the late differentiation markers loricrin (LOR), filaggrin (FLG) and transglutaminase 1 (TGM1). The LXR modulator can induce the expression of these genes directly or indirectly. [0198] The modulators of LXR increase the expression of genes involved in the synthesis of fatty acids and the transport of lipids in the skin. The ligand of LXR induced the expression of genes involved in the synthesis of fatty acids, namely SREBF1, SREBF2, FASN and SCD, and genes involved in the transport of cholesterol and phospholipids, namely APOE, APOD, ABCG1, ABCA1, ABCA12, ABCA2 and ABCA13. LXR modulators increase the expression of LASS4 and SMPD2 in the skin. [0199] Pharmaceutical compositions and administration methods of LXR modulators [0200] The LXR modulators are administered to subjects in a biologically compatible form suitable for topical administration to treat or prevent skin aging. By "biologically compatible form suitable for topical administration" is meant a form of the LXR modulator to be administered in which any toxic effect is overcome by the therapeutic effects of the modulator. The term "subject" is intended to include living organisms in which an immune response can be elicited, e.g., mammals. The administration of LXR modulators as described herein can be in any pharmacological form that includes a therapeutically effective amount of an LXR modulator alone or in combination with a pharmaceutically acceptable carrier. [0201] The therapeutic or pharmaceutical compositions described herein can be administered by any other appropriate route known in the art including, for example, oral, intravenous, subcutaneous, intramuscular or transdermal, or administration to cells in ex vivo treatment protocols. Administration can be either rapid or by injection or over a period of time such as by slow infusion or administration of slow release formulation. To treat or prevent skin aging, administration of the therapeutic or pharmaceutical compositions described herein can be performed, for example, by topical administration. [0202] Topical administration of an LXR modulator may be in the form of an aerosol, a semi-solid pharmaceutical composition, a powder or a solution. By the term "semi-solid composition" is meant an ointment, cream, balsam, gelatin or other pharmaceutical composition of substantially similar consistency suitable for application to the skin. Examples of semi-solid compositions are given in Chapter 17 of The Theory and Practice of Industrial Pharmacy, Lachman, Lieberman and Kanig, published by Lea and Febiger (1970) and in Chapter 67 of Remington's Pharmaceutical Sciences, 15th Edition (1975) published by Mack Publishing Company. [0203] Dermal or skin patches are another method for the transdermal administration of the therapeutic or pharmaceutical compositions described herein. The patches can provide an absorption enhancer such as DMSO to increase the absorption of the compounds. The patches may include those that control the rate of drug delivery to the skin. The patches can provide a variety of dosing systems that include a reservoir system or a monolithic system, respectively. The reservoir design can have, for example, four layers: the adhesive layer that comes into direct contact with the skin, the control membrane, which controls the diffusion of drug molecules, the deposition of drug molecules and a carrier resistant to the drug. Water. Such a design provides uniform amounts of the drug over a specific period of time, the rate of administration must be less than the saturation limit of different skin types. The monolithic design, for example, typically has only three layers: the adhesive layer, a polymer matrix containing the compound and an impermeable backing. This design brings a quantity of medicine saturation to the skin. Therefore, the administration is controlled by the skin. As the amount of drug decreases in the patch below the saturation level, the rate of administration decreases. [0204] A therapeutically effective amount of an LXR modulator may vary according to factors such as the skin's aging state, age, sex and weight of the individual, and the ability of the LXR modulator to elicit a desired response in the individual. The dosage regimen can be adjusted to provide the optimal aesthetic response. For example, several divided doses may be administered daily, or the dose may be reduced proportionally as indicated by the demands of skin aging. [0205] LXR modulators can also be linked or conjugated with agents that provide desirable pharmaceutical or pharmacodynamic properties. For example, LXR modulators can be stably attached to a polymer such as polyethylene glycol to obtain desirable properties of solubility, stability, half-life and other pharmaceutically advantageous properties (see, for example, Davis et al., Enzyme Eng. 4: 169 73 (1978), Burnham NL, Am. J. Hosp. Pharm. 51: 210-18 (1994)). [0206] The LXR modulators may be in a composition that aids the administration to the cytosol of a cell. For example, an LXR modulator can be conjugated to a carrier moiety such as a liposome that is capable of delivering the modulator in the cytosol of a cell. Such methods are well known in the art (see, for example, Amselem S et al., Chem. Phys. Lipids 64: 219-37 (1993)). [0207] The LXR modulators can be used in the form of pharmaceutical preparations. Such preparations are made in a manner well known in the pharmaceutical art. A preferred preparation uses a physiological saline vehicle, but it is contemplated that other pharmaceutically acceptable carriers may also be used such as physiological concentrations of other non-toxic salts, five percent aqueous glucose solution, sterile water or the like. As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, absorption and isotonic retardants, and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except to the extent that any conventional media or agent is incompatible with the LXR modulator, its use in cosmetic compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions. It may also be desirable that an appropriate buffer be present in the composition. Such solutions can, if desired, be lyophilized and stored in a sterile ampoule ready for reconstitution by the addition of sterile water for rapid injection. The primary solvent may be aqueous or alternatively non-aqueous. [0208] The anti-aging skin compositions described herein may additionally comprise a retinoic acid receptor (RAR) ligand. Useful RAR ligands include, for example, the all-trans-retinoic acid (tretinoin) and / or the synthetic retinoic acid receptor ligands. Tretinoin is sold under brand names such as Atragen®, Avita®, Renova®, Retin-A®, Vesanoid®, and Vitinoin®. Exemplary synthetic retinoic acid receptor ligands include tazarotene (Avage®; ethyl 6- [2- (4,4-dimethylthiochroman-6-yl) ethynyl] pyridine-3-carboxylate) and Differin® (adapalene; [3- (1-adamantyl) -4-methoxyphenyl] -2-naphthoic acid, CD271). [0209] Topical compositions can be prepared by combining the antiaging composition of the skin with conventional pharmaceutically acceptable diluents and vehicles commonly used in dry topical, liquid, cream and aerosol formulations. The ointment and the creams can, for example, be formulated with an aqueous or oily base with the addition of appropriate thickening and / or gelling agents. An exemplary basis is water. Thickening agents which may be used according to the nature of the base include aluminum stearate, cetostearyl alcohol, propylene glycol, polyethylene glycols, hydrogenated lanolin and the like. The lotions can be formulated with an aqueous base and, in general, will also include one or more of the following: stabilizing agents, emulsifiers, dispersants, suspending agents, thickeners, colorants, perfumes and the like. The powders can be formed with the aid of any suitable powder base, for example, talc, lactose, starch and the like. The drops can be formulated with an aqueous base or a non-aqueous base, and can also include one or more dispersing agents, suspending agents, solubilizing agents. [0210] The topical composition can, for example, take the form of a hydrogel based on poly (acrylic acid) or polyacrylamide; in the form of an ointment, for example, with polyethylene glycol (PEG) as vehicle, as the standard DAB 8 ointment (50% PEG 300, 50% PEG 1500); or as an emulsion, especially a microemulsion based on water in oil or oil in water, optionally with added liposomes. Suitable permeation accelerators (entraining agents) include sulfoxide derivatives such as dimethylsulfoxide (DMSO) or decylmethylsulfoxide (decyl-MSO) and transcutol (diethylene glycol monoethyl ether) or cyclodextrin; as well as pyrrolidones, for example, 2-pyrrolidone, N-methyl-2-pyrrolidone, 2-pyrrolidone-5-carboxylic acid, or the biodegradable N- (2-hydroxyethyl) -2-pyrrolidone and the fatty acid esters of the same; urea derivatives such as dodecylurea, 1,3-didodecylurea and 1,3-diphenylurea; terpenes, for example D-limonene, menthone, a-terpinol, carvol, limonene oxide or 1,8-cineol. [0211] Ointments, pastes, creams and gels may also contain excipients, such as starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid and talc, or mixtures thereof. The powders and aerosols may also contain excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. The nanocrystalline antimicrobial metal solutions can be converted into aerosols or sprays by any of the known means routinely used to manufacture aerosolized pharmaceutical products. In general, such methods comprise pressurizing or providing a means for pressurizing a solution vessel, usually with a gaseous vehicle, and passing the pressurized gas through a small orifice. The aerosols may additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane. [0212] The vehicle may also contain other pharmaceutically acceptable excipients to modify or maintain the pH, osmolarity, viscosity, transparency, color, sterility, stability, rate of dissolution or odor of the formulation. Anti-aging skin compositions may also comprise antioxidants, sunscreens, natural retinoids (eg, retinol) and other additives commonly found in skin treatment compositions. [0213] The administration of the dose can be repeated depending on the pharmacokinetic parameters of the dosage formulation and the route of administration used. [0214] [0215] It is especially advantageous to formulate compositions in the form of a dosage unit for ease of administration and uniformity of dosage. The dosage unit form as used herein refers to physically discrete units suitable as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for dosage unit forms is dictated and directly dependent on (a) the unique characteristics of the LXR modulator and the particular therapeutic effect that it is desired to achieve and (b) the inherent limitations in the composition technique of such active compound for the treatment of sensitivity in individuals. The specific dose can easily be calculated by a person of average skill in the art, for example, according to the approximate body weight or surface area of the patient's body or the volume of the body space to be occupied. The dose will also be calculated depending on the particular administration route selected. Further refinement of the calculations necessary to determine the appropriate dose for the treatment is routinely done by persons of average skill in the art. Such calculations can be made without undue experimentation by a person skilled in the art in light of the LXR modulator activities described herein in target cell assay preparations. The exact doses are determined together with the standard dose-response studies. It will be understood that the amount of the composition administered will in fact be determined by a physician, in light of the relevant circumstances that include the condition or conditions to be treated, the choice of composition to be administered, the age, weight, and response of the patient. individual patient, the severity of the patient's symptoms, and the chosen route of administration. [0216] [0217] The toxicity and therapeutic efficacy of such LXR modulators can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, for example, to determine LD 50 (the lethal dose for 50% of the population) and ED 50 (the therapeutically effective dose in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and can be expressed as the LD 50 / ED 50 ratio. Preferred are LXR modulators that show high therapeutic indices. Although LXR modulators that exhibit toxic side effects may be used, care must be taken in designing a delivery system that directs such modulators to the site of the affected tissue to minimize potential damage to uninfected cells and, therefore, reduce side effects. [0218] [0219] The data obtained from cell culture assays and animal studies can be used to formulate a dosage range for use in humans. The dosage of such modulators l X r is preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity. The dosage may vary within this range depending on the dosage form employed and the route of administration used. For any LXR modulator used in a method described herein, the therapeutically effective dose can be estimated initially from cell culture assays. A dose can be formulated in animal models to achieve a concentration range in circulating plasma that includes IC 50 (ie, the concentration of the LXR modulator that achieves an inhibition of half the maximum of the symptoms) as determined in culture. mobile. Such information can be used to more accurately determine useful doses in humans. Plasma levels can be measured, for example, by high performance liquid chromatography. [0220] [0221] Monitoring the influence of LXR modulators on the induction of TIMP1, ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1 and / or decorin expression and / or the inhibition of expression of TNFa, MMP1, MMP3 and / or IL-8 is applied in clinical trials. For example, the effectiveness of an LXR modulator is monitored in clinical trials of subjects exhibiting increased expression of TIMP1, ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1 and / or decorin and / or decreased expression of TNFa, MMP1, MMP3 and / or IL-8. In such clinical trials, the expression of TIMP1, ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA 1 2 , ABCA13, ABCG1, decorin, TNFa, MMP1, MMP3, and / or IL-8 is used as a "reading" or markers of different phenotypes of skin aging. [0222] [0223] Thus, to study the effect of LXR modulators on skin aging, for example, in a clinical trial, the cells are isolated and RNA is prepared and analyzed to determine the expression levels of TIMP1, ASAH1, SPTLC1 , SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1, decorin, TNFa, MMP1, MMP3 and / or IL-8. Gene expression levels (i.e., a gene expression pattern) are quantified, for example, by Northern blot analysis or RT-PCR, by measuring the amount of protein produced, or by measuring the activity levels of TIMP1, ASAH1, SPTLC1 , SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1, decorin, TNFa, MMP1, MMP3 and / or IL-8, all by methods well known to people of average experience in the technique. In this way, the gene expression pattern serves as a marker, indicative of the physiological response of the cells to the LXR modulator. Accordingly, this state of response is determined before, and at various points during, the treatment of the individual with the LXR modulator. [0224] [0225] Also described is a method for monitoring the effectiveness of the treatment of a subject with a modulator of LXR comprising the steps of (i) obtaining a pre-administration sample from a subject prior to administration of the LXR modulator; (ii) detect the level of expression of TIMP1, ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1, decorin, TNFa, MMP1, MMP3, and / or IL-8; (iii) obtaining one or more post-administration samples from the subject; (iv) detect the level of expression of TIMP1, ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1, decorin, TNFa, MMP1, MMP3 and / or IL -8 in the samples after the administration; (v) comparing the expression level of TIMP1, ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, to BcA12, ABCA13, ABCG1, decorin, TNFa, MMP1, MMP3, and / or IL-8 in the pre-administration sample with the expression of TIMP1, ABCA12, decorin, TNFa, m Mp1, MMP3 and / or IL-8 in the sample or samples subsequent to administration; and (vi) altering the administration of the LXR modulator to the subject accordingly. [0226] For example, an increased administration of the LXR modulator may be desirable to increase the expression of TIMP1, ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1 and / or decorin at levels higher than those detected and / or reducing the expression of TNFa, MMP1, MMP3 and / or IL-8 at levels lower than those detected, that is, to increase the effectiveness of the LXR modulator. Alternatively, decreasing the administration of the LXR modulator may be desirable to decrease the expression of TIMP1, ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1 and / or decorin at levels lower than those detected or activity and / or to increase the expression of TNFa, MMP1, MMP3 and / or IL-8 at levels higher than those detected, that is, to decrease the effectiveness of the LXR modulator. Accordingly, the expression of TIMP1, ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1, decorin, TNFa, MMP1, MMP3 and / or IL can be used. -8 as an indicator of the effectiveness of an LXR modulator, even in the absence of an observable phenotypic response. [0227] In addition, in the treatment of skin aging, compositions containing LXR modulators are administered exogenously, and it is desirable to achieve certain target levels of LXR modulator in sera, in any desired tissue compartment, and / or tissue affected. Therefore, it is advantageous to be able to monitor the levels of LXR modulator in a patient or in a biological sample that includes a tissue biopsy sample obtained from a patient and, in some cases, also to monitor the expression levels of TIMP1, ASAH1 , SPTLC1, Expression of SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1, decorin, TNFa, MMP1, MMP3 and / or IL-8. Accordingly, methods for detecting the presence of the LXR modulator in a sample of a patient using the techniques described herein are also described herein. [0228] Screening assays [0229] The expression levels of cytokines and metalloproteases described herein are used to facilitate the design and / or identification of compounds that treat skin aging by means of an LXR-based mechanism. Accordingly, methods (also referred to herein as "screening assays") are provided to identify modulators, ie, LXR modulators, which have a stimulatory or inhibitory effect on, for example, the expression of TIMP1, ASAH1, SPTLC1, SMPD1 , LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1, decorin, TNFa, MMP1, MMP3 and / or IL-8. The compounds thus identified are used as antiaging compounds of the skin as described herein elsewhere. [0230] An exemplary screening assay is a cell-based assay in which a cell expressing LXR is contacted with a test compound and the ability of the test compound to modulate the expression of TIMP1, ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1, decorin, TNFa, MMP1, MMP3 and / or IL-8 by means of a mechanism based on LXR. Determine the ability of the test compound to modulate the expression of TIMP1, ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12, ABCA13, ABCG1, decorin, TNFa, MMP1, MMP3, and / or IL-8 is achieved by monitoring, for example, the levels of DNA, mRNA or protein, or by measuring the activity levels of TIMP1, ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1 , ABCA2, ABCA12, ABCA13, ABCG1, decorin, TNFa, MMP1, MMP3 and / or IL-8. The cell, for example, is of mammalian, e.g., human, origin. [0231] The modulators identified by the screening assays described above are used for treatments as described herein. [0232] Examples [0233] The following examples are offered for illustrative purposes. The starting materials and reagents used for the synthesis of the compounds described herein can be synthesized or obtained from commercial sources, such as, but not limited to, Sigma-Aldrich, Acros Organics, Fluka and Fischer Scientific. [0234] Example 1: Synthesis of intermediate 3- (ethylsulfonyl) benzohydrazide ( 3 ) [0235] [0236] [0237] Stage A: Synthesis of methyl 3- (methylsulfonyl) benzoate ( 2 ) [0238] To a stirred solution of 3- (methylsulfonyl) benzoic acid ( 1 ) (1.5 g, 7.5 mmol) in methanol (25 ml) was added sulfuric acid (1.5 ml) and the reaction mixture was heated to a reflux for 8 h. At the end, the solvent was removed, diluted with water (40 ml) and extracted with ethyl acetate (60 ml x 3). The combined organic layer was washed with a saturated solution of sodium bicarbonate, brine, dried over sodium sulfate and concentrated to give the title compound. 2 (1.6 g, 99%) which was used for a subsequent reaction. [0239] Stage B: Synthesis of 3- (methylsulfonyl) benzohydrazide ( 3 ) [0240] A stirred solution of compound 2 (2.1 g, 9.8 mmol) and hydrazine hydrate (2.4 mL, 49.0 mmol) in ethanol (70 mL) was heated to reflux for 8 h. At the end, the solvent was removed, diluted with water (30 ml) and extracted with 10% methanol in dichloromethane (60 ml x 4). The combined organic layer was washed with brine, dried over sodium sulfate and concentrated to give the title compound 3 (2.0 g, 91%) which was used for the subsequent reaction. [0241] Example 2: Synthesis of 1- (2-chlorophenyl) -5- (5- (3- (methylsulfonyl) phenyl) -1,3,4-thiadiazol-2-yl) -1H-pyrazole-3-carboxylic acid ( 10 ) [0242] [0243] [0244] [0245] Stage 1: Synthesis of methyl 4- (furan-2-yl) -2,4-dioxobutanoate ( 5 ) [0246] To a stirred solution of compound 4 (5 g, 45.0 mmol) in THF (100 ml) at -78 ° C, LiHMDS (58.5 ml, 58.5 mmol) was added dropwise. After 30 minutes, a solution of dimethyl oxalate (7.9 g, 67.5 mmol) in THF (20 ml) was added. The reaction mixture was allowed to reach room temperature in the cooling bath gradually and was stirred overnight. The solvent was removed, water (100 ml) was added and extracted with ethyl acetate (50 ml x 3). The combined organic layer was washed with brine, dried over sodium sulfate and concentrated to give the title compound 5 (8.0 g, 89.9%) which was used for the subsequent reaction. [0247] Stage 2: Synthesis of ethyl 1- (2-chlorophenyl) -5- (furan-2-yl) -1H-pyrazole-3-carboxylate ( 6 ) [0248] A stirred solution of compound 5 (10.0 g, 50 mmol) and (2-chlorophenyl) hydrazine hydrochloride (10.0 g, 56 mmol) in 80 mL of methanol was heated to reflux for 6 h. Upon cooling to 0 ° C, the solid was precipitated, filtered and washed with cold methanol to give the title compound 6 (12.0 g, 80.0%) which was used for a subsequent reaction. [0249] Step 3: Synthesis of 1- (2-dorophenyl) -3- (methoxycarbonyl) -1H-pyrazole-5-carboxylic acid ( 7 ) [0250] To a stirred mixture of compound 6 (3.0 g, 9.9 mmol) in a mixture of acetonitrile (60 ml), water (80 ml) and carbon tetrachloride (60 ml) was added sodium periodate (8.3 g, 39 mmol) and ruthenium chloride (125 mg, 0.03 mmol) and the reaction mixture was stirred at room temperature for 72 h. At the end, the solvent was removed, the crude mass was dissolved in a saturated solution of sodium bicarbonate, extracted with ether (50 ml × 3). The aqueous layer was acidified with 1N HCl and extracted with ethyl acetate (60 ml x 4). The combined ethyl acetate layer was washed with brine, dried over sodium sulfate and concentrated to give the title compound 7 (1.2 g, 44%) which was used for a subsequent reaction. [0251] Step 4: Synthesis of 1- (2-chlorophenyl) -5- (2- (3- (methylsulfonyl) benzoyl) hydrazinocarbonyl) -1H-pyrazole-3-carboxylic acid methyl ester ( 8 ) [0252] To a stirred solution of compound 7 (500 mg, 1.78 mmol) in DMF (20 ml at 0 ° C, EDCI (500 mg, 2.6 mmol) was added and stirred for 15 min. HOBt (351) was added. mg, 2.6 mmol) to the reaction mixture and after 30 minutes of stirring was added 3- (methylsulfonyl) benzohydrazide 3 of Example 1 (450 mg, 2.1 mmol) .The reaction mixture was allowed to reach the Room temperature was stirred overnight.Water (100 ml) was added to the reaction mixture and extracted with ethyl acetate (50 ml x 4) .The combined organic layer was washed with water, brine, dried over sulfate Sodium and concentrated The crude product using column chromatographic purification with 2% methanol in dichloromethane gave the title compound 8 (452 mg, 52%). [0253] Step 5: Synthesis of 1- (2-chlorophenyl) -5- (5- (3- (methylsulfonyl) phenyl) -1,3,4-thiadiazol-2-yl) -1H-pyrazole-3-carboxylic acid methyl ester ( 9 ) [0254] To a stirred mixture of compound 8 (500 mg, 1.05 mmol) and Lawesson's reagent (637 mg, 1.57 mmol) in toluene (10 ml) was added pyridine (0.15 ml) and the reaction mixture was added. heated at reflux for 3 h. At the end, the solvent was removed and the crude reaction mixture was purified by column chromatography using 0.5% methanol in dichloromethane and gave the title compound 9 (250 mg, 51%). LCMS: 475.15 (M 1) +; HPLC: 94.29% (@ 210 nm-370 nm) (Rt; 6.771; method: column: YMC ODS-A 150 mm x 4.6 mm x 5 p; mobile phase: A; 0.05% TFA in water / B, 0.05% TFA in acetonitrile, initial volume: 10 pl, column temperature: 30 ° C, flow rate: 1.4 ml / min, gradient: from 5% B to 95% B in 8 min, maintain for 1.5 min, 9.51-12 min 5% of B); 1 H NMR (CDCh, 400 MHz) 8 8.38 (s, 1H), 8.23 (d, 1H, J = 7.6 Hz), 8.06 (d, 1H, J = 7.6 Hz), 7.70 (t, 1H, J = 8 and 7.6 Hz), 7.66 (s, 1 H), 7.62 - 7.50 (m, 4 H), 4.00 (s, 3 H) ), 3.09 (s, 3 H). [0255] Step 6: Synthesis of 1- (2-chlorophenyl) -5- (5- (3- (methylsulfonyl) phenyl) -1,3,4-thiadiazol-2-yl) -1H-pyrazole-3-carboxylic acid ( 10 ) To a stirred solution of compound 9 (120 mg, 0.25 mmol) in THF (6.0 ml) at room temperature, a solution of lithium hydroxide (52 mg, 1.25 mmol) in water (6 mg) was added. , 0 ml) and stirring was continued for 2 h. At the end, the solvent was removed, diluted with water (20 ml) and washed with ether (30 ml x 3). The aqueous layer was acidified with 1 N HCl and extracted with 10% methanol in dichloromethane (50 ml x 3). The combined organic layer was washed with brine, dried over sodium sulfate and concentrated. The crude product after washing with ether and pentane gave the title compound 10 (80 mg, 69%). LCMS: 461.25 (M 1) +; HPLC: 94.34% (@ 210 nm-370 nm) (Rt; 5.989; method: column: YMC ODS-A 150 mm x 4.6 mm x 5 p; mobile phase: A; 0.05% TFA in water / B, 0.05% TFA in acetonitrile, initial volume: 10 pl, column temperature: 30 ° C, flow rate: 1.4 ml / min, gradient: 5% B to 95% B in 8 min, maintain for 1.5 min, 9.51-12 min 5% of B); 1 H NMR (DMSO-d 6, 400 MHz) 88.39 (s, 1 H), 8.24 (d, 1 H, J = 8 Hz), 8.12 (d, 1 H, J = 8 Hz), 7.85 -7.60 (m, 6H), 3.30 (s, 3H). [0256] Example 3: Synthesis of 1- (2-chlorophenyl) -5- (5- (3- (methylsulfonyl) phenyl) -1,3,4-oxadiazol-2-yl) -1H-pyrazole-3-carboxylic acid ( 12 ) [0257] [0258] [0259] Step 1: Synthesis of 1- (2-dorophenyl) -5- (5- (3- (methylsulfonyl) phenyl) -1,3,4-oxadiazol-2-yl) -1H-pyrazole-3-carboxylic acid ethyl ester ( 11 ) [0260] [0261] To an ice-cooled stirred solution of compound 7 of Example 2 (750 mg, 2.67 mmol) and 3- (methylsulfonyl) benzohydrazide 3 of Example 1 (575 mg, 2.67 mmol) in dichloromethane (25 ml) was added. Imidazolinium chloride (902 mg, 5.34 mmol) and stirred for 30 min. Triethylamine (1.5 ml, 10.68 mmol) was added to the reaction mixture slowly over a period of 30 minutes, allowed to come to room temperature and stirring was continued overnight. Water (50 ml) was added to the reaction mixture and extracted with 10% methanol in dichloromethane (50 ml x 3). The combined organic layer was washed with a saturated solution of sodium bicarbonate, brine, dried over sodium sulfate and concentrated. The crude reaction mixture by chromatographic column purification using 2% methanol in dichloromethane gave the title compound 11 (350 mg, 50%). LCMS: 459.25 (M 1) +; HPLC: 93.72% (@ 210 nm-370 nm) (Rt; 6.704; method: column: YMC ODS-A 150 mm x 4.6 mm x 5 p; mobile phase: A; 0.05% TFA in water / B, 0.05% TFA in acetonitrile, initial volume: 10 pl, column temperature: 30 ° C, flow rate: 1.4 ml / min, gradient: from 5% B to 95% B in 8 min, maintain for 1.5 min, 9.51-12 min 5% of B); 1 H NMR (CDCl 4, 400 MHz) 88.28 (s, 1 H), 8.26 (d, 1 H), 8.11 (d, 1 H, J = 7.6 Hz), 7.75 7.71 (m , 2H), 7.62-7.50 (m, 4H), 4.02 (s, 3H), 3.09 (s, 3H). [0262] [0263] Step 2: Synthesis of 1- (2-chlorophenyl) -5- (5- (3- (methylsulfonyl) phenyl) -1,3,4-oxadiazol-2-yl) -1H-pyrazole-3-carboxylic acid ( 12 ) [0264] [0265] To a stirred solution of compound 11 (100 mg, 0.21 mmol) in THF (4.0 ml) at room temperature, a solution of lithium hydroxide (44 mg, 1.05 mmol) in water (4, 0 ml) and stirring was continued for 2 h. At the end, the solvent was removed, diluted with water (20 ml) and washed with ether (30 ml x 3). The aqueous layer was acidified with 1 N HCl and extracted with 10% methanol in dichloromethane (50 ml x 3). The combined organic layer was washed with brine, dried over sodium sulfate and concentrated. The crude product after washing with ether and pentane gave the title compound 12 (30 mg, 31%). LCMS: 445.10 (M 1) +; HPLC: 96.20% (@ 210 nm-370 nm) (Rt; 5,847; method: column: YMC ODS-A 150 mm x 4.6 mm x 5 p; mobile phase: A; 0.05% TFA in water / B, 0.05% TFA in acetonitrile, initial volume: 10 pl, column temperature: 30 ° C, flow rate: 1.4 ml / min, gradient: from 5% B to 95% B in 8 min, maintain for 1.5 min, 9.51-12 min 5% of B); 1 H NMR (CDCl 3 , 400 MHz) 88.29 (s, 1 H), 8.27 (d, 1 H), 8.12 (d, 1 H, J = 7.6 Hz), 7.79 (s, 1 H ), 7.74 (t, 1H, J = 7.6 and 7.2 Hz), 7.64-7.50 (m, 4 H), 3.10 (s, 3 H). [0266] [0267] Example 4: Synthesis of 2- (5- (5- (3- (methylsulfonyl) phenyl) thiophen-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) ethyl acetate ( 16 ) [0268] [0269] Stage 1: Synthesis of 1- (5-bromothiophen-2-yl) -4,4,4-trifluorobutane-1,3-dione ( 14 ) [0270] To a stirred solution of compound 13 (5.0 g, 24.0 mmol) in THF (50 ml) at -78 ° C, LiHMDS (37 ml, 36.0 mmol) was added dropwise. After 30 minutes, a solution of dimethyl oxalate (2.7 g, 36.0 mmol) in THF (20 ml) was added. The reaction mixture was allowed to gradually return to room temperature in a cooling bath and was stirred overnight. The solvent was removed, water (100 ml) was added and the solution was extracted with ethyl acetate (50 ml x 3). The combined organic layers were washed with brine, dried over sodium sulfate and concentrated to give crude compound 14 (6.0 g, 82.1%) which was used without purification for subsequent reactions. [0271] Step 2: Synthesis of 2- (5- (5-bromothiophen-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) ethyl acetate ( 15 ) [0272] A stirred solution of compound 14 (5.2 g, 17.0 mmol) and ethyl 2-hydrazinylacetate (2.94 g, 19.0 mmol) in methanol (60 mL) was heated to reflux for 1.5 h. Upon cooling to room temperature, the solvent was removed, water (100 ml) was added and the solution was extracted with ethyl acetate (50 ml x 3). The combined organic layers were washed with brine, dried over sodium sulfate and concentrated. The crude product was purified by column chromatography using 10% ethyl acetate in hexane to give compound 15 (1.5 g) and used for other reactions. [0273] Step 3: Synthesis of 2- (5- (5- (3- (methylsulfonyl) phenyl) thiophen-2-yl) -3- (trifluoromethyl) -1H-pyrazole-1-yl) ethyl acetate ( 16 ) A solution Agitation of compound 15 (1.5 g, 3.9 mmol) and 3- (methylsulfonyl) phenyl boronic acid (1.2 g, 5.8 mmol) in DMF (10 mL) was degassed with argon. Tetrakis (triphenylphosphine) palladium (0) (450 mg, 0.3 mmol) was added and the reaction was degassed for 30 minutes. Sodium carbonate (1.03 g, 9.0 mmol) was added to the reaction mixture, the reaction was degassed with argon for 30 minutes and heated at 90 ° C for 3 h. The solvent was removed and the reaction mixture was diluted with water (30 ml) and extracted with ethyl acetate (50 ml x 3). The combined organic layers were washed with saturated brine, dried over sodium sulfate and concentrated. The crude reaction mixture was purified by column chromatography using 30% ethyl acetate in hexane to give 16 (1.2 g, 67%). LCMS: 459.20 (M 1) +; 1 H NMR (DMSO-d 6,400 MHz) 88.13 (s, 1H), 8.05 (d, 1H, J = 7.6 Hz), 7.91 (d, 1H, J = 7.6 Hz) , 7.82 (d, 1H, J = 4 Hz), 7.75 (t, 2H, J = 7.6 Hz), 7.20 (s, 1H), 5.40 (s, 2H), 4 , 01-4.23 (m, 2H), 3.32 (s, 3H), 1.1-1.25 (m, 3H). [0274] Example 5: Synthesis of 2- (4-bromo-5- (5- (3- (methylsulfonyl) phenyl) thiophen-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) ethyl acetate ( 17 ) [0275] [0276] [0277] [0278] After bromination with N-bromosuccinimide, the title compound 17 is prepared from 2- (5- (5- (3- (methylsulfonyl) phenyl) thiophen-2-yl) -3- (trifluoromethyl) -1H- pyrazol-1-yl) ethyl acetate 16 . [0279] Example 6: Synthesis of 2- (5- (3- (methylsulfonyl) phenyl) thiophen-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) 2- (dimethylamino) ethyl acetate ( 19 ) [0280] [0281] [0282] Step 1: Synthesis of 2- (5- (5- (3- (Methylsulfonyl) phenyl) thiophen-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) acetic acid) ( 18 ) A stirred solution of 16 (1.2 g, 2.62 mmol) in THF (4.0 ml) at room temperature, a solution of lithium hydroxide (94 mg, 3.930 mmol) in water (4.0 ml) was added. and the stirring was continued for 2 h. At the end, the solvent was removed, the reaction mixture was diluted with water (20 ml) and washed with ether (30 ml x 3). The aqueous layer was acidified with 1 N HCl and extracted with 10% methanol in dichloromethane (50 ml x 3). The combined organic layer was washed with brine, dried over sodium sulfate and concentrated. The crude product after washing with ether and pentane gave 18 (700 mg, 63%). LCMS: 431.15 (M 1) +; 1 H NMR (DMSO-da, 400 MHz) 813.6 (s, 1H), 8.15 (s, 1H), 8.05 (d, 1H, J = 7.6 Hz), 7.91 (d, 1H, J = 8 Hz), 7.81 (d, 2H, J = 3.6 Hz), 7.76 (t, 1H, J = 7.6 Hz), 7.16 (s, 1 H), 5.23 (s, 2 H), 3.35 (s, 3 H). [0283] Step 2: Synthesis of 2- (5- (5- (3- (methylsulfonyl) phenyl) thiophen-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) 2- (dimethylamino) ethyl acetate ( 19 ) [0284] To a stirred solution of compound 18 (0.2 g, 0.471 mmol), 2- (dimethylamino) ethanol (0.14 mL, 1.41 mmol) and triethylamine (0.13 mL, 0.942 mmol) in DMSO (5 mL ) at RT Pybop (0.360 g, 0.706 mmol) was added and the reaction mixture was stirred overnight. The reaction was then diluted with water (30 ml) and extracted with ethyl acetate (50 ml x 3). The combined organic layer was washed with saturated brine, dried over sodium sulfate and concentrated. The crude reaction mixture was purified by column chromatography using 3% methanol in dichloromethane to give 19 (15 mg, 6%). [0285] Example 7: Synthesis of 2- (4-bromo-5- (5- (3- (methylsulfonyl) phenyl) thiophen-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) 2-acetate (dimethylamino) ethyl ( 20 ) [0286] [0287] [0288] [0289] After bromination with N-bromosuccinimide, the title compound 20 is prepared from 2- (5- (5- (3- (methylsulfonyl) phenyl) thiophen-2-yl) -3- (trifluoromethyl) -1H- 2- (dimethylamino) ethyl pyrazole-1-yl) acetate 19 . [0290] Example 8: Synthesis of methylthiomethyl 2- (5- (5- (3- (methylsulfonyl) phenyl) thiophen-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) acetate ( 21 ) [0291] [0292] [0293] [0294] A stirred mixture of compound 18 (0.3 g, 0.690 mmol), (chloromethyl) (methyl) sulphan (0.269 g, 2.79 mmol) and potassium carbonate (0.480 g, 3.48 mmol) in DMF (5 ml. ),) was heated at 100 ° C for 10 h, diluted with water (30 ml) and extracted with ethyl acetate (50 ml x 3). The combined organic layer was washed with saturated brine, dried over sodium sulfate, and concentrated. The crude reaction mixture was purified by preparative HPLC to give 21 (0.020 g, 6%). LCMS: 491.20 (M 1) +; 1 H NMR (DMSO-da, 400 MHz) 813.6 (s, 1H), 8.18 (s, 1H), 8.08 (d, 1H, J = 8 Hz), 7.92 (d, 1H, J = 8 Hz), 7.85 (d, 1H, J = 3.6 Hz), 7.75 (t, 1H), 7.50 (d, 2H), 5.6-5.3 (m, 2H), 3.4-3.20 (m, 5H), 1.84 (s, 3H). [0295] Example 9: Synthesis of methylthiomethyl 2- (4-bromo-5- (5- (3- (methylsulfonyl) phenyl) thiophen-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) acetate ( 22 ) [0296] [0297] [0298] [0299] After bromination with N-bromosuccinimide, the title compound 22 is prepared from 2- (5- (5- (3- (methylsulfonyl) phenyl) thiophen-2-yl) -3- (trifluoromethyl) -1H- methylthiomethyl pyrazole-1 -yl) acetate 21 . [0300] Example 10: Synthesis of 2- (5- (5- (3- (methylsulfonyl) phenyl) -1H-pyrrol-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) ethyl acetate ( 26 ) [0301] [0302] [0303] [0304] [0305] Following the above reaction sequence, the title compound 26 is prepared from an appropriately protected pyrrole 23 . [0306] Example 11: Synthesis of 2- (5- (4-bromo-5- (3- (methylsulfonyl) phenyl) -1H-pyrrol-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) acetate ethyl ( 27 ) [0307] [0308] [0309] [0310] After bromination with N-bromosuccinimide, the title compound 27 is prepared from 2- (5- (5- (3- (methylsulfonyl) phenyl) -1H-pyrrol-2-yl) -3- (trifluoromethyl) H-pyrazol-1-yl) ethyl acetate 26 . [0311] Example 12: Synthesis of 2- (5- (5- (3- (methylsulfonyl) phenyl) -1H-pyrrol-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) acetate dimethylamino) ethyl ( 29 ) [0312] [0313] Following the above two-step reaction sequence, the title compound 29 is prepared from 2- (5- (5- (3- (methylsulfonyl) phenyl) -1H-pyrrole-2-M) -3- (trifluoromethyl) ) -1H-pyrazol-1-yl) ethyl acetate 26 . [0314] Example 13: Synthesis of 2- (4-bromo-5- (5- (3- (methylsulfonyl) phenyl) thiophen-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) acetate dimethylamino) ethyl ( 30 ) [0315] [0316] [0317] [0318] After bromination with N-bromosuccinimide, the title compound 30 is prepared from 2- (5- (5- (3- (methylsulfonyl) phenyl) -1H-pyrrol-2-yl) -3- (trifluoromethyl) H-pyrazol-1-yl) 2- (dimethylamino) ethyl acetate 29 . [0319] Example 14: Synthesis of methylthiomethyl 2- (5- (3- (3- (methylsulfonyl) phenyl) -1H-pyrrol-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) acetate ( 31 ) [0320] [0321] [0322] [0323] After alkylation, the title compound 31 is prepared from 2- (5- (5- (3- (methylsulfonyl) phenyl) -1H-pyrrol-2-yl) -3- (trifluoromethyl) -1H- pyrazol-1-yl) acetic 28 . [0324] Example 15: Synthesis of 2- (4-bromo-5- (5- (3- (methylsulfonyl) phenyl) -1H-pyrrol-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) acetate of methylthiomethyl ( 32 ) [0325] [0326] [0327] [0328] After bromination with N-bromosuccinimide, the title compound 32 is prepared from 2- (5- (5- (3- (methylsulfonyl) phenyl) -1H-pyrrol-2-yl) -3- (trifluoromethyl) 3 L methylthiomethyl -1H-pyrazol-1-yl) acetate. [0329] Example 16: Synthesis of 2- (5- (3 '- (methylsulfonyl) biphenyl-4-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) ethyl acetate ( 36 ) [0330] [0331] To a stirred solution of compound 33 (15.0 g, 76.0 mmol) in THF (50 mL) at -78 ° C, LiHMDS (114 mL, 114.0 mmol) was added dropwise. After 30 minutes, a solution of dimethyl oxalate (13.6 mL, 114.0 mmol) in THF (100 mL) was added. The reaction mixture was gradually left at room temperature in a cooling bath and stirred overnight. The solvent was removed, water (100 ml) was added and the mixture was extracted with ethyl acetate (50 ml x 3). The combined organic layer was washed with brine, dried over sodium sulfate and concentrated to give compound 34 (16 g, 71%) which was used without purification in subsequent reactions. [0332] Step 2: Synthesis of 5- (4-bromophenyl) -3- (trifluoromethyl) -1H-pyrazole ( 34a ) [0333] A stirred solution of compound 34 (15.0 g, 50.0 mmol) and hydrazine hydrate (10.0 g, 56 mmol) in 150 mL methanol was heated to reflux for 1 h. After cooling in an ice bath for 10 minutes, the solvent was removed, water (100 ml) was added and the reaction mixture was extracted with ethyl acetate (50 ml x 3). The combined organic layer was washed with brine, dried over sodium sulfate and concentrated crude. The crude product was purified by column chromatography using 10% ethyl acetate in hexane, to give the crude pyrazole intermediate 34a . [0334] Step 3: Synthesis of 2- (5- (4-bromophenyl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) ethyl acetate ( 35 ) [0335] To 6 g of 34a in acetonitrile (150 ml) was added cesium carbonate (13.5 g, 41 mmol) followed by ethyl bromoacetate (1.3 ml, 30 mmol) and the reaction mixture was heated to 80 ° C. for 6 h. Upon completion, the solvent was removed, water (100 ml) was added and the crude reaction mixture was extracted with ethyl acetate (50 ml x 3). The combined organic layer was washed with brine, dried over sodium sulfate and concentrated. The crude product was purified by column chromatography using 10% ethyl acetate in hexane to give 35 (2 g, 26%). [0336] Step 4: Synthesis of 2- (5- (3 '- (methylsulfonyl) - [1,1'-biphenyl] -4-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) ethyl acetate ( 35b )) A stirred solution of compound 35 (1.6 g, 4.0 mmol) and (3- (methylthio) phenyl) boronic acid (1.06 g, 6.0 mmol) in DMF (20 ml) was degassed with argon. Tetrakis (triphenylphosphine) palladium (0) (462 mg, 0.4 mmol) was added and the reaction degassed again for 30 minutes. Sodium carbonate (1.06 g, 10.0 mmol) was added to the reaction mixture, and the reaction was degassed again with argon for another 30 minutes. The reaction mixture was heated at 90 ° C for 3 h. The solvent was removed, the reaction mixture was diluted with water (30 ml) and extracted with ethyl acetate (50 ml x 3). The combined organic layer was washed with saturated brine, dried over sodium sulfate and concentrated. The crude reaction mixture was purified by column chromatography using 30% ethyl acetate in hexane to give 35b (1.5 g, 99%). [0337] Step 5: Synthesis of 2- (5- (3 '- (methylsulfonyl) - [1,1'-biphenyl] -4-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) ethyl acetate ( 36 ) To a stirred solution of 35b (1.5 g, 3.5 mmol) in methanol (20.0 ml) and water (20 ml) at room temperature, oxone (5.46 g, 8.9 mmol) was added. ) and stirring was continued for 1.5 h. At the end, the solvent was removed and the reaction mixture was diluted with water (30 ml) and extracted with ethyl acetate (50 ml x 3). The combined organic layer was washed with saturated brine, dried over sodium sulfate and concentrated. The crude reaction mixture was purified by column chromatography using 50% ethyl acetate in hexane to give 36 (1.3 g, 81%). LCMS: 453.20 (M 1) +; 1 H NMR (CD 3 OD, 400 MHz) 88.24 (s, 1 H), 8.04 (d, 1 H, J = 7.6 Hz), 7.99 (d, 1 H, J = 8.4 Hz) , 7.86 (d, 1H, J = 8 Hz), 7.77 (m, 1 H, J = 8.4 Hz), 7.62 (d, 2 H, J = 8.4 Hz), 7 , 62 (d, 2 H, J = 8.4 Hz), 6.80 (s, 1 H), 5.09 (s), 1H), 4.25 -4, 15 (m, 2H), 3 , 19 (s, 3H), 1.25-1.2 (m, 3H). [0338] Example 17: Synthesis of 2- (4-bromo-5- (3 '- (methylsulfonyl) biphenyl-4-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) ethyl acetate ( 37 ) [0339] [0340] After bromination with N-bromosuccinimide, the title compound 37 is prepared from 2- (5- (3 '- (methylsulfonyl) biphenyl-4-yl) -3- (trifluoromethyl) -1H-pyrazole-1 - M) ethyl acetate 36 . [0341] Example 18: Synthesis of 2- (5- (3 '- (methylsulfonyl) biphenyl-4-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) 2- (dimethylamino) ethyl acetate ( 39 ) [0342] [0343] [0344] [0345] Step 1: Synthesis of 2- (5- (3 '- (methylsulfonyl) - [1,1'-biphenyl] -4-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) acetic acid) ( 38 ) To a stirred solution of 36 (1.3 g, 2.0 mmol) in THF (10.0 ml) at room temperature, a solution of lithium hydroxide (103 mg, 4.0 mmol) in water was added. (10.0 ml) and stirring was continued for 2 h. Upon completion, the solvent was removed and the reaction mixture was diluted with water (20 ml) and washed with ether (30 ml x 3). The aqueous layer was acidified with 1 N HCl and extracted with 10% methanol in dichloromethane (50 ml x 3). The combined organic layer was washed with brine, dried over sodium sulfate and concentrated. The crude product after washing with ether and pentane gave 38 (1g, 83%). LCMS: 425.15 (M 1) +; 1 H NMR (DMSO-da, 400 MHz) 813.3 (s, 1H), 8.24 (s, 2H), 8.12 (d, 1H, J = 8 Hz), 7.78 (t, 1H, J = 8 Hz), 7.67 (d, 1H, J = 8.4 Hz), 7.04 (s, 1 H), 5.13 (s, 1 H), 3.25 (s, 3 H) ). Step 2: Synthesis of 2- (5- (3 '- (methylsulfonyl) - [1,1'-biphenyl] -4-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) 2-acetate (dimethylamino) ethyl ( 39 ) [0346] To a stirred solution of compound 38 (0.2 g, 0.471 mmol), 2- (dimethylamino) ethanol (0.14 mL, 1.41 mmol) and triethylamine (0.126 mL, 0.942 mmol) in DMSO (5 mL) a TA was added Pybop (0.360 g, 0.706 mmol) and the reaction mixture was stirred overnight. The reaction was then diluted with water (30 ml) and extracted with ethyl acetate (50 ml x 3). The combined organic layer was washed with saturated brine, dried over sodium sulfate and concentrated. The crude reaction mixture was purified by column chromatography using 3% methanol in dichloromethane to provide 39 (15 mg, 6%). LCMS: 496.25 (M 1) +; 1 H NMR (CD 3 OD, 400 MHz) 88.23 (s, 1H), 8.05 (d, 1H, J = 8 Hz), 8.01 (d, 1H, J = 7.6 Hz), 7 , 88 (d, 1H, J = 8.4 Hz), 7.78 (t, 3H, J = 8 Hz), 7.64 (d, 2H, J = 8 Hz), 6.84 (s, 1H ), 5.2 (s, 2H), 4.5 (t, 2H, J = 4.8 Hz), 3.49 (d, 2H, J = 4.8 Hz), 3.20 (s, 3H) ), 2.9 (s, 6H). [0347] Example 19: Synthesis of 2- (4-bromo-5- (3 '- (methylsulfonyl) biphenyl-4-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) 2- (dimethylamino) ethyl acetate ( 40 ) [0348] After bromination with N-bromosuccinimide, the title compound 40 is prepared from 2- (5- (3 '- (methylsulfonyl) biphenyl-4-yl) -3- (trifluoromethyl) -1H-pyrazole-1 - M) 2- (dimethylamino) ethyl acetate 39 . [0349] Example 20: Synthesis of methylthiomethyl 2- (5- (3 '- (methylsulfonyl) biphenyl-4-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) acetate ( 41 ) [0350] [0351] [0352] [0353] A mixture of compound 38 (0.2 g, 0.471 mmol), chloromethylmethylsulfan (0.182 g, 1.89 mmol) and potassium carbonate (0.324 g, 2.36 mmol) in DMF (5 mL) was stirred at room temperature for 1 h, then it was heated at 100 ° C for 10 h. At the end, the reaction mixture was diluted with water (30 ml) and extracted with ethyl acetate (50 ml x 3). The combined organic layer was washed with saturated brine, dried over sodium sulfate and concentrated. The crude reaction mixture was purified by preparative HPLC to give 41 (15 mg, 7%). LCMS: 485.20 (M 1) +; 1 H NMR (DMSO-d 6,400 MHz) 68.26 (s, 1H), 8.06 (d, 1H, J = 7.2 Hz), 7.99 (d, 1H, J = 8 Hz), 7 , 93 (d, 2H, J = 8.4 Hz), 7.8-7.6 (m, 3H), 6.75 (s, 1H), 5.10-5.25 (m, 2H), 3.45-3.3 (m, 2H), 3.30 (s, 3H), 1.78 (s, 3H), Example 21: Synthesis of 2- (4-bromo-5- (3 '- ( Methylthiomethyl methylsulfonyl) biphenyl-4-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) acetate ( 42 ) [0354] [0355] [0356] [0357] After bromination with N-bromosuccinimide, the title compound 42 is prepared from 2- (5- (3 '- (methylsulfonyl) biphenyl-4-yl) -3- (trifluoromethyl) -1H-pyrazole-1- il) methylthiomethyl acetate 41 . [0358] Example 22: Synthesis of 2- (5- (5- (3- (Methylsulfonyl) phenyl) furan-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) acetic acid ( 48 ) [0359] [0360] [0361] [0362] Stage 1: Synthesis of 1- (5-bromofuran-2-yl) ethanone ( 43 ) [0363] To a stirred solution of compound 4 (5.0 g, 45.45 mmol) and DMF (50 ml), NBS (8.8 g, 50 mmol) was added portionwise at room temperature with stirring. The reaction mixture was allowed to stir at room temperature overnight. There was 50% of starting material left by TLC and LCMS. The reaction mixture was poured into cold water and the compound was extracted with diethyl ether (150 ml x 3). The combined organic layer was washed with brine, dried over sodium sulfate and concentrated under reduced pressure. The crude compound was purified by column chromatography using 5% ethyl acetate in n-hexane as eluent to give compound 43 (2.4 g, 28%) as a white solid. [0364] Stage 2: Synthesis of 1- (5-bromofuran-2-yl) -4,4,4-trifluorobutane-1,3-dione ( 44 ) [0365] To a stirred solution of compound 43 (4.8 g, 25.4 mmol) in THF (60 mL) at -78 ° C, LiHMDS (38 mL, 38.1 mmol) was added dropwise and the reaction was stirred at -78 ° C for 1 h. Then ethyl 2,2,2-trifluoroacetate (4.5 g, 38.1 mmol) was added dropwise. The reaction mixture was allowed to warm gradually to room temperature in a cooling bath and was stirred overnight. The solvent was removed, cold water (50 ml) was added and the reaction mixture was extracted with diethyl ether (100 ml x 3). The combined organic layer was washed with brine, dried over sodium sulfate and concentrated under reduced pressure. The crude compound was washed with diethyl ether and hexane to give compound 44 (5 g, 69%) as a white solid. [0366] Step 3: Synthesis of 2- (5- (5-bromofuran-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) ethyl acetate ( 45 ) [0367] A mixture of compound 44 (4 g, 14.0 mmol), ethyl 2-hydrazinylacetate (2.37 g, 15.0 mmol) and methanol (60 mL) was heated for 1.5 h. The reaction mixture was cooled to 0 ° C and the solvent was removed under reduced pressure. Water (100 ml) was added and the reaction mixture was extracted with ethyl acetate (50 ml x 3). The combined organic layer was washed with brine, dried over sodium sulfate and concentrated under reduced pressure. The crude compound was purified by column chromatography using 10% ethyl acetate in n-hexane as eluent to give the separated isomer 45 (1.2 g, 24%). [0368] Step 4: Synthesis of 2- (5- (5- (3- (methylthio) phenyl) furan-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) ethyl acetate ( 46 ) A mixture of compound 45 (1.25 g, 3.42 mmol) and (3- (methylthio) phenyl) boronic acid (1.15 g, 6.84 mmol) in DMF (50 mL) was degassed with argon for 30 minutes. To the mixture was added tetrakis (triphenylphosphine) palladium (0) (390 mg, 0.34 mmol), degassed for 30 minutes followed by sodium carbonate (0.91 g, 8.56 mmol). The reaction was degassed again with argon for 30 minutes and heated at 90 ° C for 3 h. After completion of the reaction, the solvent was removed under reduced pressure and the reaction mixture was diluted with water (30 ml). The mixture was extracted with ethyl acetate (50 ml x 3) and the combined organic layer was washed with saturated brine (50 ml x 2), dried over sodium sulfate and concentrated under reduced pressure. The crude compound was purified by column chromatography using 5% ethyl acetate in n-hexane as eluent to give compound 46 (1 g, 71%). [0369] Step 5: Synthesis of 2- (5- (5- (3- (methylsulfonyl) phenyl) furan-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) ethyl acetate ( 47 ) A Dissolution of compound 46 (1 g, 2.44 mmol) in DCM (150 ml), mCPBA (1.26 g, 7.31 mmol) was added at room temperature. The reaction mixture was stirred at room temperature for 90 minutes. The reaction mixture was diluted with DCM (100 ml) and washed with water (50 ml x 3). The combined organic layer was washed with saturated brine, dried over sodium sulfate and concentrated under reduced pressure. The crude reaction mixture was purified by column chromatography using 40% ethyl acetate and n-hexane as eluent, to give 47 (0.5 g, 46%). LCMS: 443.20 (M 1) +; 1 H NMR (CDCl 4, 400 MHz) 88.19 (s, 1H), 7.92-7.87 (m, 2H), 7.65-7.62 (t, J = 7.8 Hz, 1H), 6.92-9.91 (d, J = 3.2 Hz, 1H), 6.85 (s, 1H), 6.77-6.76 (d, J = 3.6 Hz, 1H), 5 , 28 (s, 2H), 4.27-4.22 (q, J = 7.06 Hz, 2H), 3.13 (s, 3H), 1.21-1.18 (t, J = 7 Hz, 3H). [0370] Step 6: Synthesis of 2- (5- (5- (3- (Methylsulfonyl) phenyl) furan-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) acetic acid ( 48 ) To a solution Agitation of compound 47 (0.5 g, 1.13 mmol) in THF (5.0 ml) was added lithium hydroxide (41 mg, 1.69 mmol) in water (5.0 ml) and the mixture was stirred at room temperature for 2 h. At the end, the solvent was removed under reduced pressure and the reaction mixture was diluted with water (20 ml) and extracted with diethyl ether (30 ml x 3). The aqueous layer was acidified with 1N HCl (pH = 3) and extracted with 10% methanol in dichloromethane (50 ml x 3). The combined organic layer was washed with brine, dried over sodium sulfate and concentrated under reduced pressure. The crude product after washing with ether and pentane gave 48 (0.4 g, 86%). LCMS: 415.10 (M 1) +; 1 H NMR (DMSO-d 6, 400 MHz) 813.50 (br, 1H), 8.28 (s, 1H), 8.13 to 8.11 (d, J = 8 Hz, 1H), 7.89- 7.87 (d, J = 8 Hz, 1H), 7.76-7.72 (t, J = 8 Hz, 1H), 7.41-7.40 (d, J = 3.6 Hz, 1 H), 7.34 (s, 1 H), 7.19-7.18 (d, J = 3.6 Hz, 1 H), 5.43 (s, 2 H), 3.30 (s, 3 H). [0371] Example 23: Synthesis of 2- (5- (5- (3- (methylsulfonyl) phenyl) thiophen-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) ethyl acetate ( 49 ) [0372] [0373] To a solution of 18 (1 eq) in i-PrOH (50 ml) was added 5 drops of conc. H 2 SO 4 . and the reaction was heated at 90 ° C for 16 h: TLC and crude LCMS showed the formation of the desired ester purified by column chromatography to give 100 mg of 49 . LCMS: 473.6 (M 1) +. [0374] Example 24: Synthesis of ethyl 2- (5- (5- (4- (hydroxymethyl) -3- (methylsulfonyl) phenyl) thiophen-2-yl) -3- (trifluoromethyl) -1H-pyrazoM-yl) ethyl acetate ( 50 ) [0375] [0376] [0377] [0378] The Suzuki coupling of 15 (300 mg) with 2 eq of boronate, 0.1 eq of tetrakis (triphenylphosphine) palladium (0), and 2.5 equivalents of Na 2 CO 3 in 20 ml of DMF at 80 ° C during 2 hours after column purification, 160 mg of compound 50 was given . LCMS: 489.10 (M 1) +. [0379] Example 25: Synthesis of 2- (5- (4 '- (hydroxymethyl) -3' - (methylsulfonyl) biphenyl-4-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) ethyl acetate ( 51 ) [0380] [0381] [0382] [0383] The Suzuki coupling of 35 (300 mg) with 2 eq of boronate, 0.1 eq of tetrakis (triphenylphosphine) palladium (0), and 2.5 equivalents of Na 2 CO 3 in 20 ml of DMF at 80 ° C for 2 hours after purification on a column, 100 mg of compound 51 was given . LCMS: 483.20 (M 1) +. [0384] Example 26: Synthesis of Chloromethyl 2- (5- (5- (3- (methylsulfonyl) phenyl) thiophen-2-yl) -3- (trifluoromethyl) -1H-pyrazol-1-yl) acetate ( 52 ) [0385] [0386] [0387] [0388] Esterification of the carboxylic acid 18 with chloromethylsulfuryl chloride as shown above gave the chloromethyl ester 52 . CMS: 479.2 (M 1) +. [0389] Example 27: RNA extraction [0390] Add QIAzol ® lysis reagent (QIAGEN, catalog number 79306) to the cells. Scrape the cells and place them in a Falcon polypropylene tube. Let stand at room temperature for 5 minutes. Add 1 ml of cells to the microcentrifuge tubes. Add 200 μl of chloroform, shake in a vortex, let stand for 5 minutes. Centrifuge at 4 ° C for 15 minutes at 14,000 RPM. Add an equal volume of 70% EtOH (diluted with DEPC water). Add 600 μl to the RNeasy® column of the RNeasy® Mini Kit (QIAGEN, catalog number 74106), centrifuge at 14,000 RPM at room temperature for 1 minute, discard the filtrate. Add the rest of the sample to the column, centrifuge, discard the filtrate. Add 350 μl of RW1 buffer from the RNeasy® Mini Kit to the column, centrifuge at room temperature for 1 minute, discard the filtrate. DNase column with RNase-Free DNase Set (QIAGEN, catalog number 79254) by preparing the DNase I standard solution, add 550 μl of water to the DNase, add 10 μl of DNase to 70 μl of BufferRDD for each sample , mix, add 80 jla the column, let stand for 15 minutes. Add 350 μl of RW1 buffer to the column, centrifuge for 1 minute, discard the filtrate. Add 500 μl of RPE buffer to the column, centrifuge for 1 minute, discard the filtrate. Add 500 μl of RPE buffer to the column, centrifuge for 1 minute, discard the filtrate. Place the column in a 2.0 ml microcentrifuge tube, centrifuge for 2 minutes. Place the column in a microcentrifuge tube, add 50 μl of water, let stand the column for 2 minutes and centrifuge for 1 minute. Quantitative PCR [0391] TaqMan Technology is used for quantitative PCR for the evaluation of MMP gene expression, TNF, TIMP, IL- 8 , ASAH1, SPTLC1, SMPD1, LASS2, TXNRD1, GPX3, GSR, CAT, ApoE, ABCA1, ABCA2, ABCA12 , ABCA13, ABCG1, decorin, and LXRa / p in keratinocytes and fibroblasts. [0392] Conditions for the use of TaqMan reverse transcriptase reagents (Applied Biosystems, catalog number N808-0234): 10 * RT buffer: 10 pl, MgCh solution: 22 l, DN t P: 20 pl mixture, random hexamers: 5 pl, Multi Scribe RT: 2.5 jl, RNase inhibitor: 2.5 jl, 2 jg of RNA. Thermal cycler: 25 ° C - 10 minutes, 48 ° C - 30 minutes, 95 ° C - 5 minutes. [0393] Configuration of the TaqMan with the QuantiTect Multiplex PCR kit (QIAGEN cat. Number 204543): 2 * master mix: 25 jl; assay in a single tube: 2.5 jl; primer probe set from Applied Biosystems (part number 4308329) -18S forward primer: 0.25 jl, reverse primer 18S: 0.25 jl, 18S probe: 0.25 jl; water up to 50 jl; 5 μl of cDNA. Thermal cycler: 50 ° C. - 2 minutes, 95 ° C - 10 minutes, 95 ° C - 15 seconds, 60 ° C - 1 minute. [0394] Example 28: Induction of LXR receptor expression [0395] Human normal epidermal keratinocytes Clonetics ® (NHEKs) are obtained from Cambrex Bio Science, Inc. Neonatal keratinocytes T-25 (C2503TA25) proliferating clusters are expanded in serum free medium Clonetics ® KGM-2 (CC-3107) and subcultured as necessary using the recommended Clonetics ® ReagentPack ™ (CC-5034). Due to a light sensitive component in the middle, all manipulations are performed in low light. [0396] For the experiments, 1.6 million NHEK cells were plated in culture medium in 100 mm plates and allowed to grow to approximately 75% confluence. On the day of treatment, the plates are washed once with KGM-2 minus hydrocortisone; then vehicle (0.1% DMSO) or 1 jM or an LXR agonist described herein is added for 6 h in KGM-2 deficient in hydrocortisone. After 6 h, the treatment medium is temporarily removed, the plates are washed with Dulbecco's phosphate-buffered saline and then half of the treatments are exposed to ultraviolet light 8 J / m 2 using a Stratagene UV Stratalinker® 2400. The treatments are replaced and 18 h later, the samples are collected for RNA processing using TRIzol®D Reagent (Invitrogen). [0397] The RNA is extracted as described above. UV irradiation of the NHEKs slightly reduced the expression of LXRa. The treatment of keratinocytes with the LXR modulator (1 jM) induces the expression of LXRa in keratinocytes both not exposed to UV rays and exposed to UV rays. UV treatment of NHEKs decreases the expression of LXRp, and this UV-mediated inhibition of LXRp expression is reversed by treatment with the LXR modulator. Therefore, the induction of the expression of both LXR receptors in keratinocytes exposed to UV rays by an LXR modulator indicates the efficiency of the LXR modulator. In addition, LXR modulators can help keratinocytes / skin exposed to UV rays respond better to their effects. [0398] Cotransfection assay of Gal4 LXRp [0399] For transient transfection of HEK 293 cells, 6 x 10 3 cells were deposited in 96-well plates. Each well was transfected with 25 ng of 5 x UAS-luciferase indicator (pG5luc) and 25 ng of plasmid pM LXRp (AA 153 461) human LBD using Fugene 6 reagent (Roche, Indianapolis, IN). The ability of the chimeric protein to transactivate a luciferase reporter plasmid responding to Gal4 in a manner responsive to the concentration of compounds (0.01 - 10 jM) was verified. Luciferase activity at each dose concentration was measured in triplicate using standard substrate reagents (BD Biosciences, San Diego, CA). The data are expressed as relative light units and are shown below in Table 1. [0400] Table 1. EC 50 values for LXR modulators in the LXRp Gal fusion assay. [0401] Compound LXRp Gal (EC 50 ) pM [0402] [0403] [0404] [0405] [0406] [0407] [0408] [0409] [0410] [0411] [0412] [0413] [0414] [0415] [0416] A, EC50 <1 pM; B, EC50 = 1-10 pM; C, EC50> 10 pM [0417] Example 29: Expression of ABCG1 [0418] The NHEKs (Cambrex / Lanza, Walkersville, MD) were grown according to the supplier's recommendations. In general, cells were trypsinized and plated on day 0, and treated with Compounds (1 pM) on day 1. Cells were harvested on day 2 with lysis buffer (AppliedBiosystems / Ambion, Foster City, CA) directly added to the cultured cells after a wash with PBS. The NHEKs were used for the purification of RNA using the Qiagen RNeasy RNA purification column (Qiagen, Hilden, Germany) according to the vendor's protocol or processed directly to cDNA using "Cell-to-cDNA" lysis buffer (Ambion, Foster City, CA). The RNA was isolated and the expression of the ABCG1 gene analyzed by real-time PCR is shown in Figure 1 for three compounds of Formula I-VI: Compound A, Compound B and Compound C. As shown in Figure 1, the Compound A, Compound B and Compound C induce ABCG1 in human keratinocytes. [0419] Example 30: Expression of TNFalpha [0420] The NHEKs are treated and RNA is extracted as described in Example 27. Exposure to UV rays of keratinocytes causes the induction of TNFa expression. A reduced expression of UV-induced TNFα expression in the presence of an LXR agonist described herein indicates less activation of the dermal fibroblasts and less production of metalloproteases that degrade the dermal matrix. [0421] Example 31: Expression of MMP3 [0422] The NHEKs are treated and the RNA extracted as described in Example 27. Exposure to UV rays of the keratinocytes causes the induction of MMP3 expression. A reduced expression of UV-induced MMP-3 expression in the presence of an LXR agonist described herein indicates a reduced degradation of the dermal matrix. [0423] Example 32: Expression of TIMP1 [0424] The NHEKs are treated and RNA is extracted as described in Example 27. Exposure to UV rays of keratinocytes causes the reduction of the basal level of expression of TIMP1 expression. It is expected that a Reduced expression of UV-induced TIMP1 expression in the presence of a LXR agonist described herein neutralizes metalloprotease activities, resulting in protection of the dermal matrix from the action of MMPs. [0425] Example 33: Expression of IL -8 [0426] NHEKs are treated and the RNA is extracted as described in Example 27. Exposure to UV rays of keratinocytes causes the induction of IL- 8 expression. Because IL -8 is a chemotactic molecule, it is expected that a reduced expression of UV-induced IL -8 expression in the presence of a LXR agonist described herein results in a lower recruitment of activated neutrophils in the dermis. Active neutrophils are also a source of MMP and elastase that degrade the dermal matrix in photoaging. [0427] Example 34: Synthesis of lipids [0428] Photo-aged or photodamaged skin shows a defective epidermal barrier function. ABCA12 is a lipid transporter that is essential for the maintenance and development of the epidermal skin barrier function. Therefore, lXr ligands can induce lipid synthesis and its loading in epidermal lamellar bodies by inducing the expression of lipid binding proteins and members of the ABC transporter family required for lipid and cholesterol efflux. These gene regulations also indicate that LXR ligands may exhibit therapeutic effect of xerosis, thus alleviating one of the main symptoms of aged skin that leads to deterioration of the epidermal barrier function and responsible for initiating other serious skin conditions. [0429] The NHEK cells are treated and RNA is extracted as described in Example 27. Exposure to UV rays of keratinocytes causes the decrease of ABCA12 expression in keratinocytes exposed to UV rays. It is expected that a reversal of the expression of UV-induced ABCA12 expression by treatment with a LXR agonist described herein results in normalization of the epidermal barrier function in photoaged skin. The improved epidermal barrier function is expected to reduce dryness of the skin, a hallmark of photodamaged / photo-aged skin. The improved epidermal barrier function is expected to reduce dryness of the skin, a hallmark of photodamaged / photo-aged skin. [0430] Example 35: Collagen [0431] Photoaged and aged skin chronologically shows reduced levels of collagen. Collagen is a component of the extracellular matrix that is required to impart rigidity to the structures of the cellular and dermal matrix. The collagen molecules are arranged in the form of collagen fibrils that are required for the normal architecture of the skin. This fibrillar architecture of collagen degrades on aged / wrinkled skin. Therefore, it is also expected that the restoration of the collagen fibrillar structure results in a therapeutic improvement of the photodamaged / photo-aged skin. [0432] Decorin is a component of the extracellular matrix that is associated with collagen I. In addition, the decorin-collagen interaction is required for the formation of collagen fibrils. In other words, decorin is a critical regulator of the fibrillar genesis of collagen 1. Therefore, it is expected that the increased expression of decorin in photodamaged skin exposed to UV rays induces the generation of collagen fibrils, a process that can improve the laxity and wrinkles of the skin. [0433] The NHEK cells are treated and RNA is extracted as described in Example 27. UV exposure of NHEKs causes the inhibition of decorin expression. It is expected that a reversal of UVB-mediated inhibition of decorin expression by treatment with a LXR agonist described herein results in a normalized decorin expression in keratinocytes exposed to UV. It is expected that the induction of decorin expression results in increased extracellular matrix formation. [0434] Example 36: Expression of MMP1 [0435] The BJ cell line (ATCC # CRL-2522) is obtained from ATCC. It is a normal human fibroblast cell line originally derived from the foreskin, which shows a prolonged lifespan in cultures of 80-90 population doublings. Cells are maintained in Eagle's minimal essential medium with Earle's BSS (EMEM) supplemented with penicillin-streptomycin, 1.0 mM sodium pyruvate, 0.1 mM non-essential amino acids, 2 mM GlutaMAX-1 ™ and fetal bovine serum ( FBS) HyClone at 10%. With the exception of serum, all reagents are obtained from Invitrogen. The cells were subcultured with 0.05% trypsin-EDTA twice a week and kept in a humidified incubator at 37 ° C and 5% CO 2 . [0436] For the experiments, 5 million BJ cells are deposited in 150 mm plates in culture medium. The next day, the culture medium containing phenol red is removed and the plates are washed once with EMEM free of phenol red without serum. The experimental medium is phenol red-free EMEM supplemented as above with the addition of 5% Lipoprotein Deficient Serum (Sigma S-5394) in place of HyClone FBS. [0437] DMSO vehicle (0.1%) or 1 pM or a LXR agonist described here is added to the plates for 6 h; in that At this time, 5 ng / ml of rhTNFa (R & D 210-TA) is added to half of the treatments. Samples are collected with TRIzol® 18 h later and processed. [0438] The RNA is extracted as described above. Treatment with TNFa of human BJ fibroblasts induces the expression of MMP1. It is expected that the inhibition of TNFα-induced MMP1 expression after treatment of human fibroblasts with a LXR agonist described herein results in a reduced degradation of the dermal matrix because MMP1 is the primary matrix collagen destroyer. dermal Example 37: Expression of MMP3 [0439] The BJ cells are treated and the RNA extracted as described in Example 27. Treatment with human fibroblast BJ TNFa induces the expression of MMP3. It is expected that the inhibition of TNFα-induced expression of MMP-3 after treatment of human fibroblasts with a LXR agonist described herein results in reduced degradation of the dermal matrix. [0440] Example 38: Expression of TIMP1 [0441] The BJ cells are treated and RNA is extracted as described in Example 27. Exposure to TNFα of human BJ fibroblasts does not cause the reduction of expression at basal level of TIMP1 expression. It is expected that an LXR agonist described herein that induces the expression of TIMP1 both in fibroblasts exposed to TNFα and in fibroblasts exposed to TNFα neutralizes the metalloprotease activities, resulting in protection of the dermal matrix from the action of MMPs. [0442] Example 39: Biosynthetic route of sphingolipids second messengers ceramide and lipids [0443] The NHEK cells are treated and the RNA is extracted as described in Example 27. Ceramide is one of the major lipids in the differentiated keratinocytes and plays a fundamental role in the skin's barrier function. A comparison of chronologically aged skin and young skin revealed a decrease in ceramide content with age. The decrease in the content of ceramide can be due to a lower differentiation of keratinocytes and to the reduction of the phosphodiesterase activities of ceramide synthase and sphingomyelin (SM) in chronological aging. Serine palmitoyltransferase (SPTLC1) catalyzes the formation of sphinganine from serine and palmitoyl-CoA. Ceramide synthase (LASS2) converts sphinganine to ceramide Phosphodiesterase SM (SMPD) also produces ceramide from SM, and acid ceramidase (ASAH1) produces lipid-second messenger sphingosine from ceramide. [0444] An induction of the expression of enzymes involved in the biosynthetic pathway of the second messenger sphingolipids ceramide and lipid by an LXR agonist described herein is indicative of therapeutic efficacy. Since ceramides and other sphingolipids are involved in the proliferation, differentiation and desquamation of keratinocytes, an increase in the expression of enzymes involved in the synthesis of sphingolipids can help in these processes and alleviate epidermal problems (dry skin, decreased proliferation and differentiation of keratinocytes, fine scales) that are derived from the decrease in sphingolipid production. [0445] Example 40: Antioxidant activities in keratinocytes [0446] The NHEK cells are treated and the RNA extracted as described in Example 27. The UV-mediated oxidative damage in both the epidermis and the dermis due to the accumulation of free radicals throughout life in all probability also promotes the cellular aging. Free radicals or reactive oxygen species cause damage to lipids, proteins and DNA, and cause the cells to enter a stage of senescent type. There are many reports describing the reduction of antioxidant enzymes in the skin with age, including superoxide dismutase, catalase and glutathione peroxidase. [0447] An induction of the expression of enzymes involved in the expression of enzymes involved in antioxidant activities in keratinocytes, for example, the expression of antioxidant enzymes, glutathione peroxidase (GPX3), thioredoxin reductase, glutathione reductase and catalase, by means of an LXR agonist described herein it is indicative of therapeutic efficacy. LXR modulators increase the body's defense system against free radicals, which can reduce the attack of hydrogen peroxide and free radicals on the proteins of skin cells, lipids and DNA. [0448] Example 41: Allergic contact dermatitis of the mouse ear [0449] The mouse contact dermatitis model (ear edema model) has previously been used for the characterization of the topical application of LXR activators for its effect on skin inflammation (Fowler et al., J Invest Dermatol 120: 246 (2003). Phorbol 12-myristate-13-acetate (PMA) was applied topically to both the internal and external surfaces (10 μl on each surface, 20 μl in total) of the left ears to induce irritant contact dermatitis. Acetone (vehicle) was applied alone to the right ears 30 minutes before and 15 minutes after the application of PMA, 20 μl of test compounds were applied to both surfaces of the left ear (40 μl in total). identical with 20 pl of the positive control, clobetasol at 0.05%, while the vehicle group received only the application of acetone, after 6 h, they were collected blood samples (approximately 60 μl) from the retroorbital plexus of 5 mice (from each group) at the 6 h time point, in labeled microtubes, containing K 2 EDTA solution as an anticoagulant. The plasma was immediately collected by centrifugation at 4,000 rpm for 10 minutes at 4 ± 2 ° C and stored at below -70 ° C until the bioassay. The PMA-induced inflammatory attack was evaluated as the percentage increase in the thickness of the ear and / or the weight of the ear in the left ear treated versus the right ear treated with the vehicle. The thickness of the ear was measured with a digital caliber followed by the total weight of the ear to determine the changes in the weights of the ears. The degree of inflammation was quantified according to the following equation: swelling of the ear (%) = 100 * (ab) / b, where a is the thickness / weight of the left ear (treated) and b is the thickness / weight of the right ear (untreated control). After obtaining the samples for thickness / weight evaluation of the ear, biopsies from adjacent sites were obtained for routine histopathological fixation in freshly prepared paraformaldehyde at 4% in phosphate buffered saline. The swelling of the ear and the weight of the ear for Compound A compared to Clobetasol (corticosteroid used to treat various skin disorders) is shown in Figure 2. Compound A reduces swelling and weight of the ear in the model of mouse contact dermatitis. [0450] Example 42: Phase II clinical trial of the safety and efficacy of the compounds of Formula (I), (II), (III), (IV), (V) or (VI) in patients with chronic mild plaque psoriasis a moderate [0451] The objective of this phase II trial is to investigate the safety and efficacy of a topical administration of a compound of Formula (I), (II), (III), (IV), (V) or (VI) in patients with psoriasis in chronic plaques from mild to moderate. Patients: eligible subjects will be men and women 18 years of age and older. [0452] Criteria: [0453] Inclusion criteria: [0454] Psoriasis in chronic plaques of mild to moderate (psoriasis vulgaris), with a duration of at least 6 months; A target plate of at least 9 cm2. [0455] Exclusion criteria: [0456] It shows "rebound" or "outbreak" of chronic plaque psoriasis; [0457] Form of psoriasis without plaques; [0458] He currently has a history of psoriatic arthritis; [0459] Currently drug-induced psoriasis; [0460] Currently in systemic therapy or was in systemic therapy for psoriasis in the previous 6 months; [0461] Currently in phototherapy for psoriasis or was in phototherapy in the previous 3 months. [0462] Study design: [0463] Assignment: randomized [0464] Endpoint classification: safety / efficacy study [0465] Intervention model: parallel assignment [0466] Masking: double blind (subject, researcher) [0467] Main purpose: treatment [0468] Measures of the main result: [0469] Percentage change with respect to the reference value in week 4 in the severity index of the target plate (TPSS) [0470] Measures of the secondary result: [0471] Proportion of subjects with response of general severity of the treatment area of "clear" (0) or "almost clear" psoriasis (1) in weeks 1, 2, 3 and 4; [0472] Proportion of subjects with a difference with respect to the reference value> = 2 stages in the total severity index of the treatment area of psoriasis in weeks 1, 2, 3 and 4 [0473] Percentage change with respect to the reference value in weeks 1, 2, 3 and 4 in the target plate area Change with respect to the reference value in weeks 1, 2, 3 and 4 in the TPSS subscripts for erythema, hardening and peeling [0474] Percentage change with respect to the reference value in TPSS in weeks 1, 2 and 3 [0475] Current and change with respect to the reference value of the itch severity (ITI) of the treatment area in weeks 1, 2, 3 and 4 [0476] Proportion of subjects in each response category of patient satisfaction with study medication (PSSM) in week 4 [0477] Incidence, nature and severity of adverse events observed and reported at the administration site during 4 weeks of treatment [0478] Incidence and severity of burning / stinging of psoriatic or perilesional skin in the treatment area during 4 weeks of treatment [0479] Incidence and severity of perilesional skin reactions in the treatment area measured by Draize score during 4 weeks of treatment [0480] Incidence and severity of adverse events during 4 weeks of treatment [0481] Incidence of clinical laboratory abnormalities and change with respect to the reference value of clinical laboratory values during 4 weeks of treatment [0482] Incidence of clinically significant changes in the physical examination with respect to the reference value during more than 4 weeks of treatment [0483] Incidence of abnormalities of vital signs (blood pressure and heart rate) and change with respect to the reference value of vital signs measurements during 4 weeks of treatment [0484] Incidence of abnormalities in the electrocardiogram (ECG) and change with respect to the reference value of ECG measurements during 4 weeks of treatment [0485] Plasma concentrations of CP-690,550, blood samples in week 4 (day 29) [0486] [0487]
权利要求:
Claims (1) [0001] 1. A compound having the structure of Formula (E): in which: A and B are each nitrogen, wherein A and B are linked together to form a five-membered heteroaryl ring; L 1 is a bond, alkyl C 1 -C 6 heteroalkyl or C1 - C6; L 2 is alkyl of C 1 -C 6 heteroalkyl or C1 - C6; R 1 is hydrogen, halogen, -CF 3 , -OR 8 , -N (R 8 ) 2 , -C (= O) R 8 , -C (= O) OR 8 , -C (= O) N (R 8 ) 2 , -C (= N-OH) R 8 , -C (= S) N (R 8 ) 2 , -C (= CH 2 ) CHa, or -C (= O) OCH 2 SCH 3 ; R 2 is -C (= O) ORg, -C (= O) N (Rg) 2 , -NR ^ C (= O) R 9, -C (= N-OH) Rg, -C (= S) N (R9) 2 , or -C (= O) OCH2SCH3; R 3 is hydrogen, halogen, C 1 -C 6 alkyl, or C 1 -C 6 haloalkyl; R 4 is aryl or heteroaryl; wherein the aryl or heteroaryl is substituted with at least one R 11 ; each R8 and each R 10 are each independently hydrogen, alkyl of C 1 -C 6 heteroalkyl C 1 -C 6 -alkyl of C1 - C6 alkylaryl, aryl, or heteroaryl; each R 9 is independently alkyl of C 1 -C 6 heteroalkyl C 1 -C 6 -alkyl of C1 - C6 alkylaryl, aryl, or heteroaryl; R 11 is independently halogen, nitro, -OR 1 Ü, -N (R 1 ü) 2 , -CN, -C (= O) R 1 ü, -C (= O) OR 1 ü, -C (= O ) N (R 1 u) 2, -NR 1 Uc (= O) R 1 ü, NR 1 2 R 1 ü use, -SOR 1 u, -SO 2 R 1 ü, -SO 2 N (R 1 ii) 2, -C (= O) OCH 2 SCH 3, alkyl of C 1 -C 6 cycloalkyl C 3 -C 8 alkyl, halo C 1 -C 6 heteroalkyl C 1 -C 6 -alkyl C 1 - Ca-aryl, optionally substituted aryl or optionally substituted heteroaryl; or a pharmaceutically acceptable salt or a pharmaceutically acceptable solvate thereof. The compound of claim 1, or a pharmaceutically acceptable salt or a pharmaceutically acceptable solvate thereof, wherein R 4 is aryl. 3. The compound of claim 2, or a pharmaceutically acceptable salt or a pharmaceutically acceptable solvate thereof, wherein R 2 is -C (= O) OR 9 ; and R 9 is alkyl of C1 - C6 or heteroalkyl C1 - C6. 4. The compound of claim 3, wherein L 2 is C 1 -C 6 alkyl. The compound of claim 4, or a pharmaceutically acceptable salt or a pharmaceutically acceptable solvate thereof, wherein L 2 is -CH 2 -. 6 The compound of claim 5, or a pharmaceutically acceptable salt or a pharmaceutically acceptable solvate thereof, wherein L 1 is a bond. The compound of claim 6 , or a pharmaceutically acceptable salt or a pharmaceutically acceptable solvate thereof, wherein R 1 is -CF 3 , -C (= O) R 8, -C (= O) OR 8, -C (= O) N (R 8 ) 2 , or -C (= CH 2) CH 3. 8 The compound of claim 7, or a pharmaceutically acceptable salt or a pharmaceutically acceptable solvate thereof, wherein R 4 is phenyl; wherein the phenyl is substituted with an R 11 . 9. The compound of claim 8 , or a pharmaceutically acceptable salt or a pharmaceutically acceptable solvate thereof, wherein R 11 is -SO 2 R 1 ü and R 1 ü is C 1 -C 6 alkyl. 1ü. A compound selected from: 4ü or a pharmaceutically acceptable salt or a pharmaceutically acceptable solvate thereof. 11. A compound, which is: or a pharmaceutically acceptable salt or a pharmaceutically acceptable solvate thereof. 12. A pharmaceutical composition comprising a pharmaceutically acceptable diluent, excipient or binder, and a compound of any one of claims 1 to 11; or a pharmaceutically acceptable salt or a pharmaceutically acceptable solvate thereof. 13. A compound, salt or solvate of any one of claims 1 to 11 for use in the treatment of a disease, disorder or dermal condition in a mammal that would benefit from the modulation of LXR. The compound, salt or solvate for use according to claim 13, wherein the disease, disorder or dermal condition is selected from skin aging, scarring, psoriasis, dermatitis, eczema, urticaria, rosacea, burns, acne, Ichthyosis, vitiligo, metastatic melanoma and xerosis 15. The compound, salt or solvate for use according to claim 14, wherein the dermatitis is an atopic dermatitis.
类似技术:
公开号 | 公开日 | 专利标题 ES2691079T3|2018-11-23|Hepatic X receptor modulators | for the treatment of diseases, disorders and dermal conditions US20190169131A1|2019-06-06|Liver x receptor | modulators US9981913B2|2018-05-29|Liver X receptor | modulators ES2861503T3|2021-10-06|PPAR agonists, compounds, pharmaceutical compositions and methods of use thereof
同族专利:
公开号 | 公开日 JP2015512883A|2015-04-30| AU2013225840A1|2014-09-18| US20180370959A1|2018-12-27| DK2820013T3|2018-10-29| JP6378630B2|2018-08-22| HRP20181623T1|2018-12-14| CN104284893B|2017-04-19| US20150045399A1|2015-02-12| SI2820013T1|2018-11-30| US8987318B2|2015-03-24| JP2018184467A|2018-11-22| EP2820013A4|2015-11-11| EP2820013B1|2018-07-11| WO2013130892A1|2013-09-06| EP3441390A1|2019-02-13| MX361349B|2018-12-04| CA2866113A1|2013-09-06| BR112014021515A2|2018-03-13| KR102112879B1|2020-05-19| JP6800922B2|2020-12-16| CN104284893A|2015-01-14| HUE040231T2|2019-02-28| PL2820013T3|2019-01-31| SG10201607345YA|2016-11-29| MX2014010537A|2015-03-09| RS57863B1|2018-12-31| CA2866113C|2020-05-12| TR201815013T4|2018-11-21| EP2820013A1|2015-01-07| RS57863B9|2019-10-31| US20210101895A1|2021-04-08| LT2820013T|2018-10-25| AU2013225840B2|2017-11-23| PT2820013T|2018-10-25| ES2691079T3|2018-11-23| IL234413A|2018-05-31| EP2820013B9|2019-01-09| CY1120742T1|2019-12-11| NZ629636A|2017-02-24| KR20140146089A|2014-12-24| SG11201405378VA|2014-09-26| US20150152094A1|2015-06-04| US20170334897A1|2017-11-23| US9637481B2|2017-05-02| HK1206030A1|2015-12-31|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题 US1199771A|1915-07-01|1916-10-03|Bates Valve Bag Co|Wire-twisting machine.| EP1318976B1|2000-09-18|2004-11-24|Glaxo Group Limited|Substituted aminopropoxyaryl derivatives useful as agonists for lxr| AUPR738301A0|2001-08-30|2001-09-20|Starpharma Limited|Chemotherapeutic agents| US7482366B2|2001-12-21|2009-01-27|X-Ceptor Therapeutics, Inc.|Modulators of LXR| CA2486651A1|2002-05-24|2003-12-04|Pharmacia Corporation|Sulfone liver x-receptor modulators| CA2486644A1|2002-05-24|2003-12-04|Pharmacia Corporation|Anilino liver x-receptor modulators| WO2004058717A1|2002-12-20|2004-07-15|X-Ceptor Therapeutics, Inc.|Isoquinolinone derivatives and their use as therapeutic agents| JP2006193426A|2003-09-05|2006-07-27|Sankyo Co Ltd|Substituted condensed-ring pyrimidin-4-one compound| WO2005113499A1|2004-05-20|2005-12-01|Sankyo Company, Limited|Indole compounds| TW200606133A|2004-06-30|2006-02-16|Sankyo Co|Substituted benzene compounds| US7592363B2|2004-08-03|2009-09-22|Wyeth|Indazoles| US20070060589A1|2004-12-21|2007-03-15|Purandare Ashok V|Inhibitors of protein arginine methyl transferases| WO2006109633A1|2005-04-07|2006-10-19|Daiichi Sankyo Company, Limited|Substituted indole compound| US7213797B2|2005-05-20|2007-05-08|Garland Jerome Gaskins|Deck board straightener| EP1910307A1|2005-06-27|2008-04-16|Exelixis, Inc.|Pyrazole based lxr modulators| SG162803A1|2005-06-27|2010-07-29|Exelixis Inc|Imidazole based lxr modulators| WO2007031791A1|2005-09-16|2007-03-22|Arrow Therapeutics Limited|Biphenyl derivatives and their use in treating hepatitis c| WO2007034279A2|2005-09-19|2007-03-29|Pfizer Products Inc.|C3a antagonists and pharmaceutical compositions thereof| CN101309915A|2005-11-14|2008-11-19|Irm责任有限公司|Compounds and compositions as lxr modulators| EP1849781A1|2006-04-28|2007-10-31|Laboratorios del Dr. Esteve S.A.|Substituted 3-Amino-4-hydroxy pyrrolidines compounds, their preparation and use as medicaments| TW200825054A|2006-10-18|2008-06-16|Wyeth Corp|Quinoline compounds| WO2008073825A1|2006-12-08|2008-06-19|Exelixis, Inc.|Lxr and fxr modulators| US8338437B2|2007-02-28|2012-12-25|Methylgene Inc.|Amines as small molecule inhibitors| AU2008284224A1|2007-05-18|2009-02-12|Wyeth Llc|Quinazoline compounds| US20090069288A1|2007-07-16|2009-03-12|Breinlinger Eric C|Novel therapeutic compounds| WO2009021868A2|2007-08-13|2009-02-19|F. Hoffmann-La Roche Ag|Novel piperazine amide derivatives| JP2011507905A|2007-12-21|2011-03-10|ワイス・エルエルシー|Benzimidazole compounds| BRPI0822237A2|2007-12-21|2015-06-30|Wyeth Llc|Imidazo [1,2-a] pyridine compounds| EP2283001A2|2008-05-13|2011-02-16|Boehringer Ingelheim International GmbH|Sulfone compounds which modulate the cb2 receptor| US8063088B2|2008-06-11|2011-11-22|Hoffmann-La Roche Inc.|Imidazolidine derivatives| WO2010054229A1|2008-11-07|2010-05-14|Wyeth Llc|Quinoxaline-based lxr modulators| EP2367801A1|2008-11-19|2011-09-28|Wyeth LLC|Polar quinazolines as liver x receptors modulators| EP2379563B1|2008-12-22|2014-07-16|Merck Sharp & Dohme Corp.|Gamma secretase modulators| US20120058133A1|2009-02-19|2012-03-08|President And Fellows Of Harvard College|Inhibition of trna synthetases and therapeutic applications thereof| EP2398474A4|2009-02-23|2012-12-05|Presidio Pharmaceuticals Inc|Inhibitors of hcv ns5a| NZ596330A|2009-05-28|2013-07-26|Exelixis Patent Co Llc|Lxr modulators| US20120263646A1|2009-10-15|2012-10-18|Guerbet|Imaging agents and their use for the diagnostic in vivo of neurodegenerative diseases, notably alzheimer's disease and derivative diseases| GB0919757D0|2009-11-12|2009-12-30|Johnson Matthey Plc|Polymorphs of bromfenac sodium and methods for preparing bromfenac sodium polymorphs| US8906943B2|2010-08-05|2014-12-09|John R. Cashman|Synthetic compounds and methods to decrease nicotine self-administration| US20130137865A1|2010-08-11|2013-05-30|Taisho Pharmaceutical Co., Ltd.|Heteroaryl-pyrazole derivative| AU2011293201B2|2010-08-27|2015-11-05|Calcimedica Inc.|Compounds that modulate intracellular calcium| AR088728A1|2011-03-25|2014-07-02|Squibb Bristol Myers Co|LXR MODULATORS AS IMIDAZOL PRODROGA| MX361349B|2012-03-02|2018-12-04|Ralexar Therapeutics Inc|Liver x receptor modulators for the treatment of dermal diseases, disorders and conditions.| EP3626309A1|2012-08-13|2020-03-25|The Rockefeller University|Lxrbeta agonist for the treatment of cancer| EP3041834A4|2013-09-04|2017-01-18|Ralexar Therapeutics, Inc.|Liver x receptor modulators| ES2804304T3|2013-09-04|2021-02-05|Ellora Therapeutics Inc|Hepatic X receptor modulators|MX361349B|2012-03-02|2018-12-04|Ralexar Therapeutics Inc|Liver x receptormodulators for the treatment of dermal diseases, disorders and conditions.| ES2694001T3|2013-03-15|2018-12-17|Bristol-Myers Squibb Company|Modulators of LXR| CN104151300B|2013-05-15|2018-07-13|中国医学科学院医药生物技术研究所|The agonist and purposes of the liver X receptor of the disubstituted Benzoquinazole classes of one group of 2,3-| ES2804304T3|2013-09-04|2021-02-05|Ellora Therapeutics Inc|Hepatic X receptormodulators| EP3041834A4|2013-09-04|2017-01-18|Ralexar Therapeutics, Inc.|Liver x receptormodulators| AU2015204572B2|2014-01-10|2020-07-30|Inspirna, Inc.|LXR agonists and uses thereof| EP3397258A2|2015-12-30|2018-11-07|Consejo Nacional De Investigaciones Cientificas Y Tecnicas |Use of brassinosteroid analogs for the treatment of dermal disorders by selectively modulating liver x receptorsand dermal disease treatment by brassinosteroid analogs acting as selective liver x receptormodulators| US20190029984A1|2016-01-11|2019-01-31|The Rockefeller University|Methods for the treatment of myeloid derived suppressor cells related disorders| EP3713575A4|2017-11-21|2021-08-25|Rgenix, Inc.|Polymorphs and uses thereof| WO2021009664A1|2019-07-15|2021-01-21|Novartis Ag|Methods for treating meibomian gland dysfunction with liver x receptor agonists| US11174220B2|2019-12-13|2021-11-16|Inspirna, Inc.|Metal salts and uses thereof|
法律状态:
优先权:
[返回顶部]
申请号 | 申请日 | 专利标题 US201261606160P| true| 2012-03-02|2012-03-02| US201261606160P|2012-03-02| PCT/US2013/028438|WO2013130892A1|2012-03-02|2013-02-28|Liver x receptormodulators for the treatment of dermal diseases, disorders and conditions| 相关专利
Sulfonates, polymers, resist compositions and patterning process
Washing machine
Washing machine
Device for fixture finishing and tension adjusting of membrane
Structure for Equipping Band in a Plane Cathode Ray Tube
Process for preparation of 7 alpha-carboxyl 9, 11-epoxy steroids and intermediates useful therein an
国家/地区
|