Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
FACTOR XIA INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2022/164735
Kind Code:
A1
Abstract:
The present invention provides a compound of Formula I and pharmaceutical compositions comprising one or more said compounds, and methods for using said compounds for treating or preventing thromboses, embolisms, hypercoagulability or fibrotic changes. The compounds are selective Factor XIa inhibitors or dual inhibitors of Factor XIa and plasma kallikrein.

Inventors:
DEBENHAM JOHN (US)
ZHU CHENG (US)
ALI AMJAD (US)
SO SUNG-SAU (US)
GAO YING-DUO (US)
Application Number:
PCT/US2022/013450
Publication Date:
August 04, 2022
Filing Date:
January 24, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MERCK SHARP & DOHME LLC (US)
DEBENHAM JOHN S (US)
ZHU CHENG (US)
ALI AMJAD (US)
SO SUNG SAU (US)
GAO YING DUO (US)
International Classes:
A61K31/4427; C07D213/89; C07D401/14; C07D487/08
Domestic Patent References:
WO2020086416A12020-04-30
WO2017095760A12017-06-08
Foreign References:
US20180339977A12018-11-29
Other References:
AL-HORANI RAMI A.: "Factor XI(a) inhibitors for thrombosis: an updated patent review (2016-present)", EXPERT OPINION ON THERAPEUTIC PATENTS, TAYLOR & FRANCIS, GB, vol. 30, no. 1, 2 January 2020 (2020-01-02), GB , pages 39 - 55, XP055959835, ISSN: 1354-3776, DOI: 10.1080/13543776.2020.1705783
Attorney, Agent or Firm:
MERCK SHARP & DOHME LLC (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS: 1. A compound of the formula I: O NH O wherein R1 is hydrogen, OCHF2, or a pyrazole optionally substituted with CF3; R2 is chloro or fluoro; R3 is hydrogen or fluoro; R4 is hydrogen or C1-3 alkyl; X is CH or N; or a pharmaceutically acceptable salt thereof. 2. The compound of Claim 1 of the formula Ia: O CF3 NH O wherein R2 is chloro or fluoro; R4 is hydrogen, CH3 or CH2CH3; or a pharmaceutically acceptable salt thereof. 3. The compound of Claim 1 of the formula Ib: O NH O wherein R2 is chloro or fluoro; R4 is CH3 or CH2CH3; X is CH or N; or a pharmaceutically acceptable salt thereof. 4. The compound of Claim 3 of the formula Ic: O O wherein R2 is chloro or fluoro; R4 is CH3 or CH2CH3; or a pharmaceutically acceptable salt thereof. 5. The compound of any of Claims 1 to 3 wherein X is CH; or a pharmaceutically acceptable salt thereof. 6. The compound of any of Claims 1 to 3 wherein X is N; or a pharmaceutically acceptable salt thereof. 7. The compound of Claim 1 wherein R1 is OCHF2; or a pharmaceutically acceptable salt thereof.

8. The compound of any one of Claims 1 to 7 wherein R4 is methyl or ethyl; or a pharmaceutically acceptable salt thereof. 9. The compound of any one of Claims 1 to 8 wherein R2 is fluoro; or a pharmaceutically acceptable salt thereof. 10. The compound of any one of Claims 1 to 8 wherein R2 is chloro; or a pharmaceutically acceptable salt thereof. 11. The compound of any one of Claims 1 to 10 wherein R3 is hydrogen; or a pharmaceutically acceptable salt thereof. 12. The compound of any one of Claims 1 to 10 wherein R3 is fluoro; or a pharmaceutically acceptable salt thereof. 13. The compound of Claim 1 selected from: ,

, , or a pharmaceutically acceptable salt thereof. 14. A pharmaceutical composition comprising a compound of any one of Claims 1 to 13 or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier. 15. A method for inhibiting thrombus formation in blood or treating thrombus formation in blood comprising administering a composition of Claim 14 to a mammal in need of thereof. 16. A method for preventing thrombus formation in blood comprising administering a composition of Claim 14 to a mammal in need thereof. 17. A method of treating venous thromboembolism and pulmonary embolism in a mammal comprising administering a composition of Claim 14 to a mammal in need thereof.

18. A method of treating deep vein thrombosis in a mammal comprising administering a composition of Claim 14 to a mammal in need thereof. 19. A method of treating thromboembolic stroke in a mammal comprising administering a composition of Claim 14 to a mammal in need thereof. 20. A compound according to any one of Claims 1 to 13, or a pharmaceutically acceptable salt thereof, for use in the manufacture of a medicament for inhibiting thrombin, inhibiting thrombus formation, treating thrombus formation or preventing thrombus formation in a mammal in need thereof. 21. A compound according to any one of Claims 1 to 13, or a pharmaceutically acceptable salt thereof, for use in therapy.

Description:
TITLE OF THE INVENTION FACTOR XIa INHIBITORS BACKGROUND OF THE INVENTION Factor XIa is a plasma serine protease involved in the regulation of blood coagulation. While blood coagulation is a necessary and important part of the regulation of an organism's homeostasis, abnormal blood coagulation can also have deleterious effects. For instance, thrombosis is the formation or presence of a blood clot inside a blood vessel or cavity of the heart. Such a blood clot can lodge in a blood vessel, blocking circulation and inducing a heart attack or stroke. Thromboembolic disorders are the largest cause of mortality and disability in the industrialized world. Blood clotting is a process of control of the blood stream essential for the survival of mammals. The process of clotting, and the subsequent dissolution of the clot after wound healing has taken place, commence after vascular damage, and can be divided into four phases. The first phase, vasoconstriction or vasocontraction, can cause a decrease in blood loss in the damaged area. In the next phase, platelet activation by thrombin, platelets attach to the site of the vessel wall damage and form a platelet aggregate. In the third phase, formation of clotting complexes leads to massive formation of thrombin, which converts soluble fibrinogen to fibrin by cleavage of two small peptides. In the fourth phase, after wound healing, the thrombus is dissolved by the action of the key enzyme of the endogenous fibrinolysis system, plasmin. Two alternative pathways can lead to the formation of a fibrin clot, the intrinsic and the extrinsic pathway. These pathways are initiated by different mechanisms, but in the later phase they converge to give a common final path of the clotting cascade. In this final path of clotting, clotting factor X is activated. The activated factor X is responsible for the formation of thrombin from the inactive precursor prothrombin circulating in the blood. The formation of a thrombus on the bottom of a vessel wall abnormality without a wound is the result of the intrinsic pathway. Fibrin clot formation as a response to tissue damage or an injury is the result of the extrinsic pathway. Both pathways comprise a relatively large number of proteins, which are known as clotting factors. The intrinsic pathway requires the clotting factors V, VIII, IX, X, XI and XII and also prekallikrein, high molecular weight kininogen, calcium ions and phospholipids from platelets. The activation of factor XIa is a central point of intersection between the two pathways of activation of clotting. Factor XIa has an important role in blood clotting. Coagulation is initiated when blood is exposed to artificial surfaces (e.g., during hemodialysis, "on-pump" cardiovascular surgery, vessel grafts, bacterial sepsis), on cell surfaces, cellular receptors, cell debris, DNA, RNA, and extracellular matrices. This process is also termed contact activation. Surface absorption of factor XII leads to a conformational change in the factor XII molecule, thereby facilitating activation to proteolytic active factor XII molecules (factor XIIa and factor XIIf). Factor XIIa (or XIIf) has a number of target proteins, including plasma prekallikrein and factor XI. Active plasma kallikrein further activates factor XII, leading to an amplification of contact activation. Alternatively, the serine protease prolylcarboxylpeptidase can activate plasma kallikrein complexed with high molecular weight kininogen in a multiprotein complex formed on the surface of cells and matrices (Shariat-Madar et al., Blood, 108:192-199 (2006)). Contact activation is a surface mediated process responsible in part for the regulation of thrombosis and inflammation, and is mediated, at least in part, by fibrinolytic-, complement-, kininogen/kinin-, and other humoral and cellular pathways (for review, Coleman, R., "Contact ActivationPathway", Hemostasis and Thrombosis, pp.103-122, Lippincott Williams & Wilkins (2001); Schmaier, A.H., "Contact Activation", Thrombosis and Hemorrhage, pp.105-128 (1998)). The biological relevance of the contact activation system for thromboembolic diseases is supported by the phenotype of factor XII deficient mice. More specifically, factor XII deficient mice were protected from thrombotic vascular occlusion in several thrombosis models as well as stroke models and the phenotype of the XII deficient mice was identical to XI deficient mice (Renne et al., J Exp. Med., 202:271-281 (2005); Kleinschmitz et al., J Exp. Med., 203:513-518 (2006)). The fact that factor XI is downstream from factor XIIa, combined with the identical phenotype of the XII and XI deficient mice suggest that the contact activation system could play a major role in factor XI activation in vivo. Plasma kallikrein is a zymogen of a trypsin-like serine protease and is present in plasma. The gene structure is similar to that of factor XI. Overall, the amino acid sequence of plasma kallikrein has 58% homology to factor XI. Proteolytic activation by factor XIIa at an internal I 389-R390 bond yields a heavy chain (371 amino acids) and a light chain (248 amino acids). The active site of plasma kallikrein is contained in the light chain. The light chain of plasma kallikrein reacts with protease inhibitors, including alpha 2 macroglobulin and Cl-inhibitor. Interestingly, heparin significantly accelerates the inhibition of plasma kallikrein by antithrombin III in the presence of high molecular weight kininogen (HMWK). In blood, the majority of plasma kallikrein circulates in complex with HMWK. Plasma kallikrein cleaves HMWK to liberate bradykinin. Bradykinin release results in increase of vascular permeability andvasodilation (for review, Coleman, R., "Contact Activation Pathway", Hemostasis and Thrombosis, pp.103-122, Lippincott Williams & Wilkins (2001); Schmaier A.H., "Contact Activation", Thrombosis and Hemorrhage, pp.105-128 (1998)). Patients presenting genetic deficiency on C1-esterase inhibitor suffer from hereditary angioedema (HAE), a lifelong disease that results in intermittent swelling throughout the body, including the hands, feet, face, throat, genitals and gastrointestinal tract. Analysis of blisters arising from acute episodes have been shown to contain high levels of plasma kallikrein, and treatment with a protein-based reversible plasma kallikrein inhibitor, Ecallantide (Kalbitor), has been approved by the FDA for the treatment of acute attacks of HAE (Schneider, L, et al., J.Allergy Clin.Immunol., 120: p.416 (2007)). Additionally, the plasma kallikrein-kinin system is abnormally abundant in patients diagnosed with advanced diabetic macular edema (DME). Recent publications have shown that plasma kallikrein contributes to observed retinal vascular leakage and dysfunction in diabetic rodent models (A. Clermont, et al., Diabetes, 60:1590 (2011)), and that treatment with a small molecule plasma kallikrein inhibitor ameliorated the observed retinal vascular permeability and other abnormalities related to retinal blood flow. SUMMARY OF THE INVENTION The present invention relates to compounds of Formula I: and pharmaceutically acceptable salts thereof. The compounds of Formula I are selective Factor XIa inhibitors or dual inhibitors of Factor XIa and plasma kallikrein, and as such may be useful in the treatment, inhibition or amelioration of one or more disease states that could benefit from inhibition of Factor XIa or plasma kallikrein, including thromboses, embolisms, hypercoagulability or fibrotic changes. The compounds of this invention could further be used in combination with other therapeutically effective agents, including but not limited to, other drugs useful for the treatment of thromboses, embolisms, hypercoagulability or fibrotic changes. The invention furthermore relates to processes for preparing compounds of Formula I, and pharmaceutical compositions which comprise compounds of Formula I and pharmaceutically acceptable salts thereof. DETAILED DESCRIPTION OF THE INVENTION The present invention relates to compounds of Formula I: O O wherein R 1 is hydrogen, OCHF2, or a pyrazole optionally substituted with CF3; R 2 is chloro or fluoro; R 3 is hydrogen or fluoro; R 4 is hydrogen or C 1-3 alkyl; X is CH or N; or a pharmaceutically acceptable salt thereof. Another embodiment of the invention relates to compounds of Formula Ia: O CF O wherein R 2 is chloro or fluoro; R 4 is hydrogen, CH 3 or CH 2 CH 3 ; or a pharmaceutically acceptable salt thereof. An embodiment of the present invention relates to compounds of Formula Ib:

wherein R 2 is chloro or fluoro; R 4 is hydrogen, CH 3 or CH 2 CH 3 ; X is CH or N; or a pharmaceutically acceptable salt thereof. Another embodiment of the invention relates to compounds of Formula Ic: O wherein R 2 is chloro or fluoro; R 4 is CH 3 or CH 2 CH 3 ; or a pharmaceutically acceptable salt thereof. In an embodiment of the invention, R 1 is hydrogen. In another embodiment of the invention, R 1 is OCHF2. In another embodiment of the invention, R 1 is a pyrazole optionally substituted with CF 3 . In a class of the embodiment, R 1 is pyrazole substituted with CF 3 . In an embodiment of the invention, R 2 is chloro. In another embodiment of the invention, R 2 is fluoro. In an embodiment of the invention, R 3 is hydrogen. In another embodiment of the invention, R 3 is fluoro. In an embodiment of the invention, R 4 is CH3. In another embodiment of the invention, R 4 is CH2CH3. In another embodiment of the invention, R 4 is hydrogen. In an embodiment of the invention, X is CH. In another embodiment of the invention, X is N. In an embodiment of the invention, the compound of Formula I is: , , or a pharmaceutically acceptable salt thereof. Reference to the preferred classes and subclasses set forth above is meant to include all combinations of particular and preferred groups unless stated otherwise. Specific embodiments of the present invention include, but are not limited to the compounds identified herein as Examples 1 to 9, or pharmaceutically acceptable salts thereof. Also included within the scope of the present invention is a pharmaceutical composition which is comprised of a compound of Formula I, Formula Ia, Formula Ib, or Formula Ic as described above and a pharmaceutically acceptable carrier. The invention is also contemplated to encompass a pharmaceutical composition which is comprised of a pharmaceutically acceptable carrier and any of the compounds specifically disclosed in the present application. These and other aspects of the invention will be apparent from the teachings contained herein. The invention also includes compositions for inhibiting loss of blood platelets, inhibiting formation of blood platelet aggregates, inhibiting formation of fibrin, inhibiting thrombus formation, inhibiting embolus formation, treating inflammatory disorders, treating diabetic retinopathy and treating hereditary angioedema in a mammal, comprising a compound of the invention in a pharmaceutically acceptable carrier. These compositions may optionally include anticoagulants, antiplatelet agents, and thrombolytic agents. The compositions can be added to blood, blood products, or mammalian organs in order to effect the desired inhibitions. The invention also includes compositions for preventing or treating unstable angina, refractory angina, myocardial infarction, transient ischemic attacks, atrial fibrillation, thrombotic stroke, embolic stroke, deep vein thrombosis, disseminated intravascular coagulation, ocular build up of fibrin, and reocclusion or restenosis of recanalized vessels, in a mammal, comprising a compound of the invention in a pharmaceutically acceptable carrier. These compositions may optionally include anticoagulants, antiplatelet agents, and thrombolytic agents. The invention also includes methods for treating unstable angina, refractory angina, myocardial infarction, transient ischemic attacks, atrial fibrillation, thrombotic stroke, embolic stroke, deep vein thrombosis, disseminated intravascular coagulation, ocular build up of fibrin, and reocclusion or restenosis of recanalized vessels, comprising administering a composition of the compound of the invention to a mammal in need thereof. The invention also includes a method for reducing the thrombogenicity of a surface in a mammal by attaching to the surface, either covalently or noncovalently, a compound of the invention. The invention also includes a compound of the invention, or a pharmaceutically acceptable salt thereof, for use in the manufacture of a medicament for inhibiting thrombin, inhibiting thrombus formation, treating thrombus formation or preventing thrombus formation in a mammal. In addition, the invention includes a compound of the invention, or a pharmaceutically acceptable salt thereof, for use in therapy. Compounds of the invention are Factor XIa inhibitors and may have therapeutic value in, for example, preventing coronary artery disease. The compounds of the invention have improved pharmacokinetic profiles compared to compounds known in the art. Furthermore, some of the compounds of the invention have a better combination of potency, efficacy and pharmacokinetic properties compared to known compounds. It will be understood that, as used herein, the compounds of the present invention include the pharmaceutically acceptable salts of the compounds of structural Formula I, Formula Ia, Formula Ib and Formula Ic, and also salts that are not pharmaceutically acceptable when they are used as precursors to the free compounds or their pharmaceutically acceptable salts or in other synthetic manipulations. The compounds of the present invention may be administered in the form of a pharmaceutically acceptable salt. The term "pharmaceutically acceptable salt" refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids including inorganic or organic bases and inorganic or organic acids. Salts of basic compounds encompassed within the term "pharmaceutically acceptable salt" refer to non-toxic salts of the compounds of this invention which are generally prepared by reacting the free base with a suitable organic or inorganic acid. Representative salts of basic compounds of the present invention include, but are not limited to, the following: acetate, ascorbate, adipate, alginate, aspirate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, butyrate, camphorate, camphorsulfonate, camsylate, carbonate, chloride, clavulanate, citrate, cyclopentane propionate, diethylacetic, digluconate, dihydrochloride, dodecylsulfanate, edetate, edisylate, estolate, esylate, ethanesulfonate, formic, fumarate, gluceptate, glucoheptanoate, gluconate, glutamate, glycerophosphate, glycollylarsanilate, hemisulfate, heptanoate, hexanoate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, 2-hydroxyethanesulfonate, hydroxynaphthoate, iodide, isonicotinic, isothionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, methanesulfonate, mucate, 2- naphthalenesulfonate, napsylate, nicotinate, nitrate, N-methylglucamine ammonium salt, oleate, oxalate, pamoate (embonate), palmitate, pantothenate, pectinate, persulfate, phosphate/diphosphate, pimelic, phenylpropionic, polygalacturonate, propionate, salicylate, stearate, sulfate, subacetate, succinate, tannate, tartrate, teoclate, thiocyanate, tosylate, triethiodide, trifluoroacetate, undeconate, valerate and the like. Furthermore, where the compounds of the invention carry an acidic moiety, suitable pharmaceutically acceptable salts thereof include, but are not limited to, salts derived from inorganic bases including aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic, mangamous, potassium, sodium, zinc, and the like. Also included are the ammonium, calcium, magnesium, potassium, and sodium salts. Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, cyclic amines, dicyclohexyl amines and basic ion-exchange resins, such as arginine, betaine, caffeine, choline, N,N- dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like. Also, included are the basic nitrogen-containing groups may be quaternized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl; and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides and others. These salts can be obtained by known methods, for example, by mixing a compound of the present invention with an equivalent amount and a solution containing a desired acid, base, or the like, and then collecting the desired salt by filtering the salt or distilling off the solvent. The compounds of the present invention and salts thereof may form solvates with a solvent such as water, ethanol, or glycerol. The compounds of the present invention may form an acid addition salt and a salt with a base at the same time according to the type of substituent of the side chain. If the compounds of Formula I, Formula Ia, Formula Ib or Formula Ic simultaneously contain acidic and basic groups in the molecule the invention also includes, in addition to the salt forms mentioned, inner salts or betaines (zwitterions). The present invention encompasses all stereoisomeric forms of the compounds of Formula I, Formula Ia, Formula Ib and Formula Ic. Unless a specific stereochemistry is indicated, the present invention is meant to comprehend all such isomeric forms of these compounds. Centers of asymmetry that are present in the compounds of Formula I, Formula Ia, Formula Ib and Formula Ic can all independently of one another have (R) configuration or (S) configuration. When bonds to the chiral carbon are depicted as straight lines in the structural Formulas of the invention, it is understood that both the (R) and (S) configurations of the chiral carbon, and hence both each individual enantiomer and mixtures thereof, are embraced within the Formula. When a particular configuration is depicted, that enantiomer (either (R) or (S), at that center) is intended. Similarly, when a compound name is recited without a chiral designation for a chiral carbon, it is understood that both the (R) and (S) configurations of the chiral carbon, and hence individual enantiomers and mixtures thereof, are embraced by the name. The production of specific stereoisomers or mixtures thereof may be identified in the Examples where such stereoisomers or mixtures were obtained, but this in no way limits the inclusion of all stereoisomers and mixtures thereof from being within the scope of this invention. Unless a specific enantiomer or diastereomer is indicated, the invention includes all possible enantiomers and diastereomers and mixtures of two or more stereoisomers, for example mixtures of enantiomers and/or diastereomers, in all ratios. Thus, enantiomers are a subject of the invention in enantiomerically pure form, both as levorotatory and as dextrorotatory antipodes, in the form of racemates and in the form of mixtures of the two enantiomers in all ratios. In the case of a cis/trans isomerism the invention includes both the cis form and the transform as well as mixtures of these forms in all ratios. The preparation of individual stereoisomers can be carried out, if desired, by separation of a mixture by customary methods, for example by chromatography or crystallization, by the use of stereochemically uniform starting materials for the synthesis or by stereoselective synthesis. Optionally a derivatization can be carried out before a separation of stereoisomers. The separation of a mixture of stereoisomers can be carried out at an intermediate step during the synthesis of a compound of Formula I, Formula Ia, Formula Ib or Formula Ic or it can be done on a final racemic product. Absolute stereochemistry may be determined by X-ray crystallography of crystalline products or crystalline intermediates which are derivatized, if necessary, with a reagent containing a stereogenic center of known configuration. Where compounds of this invention are capable of tautomerization, all individual tautomers as well as mixtures thereof are included in the scope of this invention. The present invention includes all such isomers, as well as salts, solvates (including hydrates) and solvated salts of such racemates, enantiomers, diastereomers and tautomers and mixtures thereof. In the compounds of the invention, the atoms may exhibit their natural isotopic abundances, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature. The present invention is meant to include all suitable isotopic variations of the specifically and generically described compounds. For example, different isotopic forms of hydrogen (H) include protium (1H) and deuterium (2H). Protium is the predominant hydrogen isotope found in nature. Enriching for deuterium may afford certain therapeutic advantages, such as increasing in vivo half-life or reducing dosage requirements, or may provide a compound useful as a standard for characterization of biological samples. Isotopically-enriched compounds can be prepared without undue experimentation by conventional techniques well known to those skilled in the art or by processes analogous to those described in the general process schemes and examples herein using appropriate isotopically- enriched reagents and/or intermediates. When any variable occurs more than one time in any constituent, its definition on each occurrence is independent at every other occurrence. Also, combinations of substituents and variables are permissible only if such combinations result in stable compounds. Lines drawn into the ring systems from substituents represent that the indicated bond may be attached to any of the substitutable ring atoms. If the ring system is bicyclic, it is intended that the bond be attached to any of the suitable atoms on either ring of the bicyclic moiety. It is understood that one or more silicon (Si) atoms can be incorporated into the compounds of the instant invention in place of one or more carbon atoms by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art from readily available starting materials. Carbon and silicon differ in their covalent radius leading to differences in bond distance and the steric arrangement when comparing analogous C-element and Si-element bonds. These differences lead to subtle changes in the size and shape of silicon-containing compounds when compared to carbon. One of ordinary skill in the art would understand that size and shape differences can lead to subtle or dramatic changes in potency, solubility, lack of off-target activity, packaging properties, and so on. (Diass, J. O. et al. Organometallics (2006) 5:1188-1198; Showell, G.A. et al. Bioorganic & Medicinal Chemistry Letters (2006) 16:2555-2558). It is understood that substituents and substitution patterns on the compounds of the instant invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below, from readily available starting materials. If a substituent is itself substituted with more than one group, it is understood that these multiple groups may be on the same carbon or on different carbons, so long as a stable structure results. The phrase “optionally substituted” (with one or more substituents) should be understood as meaning that the group in question is either unsubstituted or may be substituted with one or more substituents. Furthermore, compounds of the present invention may exist in amorphous form and/or one or more crystalline forms, and as such all amorphous and crystalline forms and mixtures thereof of the compounds of Formula I, Formula Ia, Formula Ib and Formula Ic are intended to be included within the scope of the present invention. In addition, some of the compounds of the instant invention may form solvates with water (i.e., a hydrate) or common organic solvents. Such solvates and hydrates, particularly the pharmaceutically acceptable solvates and hydrates, of the instant compounds are likewise encompassed within the scope of this invention, along with un-solvated and anhydrous forms. Also, in the case of a carboxylic acid (-COOH) or alcohol group being present in the compounds of the present invention, pharmaceutically acceptable esters of carboxylic acid derivatives, such as methyl, ethyl, or pivaloyloxymethyl, or acyl derivatives of alcohols, such as O-acetyl, O-pivaloyl, O-benzoyl, and O-aminoacyl, can be employed. Included are those esters and acyl groups known in the art for modifying the solubility or hydrolysis characteristics for use as sustained-release or prodrug formulations. Any pharmaceutically acceptable pro-drug modification of a compound of this invention which results in conversion in vivo to a compound within the scope of this invention is also within the scope of this invention. For example, esters can optionally be made by esterification of an available carboxylic acid group or by formation of an ester on an available hydroxy group in a compound. Similarly, labile amides can be made. Pharmaceutically acceptable esters or amides of the compounds of this invention may be prepared to act as pro-drugs which can be hydrolyzed back to an acid (or -COO- depending on the pH of the fluid or tissue where conversion takes place) or hydroxy form particularly in vivo and as such are encompassed within the scope of this invention. Examples of pharmaceutically acceptable pro-drug modifications include, but are not limited to, -C 1-6 alkyl esters and –C 1-6 alkyl substituted with phenyl esters. Accordingly, the compounds within the generic structural formulas, embodiments and specific compounds described and claimed herein encompass salts, all possible stereoisomers and tautomers, physical forms (e.g., amorphous and crystalline forms), solvate and hydrate forms thereof and any combination of these forms, as well as the salts thereof, pro-drug forms thereof, and salts of pro-drug forms thereof, where such forms are possible unless specified otherwise. Except where noted, the term "halogen" or “halo” means fluorine, chlorine, bromine or iodine. “Celite®” (Fluka) diatomite is diatomaceous earth, and can be referred to as "celite". Except where noted herein, bicyclic ring systems include fused ring systems, where two rings share two atoms, and spiro ring systems, where two rings share one atom. The invention also relates to medicaments containing at least one compound of the Formula I, Formula Ia, Formula Ib or Formula Ic and/or of a pharmaceutically acceptable salt of the compound of the Formula I, Formula Ia, Formula Ib or Formula Ic and/or an optionally stereoisomeric form of the compound of the Formula I, Formula Ia, Formula Ib or Formula Ic or a pharmaceutically acceptable salt of the stereoisomeric form of the compound of Formula I, Formula Ia, Formula Ib or Formula Ic, together with a pharmaceutically suitable and pharmaceutically acceptable vehicle, additive and/or other active substances and auxiliaries. Anticoagulant therapy is indicated for the treatment and prevention of a variety of thrombotic conditions, particularly coronary artery and cerebrovascular disease. Those experienced in this field are readily aware of the circumstances requiring anticoagulant therapy. The term “patient” used herein is taken to mean mammals such as primates, humans, sheep, horses, cattle, pigs, dogs, cats, rats, and mice. The compounds may be selective Factor XIa inhibitors or dual inhibitors of Factor XIa and plasma kallikrein. Factor XIa or dual Factor XIa/plasma kallikrein inhibition are useful not only in the anticoagulant therapy of individuals having thrombotic conditions, but are useful whenever inhibition of blood coagulation is required such as to prevent coagulation of stored whole blood and to prevent coagulation in other biological samples for testing or storage. Thus, the Factor XIa or dual Factor XIa/plasma kallikrein inhibitors can be added to or contacted with any medium containing or suspected of containing thrombin and in which it is desired that blood coagulation be inhibited, e.g., when contacting the mammal’s blood with material selected from the group consisting of vascular grafts, stents, orthopedic prosthesis, cardiac prosthesis, and extracorporeal circulation systems. Compounds of the invention may be useful for treating or preventing venous thromboembolism (e.g., obstruction or occlusion of a vein by a detached thrombus; obstruction or occlusion of a lung artery by a detached thrombus), cardiogenic thromboembolism (e.g., obstruction or occlusion of the heart by a detached thrombus), arterial thrombosis (e.g., formation of a thrombus within an artery that may cause infarction of tissue supplied by the artery), atherosclerosis (e.g., arteriosclerosis characterized by irregularly distributed lipid deposits) in mammals, and for lowering the propensity of devices that come into contact with blood to clot blood. Examples of venous thromboembolism which may be treated or prevented with compounds of the invention include obstruction of a vein, obstruction of a lung artery (pulmonary embolism), deep vein thrombosis, thrombosis associated with cancer and cancer chemotherapy, thrombosis inherited with thrombophilic diseases such as Protein C deficiency, Protein S deficiency, antithrombin III deficiency, and Factor V Leiden, and thrombosis resulting from acquired thrombophilic disorders such as systemic lupus erythematosus (inflammatory connective tissue disease). Also with regard to venous thromboembolism, compounds of the invention may be useful for maintaining patency of indwelling catheters. Examples of cardiogenic thromboembolism which may be treated or prevented with compounds of the invention include thromboembolic stroke (detached thrombus causing neurological affliction related to impaired cerebral blood supply), cardiogenic thromboembolism associated with atrial fibrillation (rapid, irregular twitching of upper heart chamber muscular fibrils), cardiogenic thromboembolism associated with prosthetic heart valves such as mechanical heart valves, and cardiogenic thromboembolism associated with heart disease. Examples of arterial thrombosis include unstable angina (severe constrictive pain in chest of coronary origin), myocardial infarction (heart muscle cell death resulting from insufficient blood supply), ischemic heart disease (local anemia due to obstruction (such as by arterial narrowing) of blood supply), reocclusion during or after percutaneous transluminal coronary angioplasty, restenosis after percutaneous transluminal coronary angioplasty, occlusion of coronary artery bypass grafts, and occlusive cerebrovascular disease. Also with regard to arterial thrombosis, compounds of the invention may be useful for maintaining patency in arteriovenous cannulas. Examples of atherosclerosis include arteriosclerosis. The compounds of the invention may also be kallikrein inhibitors and especially useful for treatment of hereditary angioedema. Examples of devices that come into contact with blood include vascular grafts, stents, orthopedic prosthesis, cardiac prosthesis, and extracorporeal circulation systems. The medicaments according to the invention can be administered by oral, inhalative, rectal or transdermal administration or by subcutaneous, intraarticular, intraperitoneal or intravenous injection. Oral administration is preferred. Coating of stents with compounds of the Formulas I, Ia, Ib, and Ic and other surfaces which come into contact with blood in the body is possible. The invention also relates to a process for the production of a medicament, which comprises bringing at least one compound of the Formulas I, Ia, Ib, and Ic into a suitable administration form using a pharmaceutically suitable and pharmaceutically acceptable carrier and optionally further suitable active substances, additives or auxiliaries. Suitable solid or galenical preparation forms are, for example, granules, powders, coated tablets, tablets, (micro)capsules, suppositories, syrups, juices, suspensions, emulsions, drops or injectable solutions and preparations having prolonged release of active substance, in whose preparation customary excipients such as vehicles, disintegrants, binders, coating agents, swelling agents, glidants or lubricants, flavorings, sweeteners and solubilizers are used. Frequently used auxiliaries which may be mentioned are magnesium carbonate, titanium dioxide, lactose, mannitol and other sugars, talc, lactose, gelatin, starch, cellulose and its derivatives, animal and plant oils such as cod liver oil, sunflower, peanut or sesame oil, polyethylene glycol and solvents such as, for example, sterile water and mono- or polyhydric alcohols such as glycerol. The dosage regimen utilizing the Factor XIa inhibitors or dual Factor XIa/plasma kallikrein inhibitors is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound or salt thereof employed. An ordinarily skilled physician or veterinarian can readily determine and prescribe the effective amount of the drug required to prevent, counter, or arrest the progress of the condition. Compounds of the Formula I, Formula Ia, Formula Ib and Formula Ic can be administered both as a monotherapy and in combination with other therapeutic agents, including antithrombotics (anticoagulants and platelet aggregation inhibitors), thrombolytics (plasminogen activators), other profibrinolytically active substances, hypotensives, blood sugar regulators, lipid-lowering agents and antiarrhythmics. The Factor XIa inhibitors or dual Factor XIa/plasma kallikrein inhibitors can also be co- administered with suitable anticoagulants, including, but not limited to, other Factor XIa inhibitors, thrombin inhibitors, thrombin receptor antagonists, factor VIIa inhibitors, factor Xa inhibitors, factor IXa inhibitors, factor XIIa inhibitors, adenosine diphosphate antiplatelet agents (e.g., P2Y12 antagonists), fibrinogen receptor antagonists (e.g. to treat or prevent unstable angina or to prevent reocclusion after angioplasty and restenosis), other anticoagulants such as aspirin, and thrombolytic agents such as plasminogen activators or streptokinase to achieve synergistic effects in the treatment of various vascular pathologies. Such anticoagulants include, for example, apixaban, dabigatran, cangrelor, ticagrelor, vorapaxar, clopidogrel, edoxaban, mipomersen, prasugrel, rivaroxaban, and semuloparin. For example, patients suffering from coronary artery disease, and patients subjected to angioplasty procedures, would benefit from coadministration of fibrinogen receptor antagonists and thrombin inhibitors. Factor XIa inhibitors may be administered first following thrombus formation, and tissue plasminogen activator or other plasminogen activator is administered thereafter. Alternatively or additionally, one or more additional pharmacologically active agents may be administered in combination with a compound of the invention. The additional active agent (or agents) is intended to mean a pharmaceutically active agent (or agents) that is active in the body, including pro-drugs that convert to pharmaceutically active form after administration, which is different from the compound of the invention, and also includes free-acid, free-base and pharmaceutically acceptable salts of said additional active agents when such forms are sold commercially or are otherwise chemically possible. Generally, any suitable additional active agent or agents, including but not limited to anti-hypertensive agents, additional diuretics, anti- atherosclerotic agents such as a lipid modifying compound, anti-diabetic agents and/or anti- obesity agents may be used in any combination with the compound of the invention in a single dosage formulation (a fixed dose drug combination), or may be administered to the patient in one or more separate dosage formulations which allows for concurrent or sequential administration of the active agents (co-administration of the separate active agents). Examples of additional active agents which may be employed include but are not limited to angiotensin converting enzyme inhibitors (e.g, alacepril, benazepril, captopril, ceronapril, cilazapril, delapril, enalapril, enalaprilat, fosinopril, imidapril, lisinopril, moveltipril, perindopril, quinapril, ramipril, spirapril, temocapril, or trandolapril); angiotensin II receptor antagonists also known as angiotensin receptor blockers or ARBs, which may be in free-base, free-acid, salt or pro-drug form, such as azilsartan, e.g., azilsartan medoxomil potassium (EDARBI ^), candesartan, e.g., candesartan cilexetil (ATACAND ^), eprosartan, e.g., eprosartan mesylate (TEVETAN ^), irbesartan (AVAPRO ^), losartan, e.g., losartan potassium (COZAAR ^), olmesartan, e.g, olmesartan medoximil (BENICAR ^), telmisartan (MICARDIS ^), valsartan (DIOVAN ^), and any of these drugs used in combination with a thiazide-like diuretic such as hydrochlorothiazide (e.g., HYZAAR ^, DIOVAN HCT ^, ATACAND HCT ^), etc.); potassium sparing diuretics such as amiloride HCl, spironolactone, epleranone, triamterene, each with or without HCTZ; neutral endopeptidase inhibitors (e.g., thiorphan and phosphoramidon); aldosterone antagonists; aldosterone synthase inhibitors; renin inhibitors; enalkrein; RO 42-5892; A 65317; CP 80794; ES 1005; ES 8891; SQ 34017; aliskiren (2(S),4(S),5(S),7(S)-N-(2-carbamoyl-2-methylpropyl)-5- amino-4-hydroxy-2,7-diisopropyl-8-[4-methoxy-3-(3-methoxypro poxy)-phenyl]-octanamid hemifumarate) SPP600, SPP630 and SPP635); endothelin receptor antagonists; vasodilators (e.g. nitroprusside); calcium channel blockers (e.g., amlodipine, nifedipine, verapamil, diltiazem, felodipine, gallopamil, niludipine, nimodipine, nicardipine); potassium channel activators (e.g., nicorandil, pinacidil, cromakalim, minoxidil, aprilkalim, loprazolam); sympatholitics; beta- adrenergic blocking drugs (e.g., acebutolol, atenolol, betaxolol, bisoprolol, carvedilol, metoprolol, metoprolol tartate, nadolol, propranolol, sotalol, timolol); alpha adrenergic blocking drugs (e.g., doxazosin, prazosin or alpha methyldopa); central alpha adrenergic agonists; peripheral vasodilators (e.g. hydralazine); lipid lowering agents, e.g., HMG-CoA reductase inhibitors such as simvastatin and lovastatin which are marketed as ZOCOR® and MEVACOR® in lactone pro-drug form and function as inhibitors after administration, and pharmaceutically acceptable salts of dihydroxy open ring acid HMG-CoA reductase inhibitors such as atorvastatin (particularly the calcium salt sold in LIPITOR®), rosuvastatin (particularly the calcium salt sold in CRESTOR®), pravastatin (particularly the sodium salt sold in PRAVACHOL®), and fluvastatin (particularly the sodium salt sold in LESCOL®); a cholesterol absorption inhibitor such as ezetimibe (ZETIA®), and ezetimibe in combination with any other lipid lowering agents such as the HMG-CoA reductase inhibitors noted above and particularly with simvastatin (VYTORIN®) or with atorvastatin calcium; niacin in immediate-release or controlled release forms, and particularly niacin in combination with a DP antagonist such as laropiprant and/or with an HMG-CoA reductase inhibitor; niacin receptor agonists such as acipimox and acifran, as well as niacin receptor partial agonists; metabolic altering agents including insulin sensitizing agents and related compounds for the treatment of diabetes such as biguanides (e.g., metformin), meglitinides (e.g., repaglinide, nateglinide), sulfonylureas (e.g., chlorpropamide, glimepiride, glipizide, glyburide, tolazamide, tolbutamide), thiazolidinediones also referred to as glitazones (e.g., pioglitazone, rosiglitazone), alpha glucosidase inhibitors (e.g., acarbose, miglitol), dipeptidyl peptidase inhibitors, (e.g., sitagliptin (JANUVIA ^), alogliptin, vildagliptin, saxagliptin, linagliptin, dutogliptin, gemigliptin), ergot alkaloids (e.g., bromocriptine), combination medications such as JANUMET ^ (sitagliptin with metformin), and injectable diabetes medications such as exenatide and pramlintide acetate; inhibitors of glucose uptake, such as sodium-glucose transporter (SGLT) inhibitors and its various isoforms, such as SGLT-1, SGLT-2 (e.g., ASP-1941, TS-071, BI-10773, tofogliflozin, LX-4211, canagliflozin, dapagliflozin, ertugliflozin, ipragliflozin, remogliflozin and sotagliflozin), and SGLT-3; or with other drugs beneficial for the prevention or the treatment of the above-mentioned diseases including but not limited to diazoxide; and including the free-acid, free-base, and pharmaceutically acceptable salt forms, pro-drug forms, e.g., esters, and salts of pro-drugs of the above medicinal agents, where chemically possible. Trademark names of pharmaceutical drugs noted above are provided for exemplification of the marketed form of the active agent(s); such pharmaceutical drugs could be used in a separate dosage form for concurrent or sequential administration with a compound of the invention, or the active agent(s) therein could be used in a fixed dose drug combination including a compound of the invention. Typical doses of Factor XIa inhibitors or Factor XIa/plasma kallikrein inhibitors of the invention in combination with other suitable anti-platelet agents, anticoagulation agents, or thrombolytic agents may be the same as those doses of Factor XIa inhibitors administered without coadministration of additional anti-platelet agents, anticoagulation agents, or thrombolytic agents, or may be substantially less that those doses of thrombin inhibitors administered without coadministration of additional anti-platelet agents, anticoagulation agents, or thrombolytic agents, depending on a patient’s therapeutic needs. The compounds are administered to a mammal in a therapeutically effective amount. By “therapeutically effective amount” it is meant an amount of a compound of the present invention that, when administered alone or in combination with an additional therapeutic agent to a mammal, is effective to treat (i.e., prevent, inhibit or ameliorate) the thromboembolic and/or inflammatory disease condition or treat the progression of the disease in a host. The compounds of the invention are preferably administered alone to a mammal in a therapeutically effective amount. However, the compounds of the invention can also be administered in combination with an additional therapeutic agent, as defined below, to a mammal in a therapeutically effective amount. When administered in a combination, the combination of compounds is preferably, but not necessarily, a synergistic combination. Synergy, as described for example by Chou and Talalay, Adv. Enzyme Regul.1984, 22, 27-55, occurs when the effect (in this case, inhibition of the desired target) of the compounds when administered in combination is greater than the additive effect of each of the compounds when administered individually as a single agent. In general, a synergistic effect is most clearly demonstrated at suboptimal concentrations of the compounds. Synergy can be in terms of lower cytotoxicity, increased anticoagulant effect, or some other beneficial effect of the combination compared with the individual components. By “administered in combination” or “combination therapy” it is meant that the compound of the present invention and one or more additional therapeutic agents are administered concurrently to the mammal being treated. When administered in combination each component may be administered at the same time or sequentially in any order at different points in time. Thus, each component may be administered separately but sufficiently closely in time so as to provide the desired therapeutic effect. The present invention is not limited in scope by the specific embodiments disclosed in the examples which are intended as illustrations of a few aspects of the invention and any embodiments that are functionally equivalent are within the scope of this invention. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the relevant art and are intended to fall within the scope of the appended claims. GENERAL METHODS Several methods for preparing the compounds of this invention are illustrated in the following Schemes and Examples. Starting materials and the requisite intermediates are in some cases commercially available, or can be prepared according to literature procedures or as illustrated herein. The compounds of this invention may be prepared by employing reactions as shown in the following schemes, in addition to other standard manipulations that are known in the literature or exemplified in the experimental procedures. Substituent numbering as shown in the schemes does not necessarily correlate to that used in the claims and often, for clarity, a single substituent is shown attached to the compound where multiple substituents are allowed under the definitions hereinabove. Reactions used to generate the compounds of this invention are carried out by employing reactions as shown in the schemes and examples herein, in addition to other standard manipulations such as ester hydrolysis, cleavage of protecting groups, etc., as may be known in the literature or exemplified in the experimental procedures. Starting materials are made according to procedures known in the art or as illustrated herein. The compounds of the present invention can be prepared in a variety of fashions. In some cases the final product may be further modified, for example, by manipulation of substituents. These manipulations may include, but are not limited to, reduction, oxidation, alkylation, acylation, and hydrolysis reactions which are commonly known to those skilled in the art. In some cases the order of carrying out the foregoing reaction schemes may be varied to facilitate the reaction or to avoid unwanted reaction products. Because the schemes are an illustration, the invention should not be construed as being limited by the chemical reactions and conditions expressed. The preparation of the various starting materials used herein is well within the skill of a person versed in the art. The following examples are provided so that the invention might be more fully understood. These examples are illustrative only and should not be construed as limiting the invention in any way. Absolute stereochemistry of separate stereoisomers in the examples and intermediates are not determined unless stated otherwise in an example or explicitly in the nomenclature. Abbreviations are used and defined as follows: S-(+)-DTBM-Segphos ® is (S)-(+)-5,5′-Bis[di(3,5-di-tert-butyl-4- methoxyphenyl)phosphino]-4,4′-bi-1,3-benzodioxole; xphos-G2 is chloro(2- dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1′- biphenyl)[2-(2′-amino-1,1′- biphenyl)]palladium(II); cataCXium® A Pd G2 is chloro[(di(1-adamantyl)-N-butylphosphine)-2- (2-aminobiphenyl)]palladium (II); which are available from Sigma Aldrich. Ac acetyl Ar aryl aq Aqueous Bn benzyl Boc tert-butyloxycarbonyl t-Bu tert-butyl 18-Crown-6 1,4,7,10,13,16-hexaoxacyclooctadecane DAST diethylaminosulfurtrifluoride DCE dichloroethane DCM dichloromethane DEA diethylamine DIAD diethyl azodicarboxylate DIEA or DIPEA N,N-diisopropylethylamine DMA Dimethylacetamide DME 1,2-dimethoxyethane DMF Dimethylformamide DMPU 1,3-dimethyl-Tetrahydropyrimidin-2 (1H)-one DMSO dimethyl sulfoxide DPPA diphenylphosphorylazide EDC or EDCI N-(3-Dimethylaminopropyl)-N’-ethylcarbodiimide ESMS Electrospray mass spectroscopy Et ethyl EtOAc ethyl acetate Et 3 N Triethylamine h hour(s) HATU 1-[bis(dimethylamino)methylene]- 1H-1,2,3-triazolo[4,5-b]pyridinium-3- oxidhexafluorophosphate Hex hexane HOBt hydroxybenzotriazole hydrate HPLC high performance liquid chromatography IPA isopropanol LCMS Liquid chromatography mass spectrometry LDA lithium diisopropylamide LiHMDS lithium bis(trimethylsilyl)amide m-CPBA m-chloroperoxybenzoic acid Me methyl MeCN acetonitrile MeOH Methanol mg milligrams min minute(s) µL microliters mL milliliters mmol millimoles NMR nuclear magnetic resonance NIS N-IodosuccinimideMS mass spectrometry MTBE methyl tert-butyl ether Pd/C Palladium on Carbon Ph phenyl Py pyridine Pd(dppf)Cl2 [1,1'-Bis(diphenylphosphino)ferrocene]dichloropalladium(II) PPh 3 Triphenylphosphine TBAI tetrabutylammonium iodide TEA Triethylamine TFA trifluoroacetic acid TFAA trifluoroacetic anhydride THF tetrahydrofuran TLC thin-layer chromatography TMEDA tetramethylethylenediamine TMS tetramethylsilane rt room temperature SEM 2-(trimethylsilyl)ethoxy)methyl SFC Supercritical Fluid Chromatography NMR spectra were measured on VARIAN NMR Systems (400, 500 or 600 MHz) and BRUKER NMR Systems (400, 500 MHz). Chemical shifts are reported in ppm downfield and up field from TMS and referenced to either internal TMS or solvent resonances ( 1 H NMR: δ 7.27 for CDCl3, δ 2.50 for (CD3)(CHD2)SO, and 13 C NMR: δ 77.02 for CDCl3, δ 39.51 for (CD3)2SO. Coupling constants (J) are expressed in hertz (Hz), and spin multiplicities are given as s (singlet), d (doublet), dd (double doublet), t (triplet), m (multiplet), and br (broad). Chiral resolutions were performed on either Waters Thar 80 SFC or Berger MG II, Thar 80 SFC, Waters 80 SFC, Sepiatec 100 SFC, Waters 200 SFC, Thar 350 SFC, Berger MG III preparative SFC systems. LCMS data were recorded on SHIMADAZU LCMS-2020, SHIMADAZU LCMS-2010EV, or Agilent 1100 series LCMS, or Waters Acquity LCMS instruments using C18 columns employing a MeCN gradient in water containing 0.02 to 0.1% TFA. UV detections were at 220 and/or 254 nm and ESI ionization was used for MS detection. When chiral resolution was achieved by chromatography using chiral columns, the chiral columns used for SFC chiral resolutions are as indicated. Also, TLC is thin layer chromatography; UV is ultraviolet; W is watts; wt. % is percentage by weight; x g is times gravity; α D is the specific rotation of polarized light at 589 nm; °C is degrees Celsius; % w/v is percentage in weight of the former agent relative to the volume of the latter agent; Hz is hertz; cpm is counts per minute; ^ H is chemical shift; d is doublet; dd is doublet of doublets; MHz is megahertz; MS is mass spectrum, and a mass spectrum obtained by ESMS may be denoted herein by “LCMS”; m/z is mass to charge ratio; n is normal; N is normal; nm is nanometer; nM is nanomolar. “Human FXIa Ki (nM)” is Human Factor XIa Ki (nM). SCHEMES Scheme 1 illustrates one synthetic sequence for the preparation of the compounds of this invention. Macrocycles of general formula 8 can be prepared as described in Scheme 1. Preparation of intermediate macrocyclic core 2 is described in the intermediates section in detail and general methods related to such systems are described in International Patent Publication WO 2017/074832. Compounds such as 1 where Y is a suitably reactive halide can be coupled with an appropriate boronic acid 2 or boronic ester using a Suzuki reaction. Methyl or ethyl ester can be introduced on the imidazole of compound 3 via iodination followed by a carbonation reaction in the presence of methanol or ethanol. Subsequent reduction of the olefin of compound 5 to compound 6 can be carried out through an asymmetric hydrogenation process in which enantiomeric enrichment can be obtained through the use of Rh catalyst/S-(+)-DTBM-segphos® reagent system. Formation of the N-oxide in 7 can be achieved through oxidation of the pyridine of 6 with an oxidant such as oxone or peracetic acid. Finally, removal of the SEM protecting group of 7 can be carried out through treatment with TFA to afford compounds of general formula 8. SCHEME 1 INTERMEDIATE 1 methyl (3-amino-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phen yl)carbamate Step 1: (4-bromo-3-nitrophenyl To 4-bromo-3-nitroaniline (500.0 g, 2.30 mol, 1 eq) in 2-Me-THF (2.5 L) was added DIEA (655.0 g, 5.07 mol, 883.0 mL, 2.2 eq) in one portion. Then methyl carbonochloridate (261.0 g, 2.76 mol, 214.0 mL, 1.2 eq) was added to the reaction over 0.5 h while maintaining the temperature between 20 - 48 °C. The reaction was heated to 50 °C and aged for 1 h. It was then cooled to 20 °C. The reaction was poured into water and extracted with EtOAc. The combined organic portions were washed with brine, and dried over anhydrous sodium sulfate. The mixture was filtered and then concentrated under vacuum. The product was slurried with petroleum ether. The product was then collected by filtration and dried under vacuum to afford methyl (4-bromo- 3-nitrophenyl)carbamate. 1 HNMR: (400 MHz, CDCl 3 ) 8.03 (s, 1 H), 7.64 (d, J = 8.8 Hz, 1 H), 7.45-7.47 (m, 1 H), 6.99(s, 1 H), 3.81 (s, 3 H). Step 2: methyl (3-nitro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phen yl)carbamate To a 3L three necked flask equipped with magnetic stirrer was added methyl (4-bromo-3- nitrophenyl)carbamate (450.0 g, 1.64 mol, 1.0 eq) in dioxane (2.7 L), 4,4,4',4',5,5,5',5'- octamethyl-2,2'-bi(1,3,2-dioxaborolane (456.9 g, 1.80 mol, 1.1 eq), KOAc (353.2 g, 3.60 mol, 2.2 eq) and Pd(dppf)Cl 2 (59.8 g, 0.08 mol, 0.05 eq) 25 °C. The reaction was purged with N 2 three times and then heated to 80 o C and aged for 7 h. The reaction was concentrated under vacuum. The residue was suspended in MTBE. The mixture was filtered, and the filtrate was concentrated under vacuum. The residue was purified by column chromatography (SiO2, Petroleum ether: Ethyl acetate = 50 : 1 to 4 : 1). The fractions containing the desired product were combined and concentrated under vacuum to afford methyl (3-nitro-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan- 2-yl)phenyl)carbamate. 1 HNMR: 400 MHz CDCl 3 δ: 8.18 (s, 1 H), 7.69-7.71 (d, J = 7.2 Hz, 1 H), 7.46-7.48 (d, J = 8.0 Hz, 1 H), 6.96 (s, 1 H), 3.80 (s, 3 H), 1.42 (s, 12 H) Step 3: methyl (3-amino-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phen yl)carbamate To a dried hydrogenation bottle was added Pd/C (3.0 g, 10.0% purity), MeOH (1.5 L), and methyl (3-nitro-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)phenyl)carbamate (60.0 g, 0.18 mol, 1.0 eq). The vessel was degassed under vacuum and purged with H2 three times. The reaction was then aged for 2 h under H2 (20 Psi) at 25 °C. The reaction mixture was filtered on celite and the filtrate concentrated under vacuum to afford methyl (3-amino-4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)carbamate. LCMS (ES, m/z): 292.9 [M+H] + . 1 HNMR: (400 MHz, CDCl 3 ) 7.51 (d, J = 8.0 Hz, 1H), 6.91 (s, 1H), 6.55 (s, 1H), 6.46-6.49 (m, 1H), 4.80 (s, 2H), 3.76 (s, 3H), 1.33 (s, 12H) INTERMEDIATE 2 (5-bromopyridin-2-yl)(4-iodo-1-((2-(trimethylsilyl)ethoxy)me thyl)-1H-imidazol-2-yl)methanone Step 1: 4-iodo-1-((2-(trimethylsilyl) midazole To a suspension of NaH (4.21 g, 105 mmol, 60%wt) in DMF (200 mL) at 0°C was added 4-iodo- 1H-imidazole (17 g, 88 mmol) in small portions. It was stirred for 1 h and SEM-Cl (16.07 g, 96 mmol) was added to the reaction. The mixture was stirred at rt for 12 h and poured into ice-water (200 mL). The mixture was extracted with EtOAc (100 mL x 3). The combined organic layers were washed with water (3x50 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by flash column chromatography on silica gel (eluting with petroleum ether: EtOAc = 10:1 to 3:1 v/v) to afford the title compound. MS (ES + ) m/z: 325 (M+H). Step 2: 5-bromo-N-methoxy-N-methylpicolinamide To a solution of 5-bromopicolinic acid (15 g, 74.3 mmol) in DCM (200 mL) at 0°C was added EDC (21.35 g, 111 mmol), N,O-dimethylhydroxylamine hydrochloride (10.86 g, 111 mmol) and pyridine (15.01 mL, 186 mmol). The mixture was stirred at rt for 16 h and concentrated under reduced pressure. The residue was diluted with EtOAc (250 mL) and washed with 1N HCl (50 mL) and then brine. The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure to give the title compound, which was used in next step without further purification. MS (ES + ) m/z: 245, 247 (M+H). Step 3: (5-bromopyridin-2-yl)(4-iodo-1-((2-(trimethylsilyl)ethoxy)me thyl)-1H-imidazol-2- yl)methanone To a solution of 4-iodo-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-imidazole (15.88 g, 49.0 mmol) in THF (100 mL) at -78°C was added dropwise a solution of LDA (26.5 mL, 53.0 mmol, 2 M in THF). The reaction was stirred for 1 h and a solution of 5-bromo-N-methoxy-N- methylpicolinamide (10 g, 40.8 mmol) in THF (20 mL) was added dropwise. The resulting mixture was stirred at -78°C for 1 h and it was quenched with saturated aqueous ammonium chloride (10 mL). The mixture was extracted with EtOAc (150 mL). The organic layer was washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by flash column chromatography on silica gel (eluting with petroleum ether: EtOAc = 10:1 v/v) to give the title compound. 1 H NMR (400 MHz, CDCl3): δ 8.85 (s, 1H), 8.23 (m, 1H), 8.02 (m, 1H), 7.52 (s, 1H), 5.81 (s, 2H),3.65 (m, 2H), 0.96 (m, 2H), -0.01 (s, 9H). INTERMEDIATE 3 methyl (3-amino-4-(2-(5-bromopicolinoyl)-1-((2-(trimethylsilyl)etho xy)methyl)-1H-imidazol-4- yl)phenyl)carbamate To a mixture of Intermedi , , .3 g, 87 mmol), tetrakis(triphenylphosphine)palladium(0) (4.55 g, 3.94 mmol), DMF (354 mL) and potassium phosphate tribasic (50.1 g, 236 mmol) was stirred at 60°C for 3.5 h. It was cooled to rt and most solvent was removed under reduced pressure. The residue was diluted with ethyl acetate (200 mL) and washed with water (3x100 mL), then brine (100 mL). The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by flash column chromatography on silica gel (eluting with 0-60% EtOAc/hexane, gradient) to give the title compound. MS (ES + ) m/z: 546, 548 [M+H]. INTERMEDIATE 4 5-(benzo[d]thiazol-2-ylsulfonyl)-2,2-dimethylpentanoic acid Step 1: methyl 5-(benzo[d]thiazol-2-ylthio)pentanoate Benzo[d]thiazole-2-thiol (220 g, 1.32 mol) and methyl 5-bromopentanoate (282.2 g, 1.45 mol) were combined with acetone (2.0 L) in a three-necked round bottom flask equipped with a magnetic stirrer at 20 °C. To this was added K2CO3 (181.8 g, 1.32 mol) in one portion at 20 °C. The reaction was stirred at 20 °C for 12 hours. The reaction was filtered, and concentrated under vacuum to provide methyl 5-(benzo[d]thiazol-2-ylthio)pentanoate that was used for the next step without further purification. Step 2: methyl 5-(benzo[d]thiazol-2-ylthio)-2-methylpentanoate A three-necked round bottom flask equipped with magnetic stirrer was charged with methyl 5- (benzo[d]thiazol-2-ylthio)pentanoate (300.0 g, 1.07 mol) and CH 3 I (151.3 g, 1.07 mol) in dry THF (1.5 L) at 20 °C. The reaction was cooled to -70 °C before addition of LiHMDS (2 M in THF, 746.3 ml) to the mixture over 2 hours while maintaining the temperature below -60 °C. The reaction aged at -60 °C for 2 hours. The reaction mixture was poured into saturated aqueous NH4Cl (8.0 L) and extracted with ethyl acetate. The organic phase was washed with brine and then concentrated under vacuum. The residue was purified by silica gel chromatography. The fractions containing the desired product were combined and concentrated under vacuum to afford methyl 5-(benzo[d]thiazol-2-ylthio)pentanoate. Step 3: methyl 5-(benzo[d]thiazol-2-ylthio)-2,2-dimethylpentanoate A three-necked round bottom flask equipped with a magnetic stirrer was charged with methyl 5- (benzo[d]thiazol-2-ylthio)-2-methylpentanoate (200.0 g, 0.68 mol) and CH 3 I (283.5 g, 2.0 mol) in dry THF (1.0 L) at 20 °C. The reaction was cooled to -70 °C before addition of LDA (2 M, 0.88 L) to the mixture over 2 hours while maintaining the temperature below -60 °C. The reaction aged at -60 °C for 2 hours. The reaction mixture was poured into saturated aqueous NH 4 Cl (4.0 L) and extracted with ethyl acetate twice (5.0 L, 4.0 L). The organic phase was washed with brine (5.0 L) and then concentrated under vacuum to afford methyl 5- (benzo[d]thiazol-2-ylthio)- 2,2-dimethylpentanoate which was used for the next step without further purification. Step 4: 5-(benzo[d]thiazol-2-ylthio)-2,2-dimethylpentanoic acid A three-necked round bottom flask equipped with a magnetic stirrer was charged with methyl 5- (benzo[d]thiazol-2-ylthio)-2,2-dimethylpentanoate (200.0 g, 0.64 mol) in 2- methyltetrahydrofuran (0.2 L) at 20 °C. Aq. NaOH (3.75 M, 1.0 L) was added in one portion to the mixture at 20 °C. The mixture aged at 80 °C reflux for 15 hours. After cooling to room temperate the reaction was extracted with ethyl acetate, and the aqueous phase was adjusted to pH = 2 with aq. HCl (4M, 1.0 L) to allow precipitation of solid. The solid was filtered, and the filter cake was washed with petroleum ether to afford 5-(benzo[d]thiazol-2-ylthio)-2,2- dimethylpentanoic acid that was used for the next step without further purification. Step 5: 5-(benzo[d]thiazol-2-ylsulfonyl)-2,2-dimethylpentanoic acid A three-necked round bottom flask equipped with a magnetic stirrer was charged with 5- (benzo[d]thiazol-2-ylthio)-2,2- dimethylpentanoic acid (730.0 g, 2.48 mol) in EtOH (3600 ml) at 20 °C. To this was added ammonium molybdate (73 g) in one portion at 20 °C. H2O2 (1403 g, 12.4 mol, 30% purity) was added to the mixture at 20 °C over 0.2 hr. The reaction aged at ~20 - 30 °C for 2 hours at which point water (15 L) was added at 20 °C. The mixture was extracted with ethyl acetate. The organic phase washed with saturated aqueous Na 2 SO 3 (4.0 L x 3), and then dried over Na2SO4. The mixture was filtered, and the filtrate was concentrated under vacuum. The product was triturated with petroleum ether: ethyl acetate = 1:1 to afford 5- (benzo[d]thiazol-2-ylsulfonyl)-2,2- dimethylpentanoic acid. LCMS (ES, m/z): 328.0 [M+H] + . INTERMEDIATE 5 methyl ((1 2 Z,8Z)-9-(5-bromopyridin-2-yl)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -en- 2 4 -yl)carbamate Step 1: 5-(benzo[d]thiazo l chloride To a mixture of 5-(benzo[d]thiazol-2-ylsulfonyl)-2,2-dimethylpentanoic acid (200 g, 0.61 mol) in acetonitrile (1000 ml) at 25℃ was added sulfurous dichloride (109 g, 0.12 mol) over 10 minutes. The reaction mixture was stirred at 25℃ for 2 h and then it was concentrated under vacuum to afford 5-(benzo[d]thiazol-2-ylsulfonyl)-2,2-dimethylpentanoyl chloride which was used without further purification. Step 2: methyl (3-(5-(benzo[d]thiazol-2-ylsulfonyl)-2,2-dimethylpentanamido )-4-(2-(5- bromopicolinoyl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-imi dazol-4-yl)phenyl)carbamate To a mixture of methyl (3-amino-4-(2-(5-bromopicolinoyl)-1-((2-(trimethylsilyl)etho xy)methyl)- 1H-imidazol-4-yl)phenyl)carbamate (276 g, 0.51 mol) in DCM (850 ml) at 25℃ was added a solution of 5-(benzo[d]thiazol-2-ylsulfonyl)-2,2-dimethylpentanoyl chloride (180 g, 0.52 mol) in DCM (1100 ml) over 5 minutes, followed by triethylamine (144.9 ml, 1.04 mol) over 10 minutes. The reaction mixture was stirred at 25℃ for 2 h. Two reactions were combined for workup. The reaction mixtures were poured into water (4.4 L). The organic phase was washed with 1 N HCl (4.4 L x 2), saturated NaHCO3 (4.4 L), brine (4.4 L), and concentrated in vacuum to dryness to afford the title compound which was used without further purification. LCMS (ES, m/z): 855.2, 857.3 [M+H] + . Step 3: methyl ((1 2 Z,8Z)-9-(5-bromopyridin-2-yl)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -en- 2 4 -yl)carbamate To a solution of LiHMDS (1.0 M, 818 ml, 0.82 mol) in THF (1250 ml) at -20℃ was added a solution of methyl (3-(5-(benzo[d]thiazol-2-ylsulfonyl)-2,2-dimethylpentanamido )-4-(2-(5- bromopicolinoyl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-imi dazol-4-yl)phenyl)carbamate (250 g, 0.20 mol) in THF (1250 mL) dropwise. The reaction mixture was stirred at -30℃ to -20℃ for 2 h. Four reactions were combined for workup. The reaction mixtures were poured into saturated NH4Cl ( 10 L) at 0~10°C. The organic phase was washed with 5% NaOH (10L, 9L and 8L). The aqueous phase was extracted with ethyl acetate (5 L, 3 L). The combined organic phases were washed with brine (5 L, 4 L), dried over anhydrous sodium sulfate and filtered. After concentration under vacuum, the product was was purified by re-crystallization from MeOH to afford the title compound. LCMS (ES, m/z): 640.2, 642.2 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ): δ 8.71 (d, J = 2.4 Hz, 1H), 7.96 - 7.89 (m, 2H), 7.64 (br s, 1H), 7.54 (d, J = 8.4 Hz, 1H), 7.40 (d, J = 8.6 Hz, 1H), 7.34 (s, 1H), 6.94 (br dd, J = 6.4, 10.4 Hz, 1H), 6.81 (br s, 1H), 5.09 (d, J = 10.8 Hz, 1H), 4.85 (d, J = 10.6 Hz, 1H), 3.85 (s, 3H), 3.37 - 3.24 (m, 2H), 2.52 - 2.26 (m, 3H), 1.86 (dt, J = 6.0, 13.8 Hz, 1H), 1.42 (s, 3H), 1.27 (s, 3H), 0.83 (t, J = 8.3 Hz, 2H), 0.00 (s, 9H). INTERMEDIATE 6 (6-((1 2 Z,8Z)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -en- 9-yl)pyridin-3-yl)boronic acid To a stirred mixture of m , 5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -en- 2 4 -yl)carbamate (1.6 g, 2.5 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (0.761 g, 3.00 mmol), potassium acetate (0.735 g, 7.49 mmol) in dioxane (20 mL) was added xphos-G2, chloro(2-dicyclohexylphosphino-2′,4′,6′-triisopropyl-1 ,1′- biphenyl)[2-(2′-amino-1,1′- biphenyl)]palladium(II) (0.197 g, 0.250 mmol) under N 2 at 25 °C and the mixture was stirred at 100 °C for 1 h under N2. LCMS showed the reaction was completed. The solvent was removed under reduced pressure. Water (20 mL) was added and the mixture was extracted with ethyl acetate (20 mL x 2). The combined organic fractions were washed with brine (20 mL x 2), dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure to give (6- ((1 2 Z,8Z)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -en- 9-yl)pyridin-3-yl)boronic acid which was used in next step without further purification. LCMS (ES, m/z): 606.2 [M+H] + . EXAMPLE 1 (R,Z)-2-(1 5 -(ethoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 H-3-aza- 1(42)-imidazola-2(1,2)-benzenacyclononaphane-9-yl)-5-(5-fluo ro-2-(4-(trifluoromethyl)-1H- pyrazol-1-yl)phenyl)pyridine 1-oxide Step 1: methyl ((1 2 Z,8Z)-9 pyrazol-1-yl)phenyl)pyridin- 2-yl)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola- 2(1,2)-benzenacyclononaphan-8-en-2 4 -yl)carbamate To a stirred mixture of methyl ((1 2 Z,8Z)-9-(5-bromopyridin-2-yl)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -en- 2 4 -yl)carbamate (4.0 g, 6.2 mmol), xphos-G2 (0.393 g, 0.499 mmol), 1-(4-fluoro-2-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-4-(trifluorometh yl)-1H-pyrazole (2.67 g, 7.49 mmol) in THF (60 mL) was added 2M potassium phosphate (9.37 mL, 18.7 mmol) under N 2 at 25 °C and the mixture was stirred at 80 °C for 16 h under N2. Water (50 mL) was added and the mixture was extracted with ethyl acetate (80 mL x 2). The combined organic fractions were washed with brine (80 mL), dried over Na2SO4, filtered and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (0-60% EtOAc/petroleum ether) to give methyl ((1 2 Z,8Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H- pyrazol-1-yl)phenyl)pyridin-2-yl)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H- 3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8-en-2 4 -yl)carbamate. LCMS (ES, m/z): 790.4 (M+H) + . Step 2: methyl ((1 2 Z,8Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol -1-yl)phenyl)pyridin- 2-yl)-1 5 -iodo-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)- imidazola-2(1,2)-benzenacyclononaphan-8-en-2 4 -yl)carbamate To a stirred mixture of methyl ((1 2 Z,8Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol -1- yl)phenyl)pyridin-2-yl)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8-en-2 4 -yl)carbamate (4.4 g, 5.6 mmol) in CH3COOH (25 mL) was added 1-iodopyrrolidine-2,5-dione (1.44 g, 6.41 mmol) at 25 °C and the reaction mixture was stirred at 40 °C for 16 h. The reaction mixture was carefully added dropwise to Na 2 CO 3 solution at 0 °C until PH=7. The mixture was extracted with ethyl acetate (200 mL x 2). The combined organic fractions were washed with brine (200 mL x 2), dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (0-50% EtOAc/petroleum ether) to give methyl ((1 2 Z,8Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol -1-yl)phenyl)pyridin-2-yl)-1 5 -iodo- 5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphan-8-en-2 4 -yl)carbamate. LCMS (ES, m/z): 916.2 (M+H) + . Step 3: ethyl (1 2 Z,8Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol -1-yl)phenyl)pyridin-2- yl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H- 3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8-ene-1 5 -carboxylate To a mixture of methyl ((1 2 Z,8Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol -1- yl)phenyl)pyridin-2-yl)-1 5 -iodo-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3- aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8-en-2 4 -yl)carbamate (8.50 g, 9.28 mmol) in EtOH (60 mL) and DMSO (60 mL) was added PdCl2(dppf) (2.037 g, 2.78 mmol) and TEA (10.35 mL, 74.3 mmol) and the mixture was stirred at 70 °C under CO atmosphere at 50 psi for 16 h. The reaction mixture was concentrated. The mixture was cooled, diluted with ethyl acetate (200 mL), washed with brine (200 mL x 4), dried over Na2SO4, filtered and the solvent was evaporated under reduced pressure. The residue was purified by chromatography (C18; Eluent of 0~50% MeCN/H2O) to give ethyl (1 2 Z,8Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol -1- yl)phenyl)pyridin-2-yl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -ene- 1 5 -carboxylate. LCMS (ES, m/z): 862.5 (M+H) + . Step 4: ethyl (R,Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl) phenyl)pyridin-2-yl)- 2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3- aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-1 5 -carboxylate To a stirred mixture of bis(norbornadiene)rhodium(I) tetrafluoroborate (0.195 g, 0.522 mmol) in 1,2-dichloroethane (15 mL) was added (S)-(+)-5,5'-bis[di(3,5-di-tert-butyl-4- methoxyphenyl)phosphino]-4,4'-bi-1,3-benzodioxole (0.616 g, 0.522 mmol) at 25 °C in a glovebox and the mixture was stirred at 25 °C for 1 h under N2 atmosphere. To a stirred mixture of ethyl (1 2 Z,8Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol -1-yl)phenyl)pyridin-2-yl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8-ene-1 5 -carboxylate (4.5 g, 5.2 mmol) in trifluoroethanol (80 mL) was added the above mixture at 25 °C and the reaction mixture was stirred at 50 °C for 48 h under H2 in 50 psi. The reaction mixture was cooled and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (0-60% EtOAc/petroleum ether) to give ethyl (R,Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)- 1H-pyrazol-1-yl)phenyl)pyridin-2-yl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 1 5 - carboxylate. LCMS (ES, m/z): 864.3 (M+H) + . Step 5: (R, Z)-2-(1 5 -(ethoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 9-yl)- 5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl)phenyl)py ridine 1-oxide The mixture of ethyl (R,Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl) phenyl)pyridin- 2-yl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)- 1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 1 5 -carboxylate (2.0 g, 2.3 mmol) in CH3COOOH (30 mL) was stirred at 30 °C for 3 h under N2 atmosphere. The mixture was added to a solution of aqueous sodium hydrogen carbonate (saturated, 200 mL) and aqueous Na 2 SO 3 (saturated, 500 mL) at 0 °C and the mixture was extracted with ethyl acetate (500 mL x 2). The combined organic fractions were dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure to give (R, Z)-2-(1 5 -(ethoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5- dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphane-9-yl)-5-(5-fluoro-2-(4-(trifluoromethy l)-1H-pyrazol-1- yl)phenyl)pyridine 1-oxide which was used in next step without further purification. LCMS (ES, m/z): 880.3 (M+H) + . Step 6: (R, Z)-2-(1 5 -(ethoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 H-3- aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-9-yl)-5-(5 -fluoro-2-(4-(trifluoromethyl)- 1H-pyrazol-1-yl)phenyl)pyridine 1-oxide To a stirred mixture of (R, Z)-2-(1 5 -(ethoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5- dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphane-9-yl)-5-(5-fluoro-2-(4-(trifluoromethy l)-1H-pyrazol-1- yl)phenyl)pyridine 1-oxide (2.0 g, 2.3 mmol), 2-amino-3-mercaptopropanoic acid (0.826 g, 6.82 mmol) in DCM (10 mL) was added 2,2,2-trifluoroacetic acid (20 mL, 2.3 mmol) and the mixture was stirred at 40 °C for 3 h under N2 atmosphere. The volatiles were evaporated under reduced pressure. The residue was purified by column chromatography on silica (0~65% CH2Cl2/EtOAc). It was further purified by chiral SFC (DAICEL CHIRALCEL OD, 40% 0.1% NH 3 H 2 O EtOH/CO 2 ) to give (R, Z)-2-(1 5 -(ethoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5- dimethyl-4-oxo-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 9-yl)-5-(5-fluoro-2- (4-(trifluoromethyl)-1H-pyrazol-1-yl)phenyl)pyridine 1-oxide. LCMS (ES, m/z): 750.3 (M+H) + . 1 H NMR (400 MHz, DMSO-d 6 ): δ 12.90 (s, 1H), 10.93 (s, 1H), 9.74 (s, 1H), 8.71 - 8.47 (m, 2H), 8.22 - 8.12 (m, 1H), 8.07 (br d, J = 4.3 Hz, 1H), 7.94 - 7.69 (m, 3H), 7.68 - 7.60 (m, 1H), 7.56 - 7.48 (m, 1H), 7.42 - 7.28 (m, 1H), 7.05 - 6.92 (m, 1H), 4.92 - 4.71 (m, 1H), 4.23 - 4.03 (m, 2H), 3.72 - 3.62 (m, 3H), 2.24 - 1.78 (m, 4H), 1.58 - 1.36 (m, 1H), 1.29 - 1.21 (m, 3H), 1.19 - 1.11 (m, 3H), 1.07 - 0.92 (m, 3H), 0.33 – 0.42 ( m, 1H). EXAMPLE 2 (R,Z)-5-(5-chloro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl)phe nyl)-2-(1 5 -(ethoxycarbonyl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphane-9-yl ridine 1-oxide Step 1: 1-(2-bromo-4-chlo ole To a solution of 2-bromo-4-chloro-1-fluorobenzene (8.32 g, 39.7 mmol) and 4-(trifluoromethyl)- 1H-pyrazole (3.20 g, 23.5 mmol) in DMA (40 mL) was added Cs2CO3 (15.32 g, 47.0 mmol). The mixture was heated to 80 °C for 20 h. Water (80 mL) was added and the mixture was extracted with ethyl acetate (40 mL x 2). The combined organic fractions were washed with brine (40 mL x 2), dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (0-20% EtOAc/petroleum ether) to give 1-(2-bromo-4-chlorophenyl)-4-(trifluoromethyl)-1H-pyrazole. Step 2: methyl ((1 2 Z,8Z)-9-(5-(5-chloro-2-(4-(trifluoromethyl)-1H-pyrazol -1-yl)phenyl)pyridin- 2-yl)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola- 2(1,2)-benzenacyclononaphan-8-en-2 4 -yl)carbamate To a stirred mixture of (6-((1 2 Z,8Z)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -en- 9-yl)pyridin-3-yl)boronic acid (1.50 g, 2.48 mmol), 1-(2-bromo-4-chlorophenyl)-4- (trifluoromethyl)-1H-pyrazole (0.766 g, 2.35 mmol), potassium phosphate (2.477 mL, 4.95 mmol) in THF (15 mL) was added cataCXium® A Pd G2 (0.166 g, 0.248 mmol) and the mixture was stirred at 80 °C for 2 h under N2 atmosphere. LCMS showed the reaction was completed. Water (10 mL) was added and the mixture was extracted with ethyl acetate (20 mL x 2). The combined organic fractions were washed with brine (30 mL x 2), dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (0-40% EtOAc/petroleum ether) to give methyl ((1 2 Z,8Z)-9-(5-(5- chloro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl)phenyl)pyridin -2-yl)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -en- 2 4 -yl)carbamate. LCMS (ES, m/z): 806.3 (M+H) + . Step 3: methyl ((1 2 Z,8Z)-9-(5-(5-chloro-2-(4-(trifluoromethyl)-1H-pyrazol -1-yl)phenyl)pyridin- 2-yl)-1 5 -iodo-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)- imidazola-2(1,2)-benzenacyclononaphan-8-en-2 4 -yl)carbamate To a stirred mixture of methyl ((1 2 Z,8Z)-9-(5-(5-chloro-2-(4-(trifluoromethyl)-1H-pyrazol -1- yl)phenyl)pyridin-2-yl)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8-en-2 4 -yl)carbamate (1.3 g, 1.6 mmol) in CH 3 COOH (10 mL) was added 1-iodopyrrolidine-2,5-dione (0.381 g, 1.69 mmol) at 25 °C and the mixture was stirred at 40 °C for 2 h . LCMS showed that the reaction was completed. The reaction mixture was carefully added dropwise to the NaHCO 3 solution at 0 °C until PH = 7. The mixture was extracted with ethyl acetate (50 mL x 2). The combined organic fractions were washed with brine (50 mL x 2), dried over Na2SO4, filtered and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (0-40% EtOAc/petroleum ether) to give methyl ((1 2 Z,8Z)-9-(5-(5-chloro-2-(4-(trifluoromethyl)-1H- pyrazol-1-yl)phenyl)pyridin-2-yl)-1 5 -iodo-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -en- 2 4 -yl)carbamate. LCMS (ES, m/z): 932.1 (M+H) + . Step 4: ethyl (1 2 Z,8Z)-9-(5-(5-chloro-2-(4-(trifluoromethyl)-1H-pyrazol -1-yl)phenyl)pyridin-2- yl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H- 3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8-ene-1 5 -carboxylate To a mixture of methyl ((1 2 Z,8Z)-9-(5-(5-chloro-2-(4-(trifluoromethyl)-1H-pyrazol -1- yl)phenyl)pyridin-2-yl)-1 5 -iodo-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3- aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8-en-2 4 -yl)carbamate (1.10 g, 1.18 mmol) in EtOH (10 mL) and DMSO (10 mL) was added PdCl2(dppf) (0.259 g, 0.354 mmol) and TEA (1.645 mL, 11.8 mmol) and the mixture was stirred at 70 °C under CO atmosphere at 50 psi for 16 h. LCMS showed the reaction was completed. The solvent was removed under reduced pressure and the residue was purified by column chromatography on silica (0-50% EtOAc/petroleum ether) to give ethyl (1 2 Z,8Z)-9-(5-(5-chloro-2-(4-(trifluoromethyl)-1H-pyrazol - 1-yl)phenyl)pyridin-2-yl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -ene- 1 5 -carboxylate. LCMS (ES, m/z): 878.3 (M+H) + . Step 5: ethyl (R,Z)-9-(5-(5-chloro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl) phenyl)pyridin-2-yl)- 2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3- aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-1 5 -carboxylate To a stirred mixture of 5,5'-bis(bis(3,5-di-tert-butyl-4-methoxyphenyl)phosphino)-4, 4'- bibenzo[d][1,3]dioxole (102 mg, 0.087 mmol) in 1,2-dichloroethane (1 mL) was added (S)-(+)- 5,5'-bis[di(3,5-di-tert-butyl-4-methoxyphenyl)phosphino]-4,4 '-bi-1,3-benzodioxole (32.4 mg, 0.087 mmol) at room temperature in glovebox and the mixture was stirred at 25 °C for 1 h under N2 atmosphere. A stirred mixture of ethyl (1 2 Z,8Z)-9-(5-(5-chloro-2-(4-(trifluoromethyl)-1H- pyrazol-1-yl)phenyl)pyridin-2-yl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -ene- 1 5 -carboxylate (760 mg, 0.865 mmol) in 2,2,2-trifluoroethan-1-ol (5 mL) was added to the above catalyst mixture at room temperature and the mixture was stirred at 45 °C for 36 h under H2 atmosphere (50 psi). The solvent was removed under reduced pressure and the residue was purified by column chromatography on silica (0-40% EtOAc/petroleum ether) to give ethyl (R,Z)-9-(5-(5-chloro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl) phenyl)pyridin-2-yl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-1 5 -carboxylate. LCMS (ES, m/z): 880.4 (M+H) + . Step 6: (R,Z)-5-(5-chloro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl)phe nyl)-2-(1 5 - (ethoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 9- yl)pyridine 1-oxide Ethyl (R,Z)-9-(5-(5-chloro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl) phenyl)pyridin-2-yl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-1 5 -carboxylate (640 mg, 0.727 mmol) in CH 3 COOOH (5 mL, 10% wt) was stirred at 25 °C for 2 h. LCMS showed the reaction was completed. The reaction mixture was carefully transferred dropwise to a cooled (0 °C) mixture of 20 g of ice, saturated NaHCO 3 (80 mL) and saturated Na 2 SO 3 (80 mL). The mixture was extracted with EtOAc (200 mL x 2). The combined organic fractions were washed with brine (200 mL), dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure to give (R,Z)-5-(5-chloro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl)phe nyl)-2-(1 5 -(ethoxycarbonyl)- 2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3- aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-9-yl)pyrid ine 1-oxide which was used in next step without further purification. LCMS (ES, m/z): 896.3 (M+H) + . Step 7: (R, Z)-5-(5-chloro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl)phenyl )-2-(1 5 - (ethoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 H-3-aza-1(4,2)-imidazola- 2(1,2)-benzenacyclononaphane-9-yl)pyridine 1-oxide To a mixture of (R,Z)-5-(5-chloro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl)phe nyl)-2-(1 5 - (ethoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 9- yl)pyridine 1-oxide (640 mg, 0.714 mmol), 2-amino-3-mercaptopropanoic acid (260 mg, 2.14 mmol) in CH2Cl2 (6 mL) was added TFA (6 mL) and the mixture was stirred at 40 °C for 2 h under N 2 atmosphere. The solvent was removed under reduced pressure. Aqueous NaHCO 3 (saturated, 5 mL) was added and the mixture was extracted with ethyl acetate (10 mL x 2). The combined organic fractions were washed with brine (15 mL x 2), dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (0-60% EtOAc/petroleum ether). It was further purified by chiral SFC (DAICEL CHIRALPAK AS, 40% 0.1% NH3H2O EtOH/CO2), and then by reverse phase HPLC (ACN/water with 0.04%NH3H2O+10mM NH4HCO3 modifier) to give (R, Z)-5-(5-chloro-2-(4- (trifluoromethyl)-1H-pyrazol-1-yl)phenyl)-2-(1 5 -(ethoxycarbonyl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphane-9-yl)pyridine 1-oxide. LCMS (ES, m/z): 766.2 (M+H) + . 1 H NMR (500 MHz, CDCl3): δ 12.39-11.97 (m, 1H), 11.70 (br s, 1H), 8.30 (s, 1H), 7.91 (br d, J = 8.5 Hz, 1H), 7.82 (s, 1H), 7.77 (br s, 1H), 7.70 (s, 1H), 7.64 - 7.48 (m, 4H), 7.33 (br d, J = 8.1 Hz, 1H), 6.97 - 6.77 (m, 2H), 4.87-4.72 (m, 1H), 4.29 (q, J = 6.9 Hz, 2H), 3.76 (s, 3H), 3.03-2.63 (m, 1H), 2.37- 2.11 (br s, 1H), 1.98-1.78 (m, 1H), 1.61-1.46 (m, 2H), 1.39 - 1.13 (m, 10H). EXAMPLE 3 (R, Z)-5-(2-(difluoromethoxy)-5-fluorophenyl)-2-(1 5 -(ethoxycarbonyl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphane-9-yl)pyridine 1-oxide Step 1: 2-bromo-1-(diflu To a stirred mixture of 2-bromo-4-fluorophenol (20.0 g, 105 mmol) in DMF (250 mL) was added K2CO3 (43.4 g, 314 mmol) and sodium 2-chloro-2,2-difluoroacetate (19.16 g, 126 mmol) and the reaction mixture was stirred at 70 °C for 15 h under N 2 . Water (500 mL) was added and the mixture was extracted with petroleum ether (500 mL x 3). The combined organic fractions were washed with brine (500 mL), dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (0-10% EtOAc/petroleum ether) to give 2-bromo-1-(difluoromethoxy)-4-fluorobenzene. 1 H NMR (400 MHz, CD3OD): δ 7.52 - 7.45 (m, 1H), 7.35 - 7.25 (m, 1H), 7.20 - 7.10 (m, 1H), 6.76 (t, J = 72 Hz, 1H). Step 2: methyl ((1 2 Z,8Z)-9-(5-(2-(difluoromethoxy)-5-fluorophenyl)pyridin -2-yl)-5,5-dimethyl- 4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphan-8-en-2 4 -yl)carbamate To a stirred mixture of (6-((1 2 Z, 8Z)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -en- 9-yl)pyridin-3-yl)boronic acid (11.3 g, 18.7 mmol) and PdCl2(dppf) (0.956 g, 1.31 mmol) in dioxane (120 mL) were added 2-bromo-1-(difluoromethoxy)-4-fluorobenzene (6.75 g, 28.0 mmol) and potassium phosphate (2M, 28.0 mL, 56.0 mmol). The reaction mixture was stirred at 80 °C for 4 hours under N 2 atmosphere. Water (150 mL) was added and the mixture was extracted with ethyl acetate (250 mL x 2). The combined organic fractions were washed with brine (250 mL), dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (0-50% EtOAc/petroleum ether) to give methyl ((1 2 Z,8Z)-9-(5-(2-(difluoromethoxy)-5-fluorophenyl)pyridin -2-yl)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -en- 2 4 -yl)carbamate. LCMS (ES, m/z): 722.6 (M+H) + . Step 3: methyl ((1 2 Z,8Z)-9-(5-(2-(difluoromethoxy)-5-fluorophenyl)pyridin -2-yl)-1 5 -iodo-5,5- dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphan-8-en-2 4 -yl)carbamate To a stirred mixture of methyl ((1 2 Z,8Z)-9-(5-(2-(difluoromethoxy)-5-fluorophenyl)pyridin -2-yl)- 5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphan-8-en-2 4 -yl)carbamate (8.0 g, 11 mmol) in CH 3 COOH (60 mL) was added 1-iodopyrrolidine-2,5-dione (2.244 g, 9.97 mmol) at 25 °C and the reaction mixture was stirred at 40 °C for 1 h. The reaction mixture was carefully added dropwise to NaHCO3 solution at 0 °C until PH = 7. The mixture was extracted with ethyl acetate (500 mL x 2). The combined organic fractions were washed with brine (500 mL), dried over Na2SO4, filtered and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (0-60% EtOAc/petroleum ether) to give methyl ((1 2 Z,8Z)-9-(5-(2-(difluoromethoxy)-5- fluorophenyl)pyridin-2-yl)-1 5 -iodo-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)- 1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -en-2 4 -yl)carbamate. LCMS (ES, m/z): 848.3 (M+H) + . Step 4: ethyl (1 2 Z,8Z)-9-(5-(2-(difluoromethoxy)-5-fluorophenyl)pyridin -2-yl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8-ene-1 5 -carboxylate To a mixture of methyl ((1 2 Z,8Z)-9-(5-(2-(difluoromethoxy)-5-fluorophenyl)pyridin -2-yl)-1 5 - iodo-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola- 2(1,2)-benzenacyclononaphan-8-en-2 4 -yl)carbamate (9.0 g, 9.6 mmol) in EtOH (90 mL) and DMSO (90 mL) were added Pd(dppf)Cl2 (1.958 g, 2.68 mmol) and Et3N (7.99 mL, 57.3 mmol) under N 2 . The suspension was degassed under vacuum and purged with CO several times. The reaction mixture was stirred under CO (50 psi) at 70 °C for 23 hours. The reaction mixture was cooled to 25 °C, water (150 mL) was added and the mixture was extracted with ethyl acetate (250 mL x 2). The combined organic fractions were washed with brine (250 mL), dried over Na2SO4, filtered and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (10-40% EtOAc/petroleum ether) to give ethyl (1 2 Z,8Z)-9-(5- (2-(difluoromethoxy)-5-fluorophenyl)pyridin-2-yl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl- 4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphan-8-ene-1 5 -carboxylate. LCMS (ES, m/z): 794.2 (M+H) + . Step 5: ethyl (R,Z)-9-(5-(2-(difluoromethoxy)-5-fluorophenyl)pyridin-2-yl) -2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-1 5 -carboxylate To a stirred mixture of bis(norbornadiene)rhodium(I) tetrafluoroborate (0.313 g, 0.838 mmol) in 1,2-dichloroethane (5 mL) was added (S)-(+)-5,5'-bis[di(3,5-di-tert-butyl-4- methoxyphenyl)phosphino]-4,4'-bi-1,3-benzodioxole (0.988 g, 0.838 mmol) at room temperature in glovebox and the mixture was stirred at 25 °C for 1 h under N 2 atmosphere. A stirred mixture of ethyl (1 2 Z,8Z)-9-(5-(2-(difluoromethoxy)-5-fluorophenyl)pyridin -2-yl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8-ene-1 5 -carboxylate (7.0 g, 8.8 mmol) in 2,2,2- trifluoroethan-1-ol (60 mL) was added the above catalyst mixture at room temperature and the reaction mixture was stirred at 50 °C for 48 h under H 2 atmosphere(50psi). The mixture was concentrated. The residue was purified by column chromatography on silica (0-40% EtOAc/petroleum ether) to give ethyl (R,Z)-9-(5-(2-(difluoromethoxy)-5-fluorophenyl)pyridin-2- yl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H- 3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-1 5 -carboxylate. LCMS (ES, m/z): 796.2 (M+H) + . Step 6: (R, Z)-5-(2-(difluoromethoxy)-5-fluorophenyl)-2-(1 5 -(ethoxycarbonyl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-9-yl)pyridine 1-oxide To a stirred mixture of ethyl (R,Z)-9-(5-(2-(difluoromethoxy)-5-fluorophenyl)pyridin-2-yl) -2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-1 5 -carboxylate (2.1 g, 2.6 mmol) and oxone (12.98 g, 21.11 mmol), 18-Crown-6 (5.58 g, 21.1 mmol) in MeOH (30 mL) was added water (8 mL) at 25 °C and the reaction mixture was stirred at 25 °C for 2 h under N2 atmosphere. Water (200 mL) was added and the mixture was extracted with ethyl acetate (200 mL x 2). The combined organic fractions were washed with aqueous Na2SO3 (100 mL), dried over Na2SO4, filtered and the solvent was evaporated under reduced pressure to give (R, Z)-5-(2- (difluoromethoxy)-5-fluorophenyl)-2-(1 5 -(ethoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5- dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphane-9-yl)pyridine 1-oxide which was used in the next step without further purification. LCMS (ES, m/z): 812.1 (M+H) + . Step 7: (R, Z)-5-(2-(difluoromethoxy)-5-fluorophenyl)-2-(1 5 -(ethoxycarbonyl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphane-9-yl)pyridine 1-oxide To a stirred mixture of (R, Z)-5-(2-(difluoromethoxy)-5-fluorophenyl)-2-(1 5 -(ethoxycarbonyl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-9-yl)pyridine 1-oxide (4.0 g, 4.9 mmol) and 2- amino-3-mercaptopropanoic acid (2.98 g, 24.6 mmol) in CH 2 Cl 2 (15 mL) was added TFA (30 mL) at 25 °C and the mixture was stirred at 25 °C for 2 h under N2 atmosphere. The solvent was removed and NaHCO 3 (200 mL) was added and the mixture was extracted with ethyl acetate (250 mL x 2). The combined organic fractions were washed with brine (200 mL), dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (0-70% EtOAc/petroleum ether). It was further purified by chiral SFC (DAICEL CHIRALCEL OD, 45% 0.1% NH3H2O MeOH/CO2) to give (R, Z)-5-(2-(difluoromethoxy)-5-fluorophenyl)-2-(1 5 -(ethoxycarbonyl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphane-9-yl)pyridine 1-oxide. LCMS (ES, m/z): 682.0 (M+H) + . 1 H NMR (400 MHz, CD3OD): δ 8.46 (br s, 1H), 8.02 - 7.51 (m, 4H), 7.40 - 7.22 (m, 4H), 6.85 (t, J = 54.6 Hz, 1H), 4.95 - 4.85 (m, 1H), 4.29 - 4.08 (m, 2H), 3.73 (s, 3H), 2.22 - 2.14 (m, 2H), 1.90 - 1.78 (m, 1H), 1.50 - 1.42 (m, 1H), 1.33 - 0.99 (m, 11H) EXAMPLE 4 (R,Z)-5'-chloro-2'-(difluoromethoxy)-6-(1 5 -(ethoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5- dimethyl-4-oxo-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 9-yl)-[3,3'- bipyridine] 1-oxide Step 1: 3-bromo-5-chlor To a mixture of 3-bromo-5-chloropyridin-2-ol (1.85 g, 8.88 mmol) and Na2CO3 (1.88 g, 17.8 mmol) in MeCN (20 mL) was added 2,2-difluoro-2-(fluorosulfonyl)acetic acid (1.581 g, 8.88 mmol). The reaction mixture was stirred at 25 °C for 15 hours. The reaction mixture was diluted with EtOAc (20 mL) and water (20 mL). The aqueous layer was extracted with EtOAc (20 mL x 2). The combined organic phases were dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by column chromatography on silica (0-30% EtOAc/petroleum ether) to give 3-bromo-5-chloro-2-(difluoromethoxy)pyridine. 1 H NMR (400 MHz, CDCL3): δ 8.08 (s, 1H), 7.95 (s, 1H), 7.38 (t, J = 72.0 Hz, 1H). Step 2: methyl ((1 2 Z,8Z)-9-(5'-chloro-2'-(difluoromethoxy)-[3,3'-bipyridi n]-6-yl)-5,5-dimethyl- 4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclonona phan-8-en-2 4 -yl)carbamate To a mixture of (6-((1 2 Z,8Z)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trime thylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -en-9-yl) pyridin-3-yl)boronic acid (0.945 g, 1.56 mmol) and 3-bromo-5-chloro-2-(difluorometh oxy)pyridine (0.565 g, 2.19 mmol) in dioxane (20 mL) were added Pd(dppf)Cl2 (0.115 g, 0.140 mmol) and K3PO4 (2M, 2.34 mL, 4.68 mmol) under N2. The reaction mixture was stirred at 80 °C for 5 hours. The reaction was then diluted with EtOAc (30 mL) and water (50 mL). The aqueous layer was extracted with EtOAc (20 mL x 2). The combined organic phases were dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by column chromatography on silica (20-50% EtOAc/petroleum ether) to give methyl ((1 2 Z,8Z)-9-(5'- chloro-2'-(difluoromethoxy)-[3,3'-bipyridin]-6-yl)-5,5-dimet hyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclonona phan-8-en- 2 4 -yl)carbamate. LCMS (ES, m/z): 739.2 (M+H) + . Step 3: methyl ((1 2 Z,8Z)-9-(5'-chloro-2'-(difluoromethoxy)-[3,3'-bipyridi n]-6-yl)-1 5 -iodo-5,5-di methyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphan-8-en-2 4 -yl)carbamate To a mixture of methyl ((1 2 Z,8Z)-9-(5'-chloro-2'-(difluoromethoxy)-[3,3'-bipyridi n]-6-yl)-5,5- dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclonona phan-8-en-2 4 -yl)carbamate (1.23 g, 1.66 mmol) in acetic acid (20 mL) was added N-iodosuccinimide (0.356 g, 1.58 mmol). The reaction mixture was stirred at 40 °C for 1 hour. The reaction mixture was concentrated in vacuo to remove AcOH and the residual oil was diluted with EtOAc (30 mL) and poured into aqueous 2 N K2CO3 till pH = 8. It was extracted with EtOAc (30 mL x 2). The combined organic phases were dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by column chromatography on silica (10-45% EtOAc/petroleum ether) to give methyl ((1 2 Z,8Z)-9-(5'-chloro-2'-(difluoromethoxy)-[3,3'- bipyridin]-6-yl)-1 5 -iodo-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8-en-2 4 -yl)carbamate. LCMS (ES, m/z): 865.0 (M+H) + . Step 4: ethyl (1 2 Z,8Z)-9-(5'-chloro-2'-(difluoromethoxy)-[3,3'-bipyridi n]-6-yl)-2 4 - ((methoxycarbo nyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3- aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8-ene-1 5 -carboxylate To a mixture of methyl ((1 2 Z,8Z)-9-(5'-chloro-2'-(difluoromethoxy)-[3,3'-bipyridi n]-6-yl)-1 5 - iodo-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola- 2(1,2)-benzenacyclononaphan-8-en-2 4 -yl)carbamate (1.00 g, 1.16 mmol) in EtOH (15 mL) and DMSO (15 mL) were added Pd(dppf)Cl2 (0.254 g, 0.347 mmol) and Et3N (1.611 mL, 11.56 mmol) under N 2 . The suspension was degassed under vacuum and purged with CO several times. The mixture was stirred under CO (50 psi) at 70 °C for 20 hours. The reaction mixture was cooled to 25 °C, concentrated in vacuo to remove EtOH. The residual was diluted with EtOAc (20 mL) and water (50 mL). The aqueous layer was extracted with EtOAc (30 mL x 2). The combined organic phases were dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by column chromatography on silica (10-45% EtOAc/petroleum ether) to give ethyl (1 2 Z,8Z)-9-(5'-chloro-2'-(difluoromethoxy)-[3,3'-bipyridi n]-6-yl)-2 4 -((methoxycarbo nyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)- imidazola-2(1,2)-benzenacyclononaphan-8-ene-1 5 -carboxylate. LCMS (ES, m/z): 811.2 (M+H) + . Step 5: ethyl (R,Z)-9-(5'-chloro-2'-(difluoromethoxy)-[3,3'-bipyridin]-6-y l)-2 4 - ((methoxycarbonyl) amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3- aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-1 5 -carboxylate To a stirred mixture of (S)-(+)-5,5'-bis[di(3,5-di-tert-butyl-4-methoxyphenyl)phosph ino]-4,4'-bi- 1,3-benzodioxole (118 mg, 0.100 mmol) in 1,2-dichloroethane (2.0 mL) was added bis(norbornadiene)rhodium(I) tetrafluoroborate (37.3 mg, 0.100 mmol) at 25 °C in a glovebox and the mixture was stirred at 25 °C for 1 h under N 2 . To a mixture of ethyl (1 2 Z,8Z)-9-(5'- chloro-2'-(difluoromethoxy)-[3,3'-bipyridin]-6-yl)-2 4 -((methoxycarbo nyl)amino)-5,5-dimethyl- 4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphan-8-ene-1 5 -carboxylate (810 mg, 0.998 mmol) in 2,2,2-trifluoroethan-1-ol (20 mL) was added the above mixture under N 2 . The suspension was degassed under vacuum and purged with H2 several times. The mixture was stirred under H2 (50 psi) at 50 °C for 48 hours. The reaction mixture was cooled to 25 °C and concentrated to dryness. The residue was purified by column chromatography on silica (10-50% EtOAc/petroleum ether) to afford ethyl (R,Z)-9- (5'-chloro-2'-(difluoromethoxy)-[3,3'-bipyridin]-6-yl)-2 4 -((methoxycarbonyl) amino)-5,5- dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphane-1 5 -carboxylate. LCMS (ES, m/z): 813.2 (M+H) + . Step 6: (R,Z)-5'-chloro-2'-(difluoromethoxy)-6-(1 5 -(ethoxycarbonyl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-9-yl)-[3,3'-bi pyridine] 1-oxide A mixture of ethyl (R,Z)-9-(5'-chloro-2'-(difluoromethoxy)-[3,3'-bipyridin]-6-y l)-2 4 -((meth oxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1 (4,2)-imidazola-2(1,2)-benzenacyclononaphane-1 5 -carboxylate (690 mg, 0.848 mmol) in CH 3 COOOH (12 mL) (8 ~ 10%) was stirred at 25 °C for 4 hours. The reaction mixture was diluted with EtOAc (20 mL) and poured into saturated aqueous Na2SO3/NaHCO3 (v/v, 1:1) till pH = 8. It was then extracted with EtOAc (30 mL x 2). The combined organic phases were dried over Na2SO4, filtered and concentrated in vacuo to afford (R,Z)-5'-chloro-2'-(difluoromethoxy)-6- (1 5 -(ethoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 9-yl)- [3,3'-bipyridine] 1-oxide which was used in the next step without further purification. LCMS (ES, m/z): 829.2 (M+H) + . Step 7: (R,Z)-5'-chloro-2'-(difluoromethoxy)-6-(1 5 -(ethoxycarbonyl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphane-9-yl)-[3,3'-bipyridine] 1-oxide To a mixture of (R,Z)-5'-chloro-2'-(difluoromethoxy)-6-(1 5 -(ethoxycarbonyl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-9-yl)-[3,3'-bi pyridine] 1-oxide (700 mg, 0.844 mmol) in TFA (10 mL) and DCM (5.0 mL) was added 2-amino-3-mercaptopropanoic acid (307 mg, 2.53 mmol). The reaction mixture was stirred at 40 °C for 2 hours. The reaction mixture was concentrated in vacuo and diluted with EtOAc (30 mL) and poured into aqueous 2 N K2CO3 until pH = 8. It was then extracted with EtOAc (30 mL x 2). The combined organic phases were dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by column chromatography on silica (25-65% EtOAc/petroleum ether). It was further purified by chiral SFC (DAICEL CHIRALCEL OD, 40% 0.1% NH3H2O EtOH/CO2) to give (R,Z)-5'-chloro-2'- (difluoromethoxy)-6-(1 5 -(ethoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo- 1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 9-yl)-[3,3'-bipyridine] 1-oxide. LCMS (ES, m/z): 699.2 (M+H) + . 1 H NMR (400 MHz, CD3OD): δ 8.61 (s, 1H), 8.30 (s, 1H), 8.14 (s, 1H), 8.07 - 7.27 (m, 6H), 5.07 - 4.90 (m, 1H), 4.31 - 4.04 (m, 2H), 3.73 (s, 3H), 2.30 - 2.01 (m, 2H), 1.72 - 1.39 (m, 2H), 1.31 - 0.97 (m, 11H). EXAMPLE 5 (R,Z)-5-(3-chloro-2-fluorophenyl)-2-(1 5 -(ethoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5- dimethyl-4-oxo-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 9-yl)pyridine 1- oxide Step 1: methyl ((1 2 Z,8Z)- yl)-5,5-d 1 imethyl-4-oxo-1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -en- 2 4 -yl)carbamate To a stirred mixture of methyl ((1 2 Z,8Z)-9-(5-bromopyridin-2-yl)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -en- 2 4 -yl)carbamate (1.0 g, 1.6 mmol) and PdCl 2 (dppf) (0.114 g, 0.156 mmol) in THF (15 mL) were added (3-chloro-2-fluorophenyl)boronic acid (0.354 g, 2.03 mmol) and potassium phosphate (2M, 2.341 mL, 4.68 mmol). The reaction mixture was stirred at 85 °C for 16 hours under N 2 atmosphere. Water (50 mL) was added and the mixture was extracted with ethyl acetate (50 mL x 2). The combined organic fractions were washed with brine (50 mL), dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (0-40% EtOAc/petroleum ether) to give methyl ((1 2 Z,8Z)-9-(5-(3- chloro-2-fluorophenyl)pyridin-2-yl)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)- 1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -en-2 4 -yl)carbamate. LCMS (ES, m/z): 690.2 (M+H) + . Step 2: methyl ((1 2 Z,8Z)-9-(5-(3-chloro-2-fluorophenyl)pyridin-2-yl)-1 5 -iodo-5,5-dimethyl-4- oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphan-8-en-2 4 -yl)carbamate To a stirred mixture of methyl ((1 2 Z,8Z)-9-(5-(3-chloro-2-fluorophenyl)pyridin-2-yl)-5,5- dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphan-8-en-2 4 -yl)carbamate (1.15 g, 1.67 mmol) in CH 3 COOH (10 mL) was added 1-iodopyrrolidine-2,5-dione (0.352 g, 1.57 mmol) at 25 °C and the reaction mixture was stirred at 40 °C for 3 h. The reaction mixture was carefully added dropwise to NaHCO 3 solution at 0 °C until pH = 7. The mixture was extracted with ethyl acetate (50 mL x 2). The combined organic fractions were washed with brine (50 mL), dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (0-40% EtOAc/petroleum ether) to give methyl ((1 2 Z,8Z)-9-(5-(3-chloro-2- fluorophenyl)pyridin-2-yl)-1 5 -iodo-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)- 1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -en-2 4 -yl)carbamate. LCMS (ES, m/z): 816.1 (M+H) + . Step 3: ethyl (1 2 Z,8Z)-9-(5-(3-chloro-2-fluorophenyl)pyridin-2-yl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8-ene-1 5 -carboxylate To a mixture of methyl ((1 2 Z,8Z)-9-(5-(3-chloro-2-fluorophenyl)pyridin-2-yl)-1 5 -iodo-5,5- dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphan-8-en-2 4 -yl)carbamate (500 mg, 0.613 mmol) in EtOH (20 mL) and DMSO (20 mL) was added PdCl2(dppf) (134 mg, 0.184 mmol) and TEA (0.256 mL, 1.84 mmol) and the mixture was stirred at 70 °C under CO atmosphere at 50 psi for 16 h. Water (50 mL) was added. The mixture was extracted with ethyl acetate (50 mL x 2). The combined organic fractions were washed with brine (50 mL), dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (0-40% EtOAc/petroleum ether) to give ethyl (1 2 Z,8Z)-9-(5-(3-chloro-2- fluorophenyl)pyridin-2-yl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -ene- 1 5 -carboxylate. LCMS (ES, m/z): 762.2 (M+H) + . Step 4: ethyl (R,Z)-9-(5-(3-chloro-2-fluorophenyl)pyridin-2-yl)-2 4 -((methoxycarbonyl)amino)- 55-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphane-1 5 -carboxylate To a stirred mixture of (S)- (+)-5,5'-bis(bis(3,5-di-tert-butyl-4-methoxyphenyl)phosphino )-4,4'- bibenzo[d][1,3]dioxole (132 mg, 0.111 mmol) in 1,2-dichloroethane (4 mL) was added bis(norbornadiene)rhodium(I) tetrafluoroborate (41.7 mg, 0.111 mmol) at 25 °C in a glovebox and the mixture was stirred at 25 °C for 1 h under N2. To a mixture of ethyl (1 2 Z,8Z)-9-(5-(3- chloro-2-fluorophenyl)pyridin-2-yl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -ene- 1 5 -carboxylate (850 mg, 1.12 mmol) in 2,2,2-trifluoroethan-1-ol (40 mL) was added the above mixture under N2. The suspension was degassed under vacuum and purged with H2 several times. The reaction mixture was stirred under H 2 (50 psi) at 50 °C for 16 hours. The reaction mixture was cooled to 25 °C and concentrated to dryness. The residue was purified by column chromatography on silica (0-40% EtOAc/petroleum ether) to afford ethyl (R,Z)-9-(5-(3-chloro-2- fluorophenyl)pyridin-2-yl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 1 5 - carboxylate. LCMS (ES, m/z): 763.9 (M+H) + . Step 5: (R,Z)-5-(3-chloro-2-fluorophenyl)-2-(1 5 -(ethoxycarbonyl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-9-yl)pyridine 1-oxide To a stirred mixture of ethyl (R,Z)-9-(5-(3-chloro-2-fluorophenyl)pyridin-2-yl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-1 5 -carboxylate (650 mg, 0.850 mmol) and oxone (4182 mg, 6.80 mmol), 18-CROWN-6 (1798 mg, 6.80 mmol) in MeOH (30 mL) was added water (8 mL) at 25 °C and the mixture was stirred at 25 °C for 2 h under N 2 atmosphere. Water (50 mL) was added. The mixture was extracted with ethyl acetate (50 mL x 2). The combined organic fractions were washed with aqueous Na 2 SO 3 (50 mL), dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure to give (R,Z)-5-(3-chloro-2- fluorophenyl)-2-(1 5 -(ethoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 9- yl)pyridine 1-oxide which was used in the next step without further purification. LCMS (ES, m/z): 780.2 (M+H) + . Step 6: (R,Z)-5-(3-chloro-2-fluorophenyl)-2-(1 5 -(ethoxycarbonyl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphane-9-yl)pyridine 1-oxide To a stirred mixture of (R,Z)-5-(3-chloro-2-fluorophenyl)-2-(1 5 -(ethoxycarbonyl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-9-yl)pyridine 1-oxide (650 mg, 0.833 mmol), (R)-2-amino-3-mercaptopropanoic acid (303 mg, 2.50 mmol) in DCM (3 mL) was added TFA (6 mL) and the mixture was stirred at 40 °C for 1 h. The reaction mixture was concentrated to dryness. Water (50 mL) was added. The mixture was extracted with ethyl acetate (50 mL x 2). The combined organic fractions were washed with aqueous NaHCO 3 (50 mL), dried over Na2SO4, filtered and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (0-90% EtOAc/petroleum ether). It was further purified by chiral SFC (DAICEL CHIRALPAK OD, 60% 0.1% NH3H2O EtOH/CO2) to give (R,Z)-5-(3-chloro-2-fluorophenyl)-2-(1 5 -(ethoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5- dimethyl-4-oxo-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 9-yl)pyridine 1- oxide. LCMS (ES, m/z): 650.2 (M+H) + . 1 H NMR (400 MHz, CD 3 OD): δ 8.54 (s, 1H), 7.95 (br d, J = 7.4 Hz, 1H), 7.81 (br d, J = 8.6 Hz, 1H), 7.71 - 7.55 (m, 3H), 7.52 (br t, J = 6.7 Hz, 1H), 7.36 - 7.28 (m, 2H), 4.99 - 4.93 (m, 1H), 4.26 - 4.08 (m, 2H), 3.73 (s, 3H), 2.17 (br s, 2H), 1.87 (br s, 1H), 1.49 (br d, J = 7.0 Hz, 1H), 1.38 - 1.24 (m, 4H), 1.21 (br t, J =7.0 Hz, 3H), 1.15 (s, 3H), 0.90 - 0.86 (m, 1H). EXAMPLE 6 (R,Z)-5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl)phe nyl)-2-(1 5 -(methoxycarbonyl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphane-9-yl ridine 1-oxide Step 1: methyl ((1 2 Z,8Z)-9 yrazol-1-yl)phenyl)pyridin-2- yl)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphan-8-en-2 4 -yl)carbamate To a stirred mixture of methyl ((1 2 Z,8Z)-9-(5-bromopyridin-2-yl)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -en- 2 4 -yl)carbamate (3 g, 4.68 mmol), xphos-G2 (0.295 g, 0.375 mmol), 1-(4-fluoro-2-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-4-(trifluorometh yl)-1H-pyrazole (2.168 g, 6.09 mmol) in THF (60 mL) was added K3PO4 (2M, 7.0 mL, 14 mmol) under N2 at 25 °C and the mixture was stirred at 80 °C for 3 h under N 2 . Water (100 mL) was added and the mixture was extracted with ethyl acetate (200 mL x 2). The combined organic fractions were washed with brine (200 mL x 2), dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (0-60% EtOAc/petroleum ether). It was suspended in 20 mL of mixed solvent (EtOAc/petroleum ether 1:2), and the suspension was stirred for 30 minutes. The mixture was filtered and the filter cake was dried under vacuum to give methyl ((1 2 Z,8Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H- pyrazol-1-yl)phenyl)pyridin-2-yl)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H- 3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8-en-2 4 -yl)carbamate. LCMS (ES, m/z): 790.5 (M+H) + . Step 2: methyl ((1 2 Z,8Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol -1-yl)phenyl)pyridin- 2-yl)-1 5 -iodo-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)- imidazola-2(1,2)-benzenacyclononaphan-8-en-2 4 -yl)carbamate To a stirred mixture of methyl ((1 2 Z,8Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol -1- yl)phenyl)pyridin-2-yl)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8-en-2 4 -yl)carbamate (8.50 g, 10.8 mmol) in CHCl 3 (100 mL) was added 1-iodopyrrolidine-2,5-dione (2.421 g, 10.76 mmol) at 25 °C and the mixture was stirred at 30 °C for 24 h. The mixture was concentrated. The residue was purified by column chromatography on silica (0-35% EtOAc/petroleum ether) to give methyl ((1 2 Z,8Z)-9-(5- (5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl)phenyl)pyri din-2-yl)-1 5 -iodo-5,5-dimethyl-4- oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphan-8-en-2 4 -yl)carbamate. LCMS (ES, m/z): 916.5 (M+H) + . Step 3: methyl (1 2 Z,8Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol -1-yl)phenyl)pyridin-2- yl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H- 3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8-ene-1 5 -carboxylate To a mixture of methyl ((1 2 Z,8Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol -1- yl)phenyl)pyridin-2-yl)-1 5 -iodo-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3- aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8-en-2 4 -yl)carbamate (4.00 g, 4.37 mmol) in MeOH (30 mL) and DMSO (30 mL) was added Pd(dppf)Cl2 (0.959 g, 1.31 mmol) and TEA (6.09 mL, 43.7 mmol) and the mixture was stirred at 70 °C under CO atmosphere at 50 psi for 16h. The mixture was concentrated, cooled, then diluted with ethyl acetate (500 mL). It was washed with brine (200 mL x4), dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (0-35% EtOAc/petroleum ether) to give methyl (1 2 Z,8Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H- pyrazol-1-yl)phenyl)pyridin-2-yl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -ene- 1 5 -carboxylate. LCMS (ES, m/z): 916.5 (M+H) + . Step 4: methyl (R,Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl) phenyl)pyridin-2- yl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H- 3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-1 5 -carboxylate To a stirred mixture of bis(norbornadiene)rhodium(I) tetrafluoroborate (0.132 g, 0.354 mmol) in 1,2-dichloroethane (5 mL) was added (S)- (+)-5,5'-bis(bis(3,5-di-tert-butyl-4- methoxyphenyl)phosphino)-4,4'-bibenzo[d][1,3]dioxole (0.417 g, 0.354 mmol) at room temperature in a glovebox and the mixture was stirred at 25 °C for 1 h under N2 atmosphere. A stirred mixture of methyl (1 2 Z,8Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol -1- yl)phenyl)pyridin-2-yl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphan-8 -ene- 1 5 -carboxylate (3.00 g, 3.54 mmol) in 2,2,2-trifluoroethan-1-ol (50 mL) was added the above catalyst mixture at room temperature and the mixture was stirred at 50 °C for 48 h under H 2 atmosphere (50 psi). The residue was purified by column chromatography on silica (0-35% EtOAc/petroleum ether) to give methyl (R,Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol- 1-yl)phenyl)pyridin-2-yl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 1 5 - carboxylate. LCMS (ES, m/z): 851.6 (M+H) + . Step 5: (R,Z)-5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl)phe nyl)-2-(1 5 - (methoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 9- yl)pyridine 1-oxide To a stirred mixture of methyl (R,Z)-9-(5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol-1- yl)phenyl)pyridin-2-yl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 1 5 - carboxylate (5.00 g, 5.88 mmol) in CH 3 COOOH (30 mL, 10% wt) at 25 °C, and the mixture was stirred at 25 °C for 3 h. It was carefully transferred dropwise to a cooled mixture of 20 g ice, saturated NaHCO 3 (80 mL) and saturated Na 2 SO 3 (80 mL) at 0 °C. The mixture was extracted with EtOAc (200 mL x 2). The combined organic fractions were washed with brine (200 mL), dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure to give (R,Z)- 5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl)phenyl)-2 -(1 5 -(methoxycarbonyl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-9-yl)pyridine 1-oxide which was used in the next step without further purification. LCMS (ES, m/z): 866.6 (M+H) + . Step 6: (R,Z)-5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl)phe nyl)-2-(1 5 - (methoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 H-3-aza-1(4,2)- imidazola-2(1,2)-benzenacyclononaphane-9-yl)pyridine 1-oxide To a stirred mixture of (R,Z)-5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol-1-yl)phe nyl)-2-(1 5 - (methoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 9- yl)pyridine 1-oxide (5.00 g, 5.77 mmol), 2-amino-3-mercaptopropanoic acid (3.50 g, 28.9 mmol) in DCM (20 mL) was added 2,2,2-trifluoroacetic acid (40 mL, 5.77 mmol) and the mixture was stirred at 40 °C for 2 h. The mixture was concentrated to dryness. The residue was dissolved in 200 mL ethyl acetate. It was carefully added dropwise to the aqueous sodium bicarbonate at 0 °C. The mixture was extracted with ethyl acetate (200 mL x 2). The combined organic fractions were washed with brine (200 mL x 2), dried over Na2SO4, filtered and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography on silica (0-80% EtOAc/petroleum ether). It was further purified by chiral SFC (DAICEL CHIRALCEL OD, 55% 0.1% NH 3 H 2 O EtOH/CO 2 ) to give (R,Z)-5-(5-fluoro-2-(4-(trifluoromethyl)-1H-pyrazol-1- yl)phenyl)-2-(1 5 -(methoxycarbonyl)-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 H-3- aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-9-yl)pyrid ine 1-oxide. LCMS (ES, m/z): 736.2 (M+H) + . 1 H NMR (400 MHz, DMSO-d 6 ): δ 12.93 (s, 0.5H), 12.19 (br s, 0.5H), 10.89 (br s, 0.5H), 9.96 - 9.65 (m, 1H), 8.64 (br d, J = 11.2 Hz, 1H), 8.57 (br s, 0.5H), 8.25 - 8.10 (m, 1H), 8.07 (br s, 1H), 7.94 (br d, J = 8.1 Hz, 0.5H), 7.82 - 7.68 (m, 2H), 7.64 - 7.63 (m, 1.5H), 7.55 - 7.53 (m, 1.5H), 7.46 - 7.35 (m, 1H), 7.32 (br d, J = 8.3 Hz, 0.5H), 7.06 - 6.92 (m, 1H), 4.95 - 4.69 (m, 1H), 3.77 - 3.55 (m, 6H), 2.27 - 1.71 (m, 3H), 1.59 - 1.30 (m, 1H), 1.29 - 0.23 (m, 8H). EXAMPLE 7 ammonium (R,Z)-9-(5-(2-(difluoromethoxy)-5-fluorophenyl)-1-oxidopyrid in-2-yl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 H-3-aza-1(4,2)-imidazola-2(1,2) benzenacyclononaphane-1 5 -carboxylate Step 1: (Z)-9-(5-(2-(dif 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-1 5 -carboxylic acid A mixture of ethyl (Z)-9-(5-(2-(difluoromethoxy)-5-fluorophenyl)pyridin-2-yl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-1 5 -carboxylate (2 g, 2.51 mmol), lithium hydroxide hydrate (0.527 g, 12.56 mmol) in THF (20 mL), water (6 mL) and MeOH (5 mL) was stirred at 40 °C for 16 h. The reaction was quenched with HCl (1M) to the pH=3, water (10 mL) was added and the mixture was extracted with ethyl acetate (2 * 100 mL). The combined organic portions were washed with brine (saturated, 2 x 50 mL), dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure. The residue was purified by silica gel chromatography to afford the title compound. LCMS (ES, m/z): 768.5 (M+H) + . Step 2: (Z)-2-(1 5 -carboxy-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2- (trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 9-yl)- 5-(2-(difluoromethoxy)-5-fluorophenyl)pyridine 1-oxide A mixture of (Z)-9-(5-(2-(difluoromethoxy)-5-fluorophenyl)pyridin-2-yl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 -((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza- 1(4,2)-imidazola-2(1,2)-benzenacyclononaphane-1 5 -carboxylic acid (1g, 1.302 mmol) in CH3COOOH (10 mL, 10% wt) was stirred at 25 °C for 6 h. The reaction was carefully transferred dropwise to a cooled 0 °C mixture of 5 g ice, sat. NaHCO 3 (50 mL)/sat. Na 2 SO 3 (50 mL). The mixture was extracted with EtOAc (2 * 150 mL). The combined organic portions were washed with brine (100 mL), dried over Na 2 SO 4 , filtered and the solvent was evaporated under reduced pressure to afford the title compound. The residue was used in next step without further purification. LCMS (ES, m/z): 784.4 (M+H) + . Step 3: (Z)-2-(1 5 -carboxy-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 H-3-aza-1(4,2)- imidazola-2(1,2)-benzenacyclononaphane-9-yl)-5-(2-(difluorom ethoxy)-5-fluorophenyl)pyridine 1-oxide To a stirred mixture of (Z)-2-(1 5 -carboxy-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 - ((2-(trimethylsilyl)ethoxy)methyl)-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 9-yl)-5-(2-(difluoromethoxy)-5-fluorophenyl)pyridine 1-oxide (1.1 g, 1.403 mmol) and DL- cysteine (0.850 g, 7.02 mmol) in DCM (5 mL) was added TFA (10 mL, 130 mmol) at room temperature and the mixture was stirred at 40 °C for 1 h. The mixture was concentrated and the residue was diluted with EtOAc (50 mL). The mixture was poured into 100 mL of NaHCO3 solution at 0 °C and then was extracted with ethyl acetate (2 * 100 mL). The combined organic portions were washed with brine (50 mL), dried over Na2SO4, filtered and the solvent was evaporated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO ® ; 12 g Agela ® Silica Flash Column, Eluent of 0~3% MeOH/DCM gradient @ 30 mL/min, 35 min, dry loaded) to give the title compound. LCMS (ES, m/z): 654.3 (M+H) + . Step 4: ammonium (R,Z)-9-(5-(2-(difluoromethoxy)-5-fluorophenyl)-1-oxidopyrid in-2-yl)-2 4 - ((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)- benzenacyclononaphane-1 5 -carboxylate (Z)-2-(1 5 -carboxy-2 4 -((methoxycarbonyl)amino)-5,5-dimethyl-4-oxo-1 1 H-3-aza-1(4,2)- imidazola-2(1,2)-benzenacyclononaphane-9-yl)-5-(2-(difluorom ethoxy)-5-fluorophenyl)pyridine 1-oxide (480 mg, 0.734 mmol) was separated with SFC (Column DAICEL CHIRALCEL OD (250 mm * 30 mm, 10 um), Condition 0.1%NH 3 H 2 O, EtOH. Begin B 50, End B 50, Gradient FlowRate (mL/min) 80, Injections 150) to give Peak 1 (S)-isomer (RT= 0.868 min) and Peak 2 (R)-isomer (RT= 3.819 min). The Peak 1 residue was further purified by Prep-HPLC (Column YMC-Actus Triart C18150 * 30 mm * 5um, Condition water (0.1%TFA) - ACN Begin B 36, End B 56, Gradient Time (min) 11, 100%B Hold Time(min) 2, FlowRate(ml/min) 25, Injections 7) and the solution was lyophilized to give (S,Z)-2-(1 5 -carboxy-2 4 -((methoxycarbonyl)amino)- 5,5-dimethyl-4-oxo-1 1 H-3-aza-1(4,2)-imidazola-2(1,2)-benzenacyclononaphane- 9-yl)-5-(2- (difluoromethoxy)-5-fluorophenyl)pyridine 1-oxide. LCMS (ES, m/z): 654.2 (M+H) + . 1 H NMR (MeOD, 400MHz) δ 8.53 (d, J=1.2 Hz, 1H), 7.89 (d, J=8.4 Hz, 1H), 7.84 - 7.79 (m, 1H), 7.72 (br d, J=8.5 Hz, 1H), 7.62 (s, 1H), 7.44 - 7.33 (m, 3H), 7.33 - 7.25 (m, 1H), 6.83 (t, J=58 Hz, 1H), 4.95 – 4.90 (m, 1H), 3.77 (s, 3H), 2.46 - 2.15 (m, 2H), 1.83 - 1.55 (m, 2H), 1.50 - 1.25 (m, 4H), 1.13 (s, 3H), 1.10 - 0.80 (m, 1H). Peak 2 was lyophilized to give the title compound. LCMS (ES, m/z): 654.2 (M+H) + . 1 H NMR (MeOD, 500MHz) δ 8.51 (br s, 1H), 7.86 - 7.63 (m, 3H), 7.44 - 7.19 (m, 5H), 6.80 (t, J=58 Hz, 1H), 5.00 - 4.93 (m, 1H), 3.75 (s, 3H), 2.39 - 1.84 (m, 2H), 1.56 - 1.33 (m, 2H), 1.32 - 1.21 (m, 4H), 1.17 - 1.12 (m, 4H). By using the procedures similar to those described above, and appropriate starting materials, the following compounds were synthesized and characterized by LC/MS. E x Structure Name MS (M+1) Factor XIa assay The effectiveness of a compound of the present invention as an inhibitor of Coagulation Factor XIa can be determined using a relevant purified serine protease, and an appropriate synthetic substrate. The rate of hydrolysis of the chromogenic or fluorogenic substrate by the relevant serine protease was measured both in the absence and presence of compounds of the present invention. Assays were conducted at room temperature or at 37°C. Hydrolysis of the substrate resulted in release of amino trifluoromethylcoumarin (AFC), which was monitored spectrofluorometrically by measuring the increase in emission at 510 nm with excitation at 405 nm. A decrease in the rate of fluorescence change in the presence of inhibitor is indicative of enzyme inhibition. Such methods are known to one skilled in the art. The results of this assay are expressed as the half-maximal inhibitory concentrations (IC50), or the inhibitory constant, Ki. Compounds were pre-incubated for 30 minutes at 25°C with human (0.04 nM) Factor XIa in 50 mM HEPES buffer with 150 mM sodium chloride, 5 mM calcium chloride, 0.1% PEG 8000, pH 7.4. Factor XIa enzymatic activity was determined by addition of the substrate glycine- proline-arginine-7-amido-4-trifluoromethylcoumarin (GPR-AFC) and measurement of the fluorescence at 400/505 nm after a 60 minute incubation at 25°C. The % inhibition for each data point was calculated from the data and analyzed using the log (inhibitor) vs. response four parameters equation to determine the half-maximal inhibitory concentrations (IC50). The IC50 were converted to equilibrium inhibitory constants (Ki) using the Cheng-Prusoff equation. The activities shown by this assay indicate that the compounds of the invention may be therapeutically useful for treating or preventing various cardiovascular and/or cerebrovascular thromboembolic conditions in patients suffering from unstable angina, acute coronary syndrome, refractory angina, myocardial infarction, transient ischemic attacks, atrial fibrillation, stroke such as thrombotic stroke or embolic stroke, venous thrombosis, coronary and cerebral arterial thrombosis, cerebral and pulmonary embolism, atherosclerosis, deep vein thrombosis, disseminated intravascular coagulation, and reocclusion or restenosis of recanalized vessels. Kallikrein assay The effectiveness of a compound of the present invention as an inhibitor of Kallikrein can be determined using a relevant purified serine protease, and an appropriate synthetic substrate. The rate of hydrolysis of the chromogenic or fluorogenic substrate by the relevant serine protease was measured both in the absence and presence of compounds of the present invention. Assays were conducted at room temperature or at 37°C. Hydrolysis of the substrate resulted in release of amino trifluoromethylcoumarin (AFC), which was monitored spectrofluorometrically by measuring the increase in emission at 510 nm with excitation at 405 nm. A decrease in the rate of fluorescence change in the presence of inhibitor is indicative of enzyme inhibition. Such methods are known to one skilled in the art. The results of this assay are expressed as the half-maximal inhibitory concentrations (IC50), or the inhibitory constant, K i . Kallikrein determinations were made in 50 mM HEPES buffer at pH 7.4 containing 150 mM NaCl, 5 mM CaCl2, and 0.1% PEG 8000 (polyethylene glycol; Fisher Scientific). Determinations were made using purified Human plasma kallikrein at a final concentration of 0.5 nM (Enzyme Research Laboratories) and the synthetic substrate, Acetyl-K-P-R-AFC (Sigma # C6608) at a concentration of 100mM. Activity assays were performed by diluting a stock solution of substrate at least tenfold to a final concentration ≤ 0.2 Km into a solution containing enzyme or enzyme equilibrated with inhibitor. Times required to achieve equilibration between enzyme and inhibitor were determined in control experiments. The reactions were performed under linear progress curve conditions and fluorescence increase measured at 405 Ex/510 Em nm. Values were converted to percent inhibition of the control reaction (after subtracting 100% Inhibition value). IC 50 was determined by inflection point from a four parameter logistic curve fit. Ki was calculated using the Cheng Prusoff equation, Ki = IC 50 /(1+([S]/Km)). The activities shown by this assay indicate that the compounds of the invention may be therapeutically useful for treating or preventing various cardiovascular and/or cerebrovascular thromboembolic conditions in patients suffering from unstable angina, acute coronary syndrome, refractory angina, myocardial infarction, transient ischemic attacks, atrial fibrillation, stroke such as thrombotic stroke or embolic stroke, venous thrombosis, coronary and cerebral arterial thrombosis, cerebral and pulmonary embolism, atherosclerosis, deep vein thrombosis, disseminated intravascular coagulation, and reocclusion or restenosis of recanalized vessels. Activated partial thromboplastin time (aPTT) Assay Activated partial thromboplastin time (aPTT) is a clotting test that measures the intrinsic coagulation cascade. The test is performed in sodium citrated plasma. Human plasma is made by collecting blood from healthy donors of both genders into Na citrate tubes (Sarstedt coagulation 9NC/10ml). Blood is centrifuged at 1500 x g and the plasma is collected. aPTT is checked on each individual donor, and those within the normal range (28-40 seconds) are pooled, aliquoted, and stored at -80C. Test samples are prepared by spiking inhibitors or vehicle into plasma. These spiked samples are then run on a coagulation analyzer (STA-R Evolution, Stago Diagnostica). In general, the analyzer performs the following steps: Factor XII is activated by addition of ellagic acid (Pacific Hemostasis), and then time to clot is measured after re-calcification of the sample. Inhibition of FXI will cause aPTT clot time to be prolonged. The data are expressed as percent increase over vehicle control clot time and the concentration that causes a 50% (1.5x) percent increase of clot time. Norepinephrine Transporter Functional Antagonist Uptake Assay Human Norepinephrine Transporter (NET) Functional Antagonist Uptake Assay was carried out at Panlabs Cerep (assay#302100). Human recombinant norepinephrine transporter expressed in MDCK cells were plated overnight. Test compound and/or vehicle was pre- incubated with cells (2 x 10E5/ml) in modified Tris-HEPES buffer pH 7.1 for 20 minutes at 25 o C and 25 nM [3H]Norepinephrine was then added for an additional 15 minute incubation period. A lysate was obtained from solubilized cells and counted to determine [3H]Norepinephrine uptake. Reduction of [3H]Norepinephrine uptake by 50 percent or more (≥50%) relative to 10 µM desipramine indicates significant inhibitory activity. Compounds are screened at 10, 3, 1, 0.3, 0.1, 0.03, 0.01 and 0.003 µM. These same concentrations are concurrently applied to a separate group of untreated cells and evaluated for possible compound-induced cytotoxicity only if significant inhibition of uptake is observed. IC50 values were determined by a non-linear, least squares regression analysis using MathIQTM (IDBusiness Solutions Ltd., UK). Inhibition of Tissue Kallikrein Enzymatic Activity Compounds were assayed with human tissue kallikrein (RayBiotech, Cat # 228-10996 or Evotec, final concentration 5 nM) in 50 mM HEPES, 150 mM NaCl, 5 mM CaCl 2 , 0.1% PEG- 8000, pH 7.4 at 25 °C in a Corning 3575 non-binding surface microplate. Compounds were tested in 10-point dose titrations starting at 166.67 uM with a 3-fold dilution series. Tissue kallikrein enzymatic activity was determined by measuring the rate of cleavage of N-acetyl-KPR- AFC substrate (Sigma, Cat # C6608, final concentration 100 uM) by continuously monitoring the fluorescence at 400/505 nm using a Tecan Safire plate-reader. The initial reaction rates (0 to 20 minutes) were used to determine percent inhibition. The % Inhibition for each data point was recalculated from the RFU/min data and analyzed using the log(inhibitor) vs. response four parameters equation with the GraphPad Prism software. High-Throughput (HT) Solubility Determination The chromatographic system consists of an Agilent 1290 UPLC/DAD system and ChemStation software, both from Agilent Technologies, USA. The separations are carried out on a Supelco Ascentis Express C18, 30 mm x 3.0 mm I.D., 2.7 µm HPLC column. The mobile phase consists of Potassium phosphate buffered at pH 7 (mobile phase A) and acetonitrile (mobile phase B). The column oven temperature is set to 30 ^C and the UPLC analysis consists of a gradient. The injection volume is 2 µL and the spectrophotometric detection is set to 215 and 238 nm. A 10 mM stock solution of the compound in DMSO is supplied for analysis. 2.5 μL of stock solution (10 mM) was diluted into 247.5 μL of organic co-solvents (10% MeCN/80% MeOH/10% DMSO, v/v/v) to create a standard solution of 100 μM. To create the solubility solutions, 4.1 μL of 10 mM DMSO stock solution was diluted into 247.5 μL of phosphate buffered saline (PBS) (pH 7) solution. A second 4.1 μL aliquot of 10 mM DMSO stock solution was added to 196 μL of PBS (pH 2) solution. A third 4.1 μL aliquot of 10 mM DMSO stock solution was added to 196 μL of FaSSIF (pH 6.5) solution. Each solubility solution was sealed and shaken for 24 hours at 25°C. The equilibrated solubility solutions were filtered by centrifugation using a filter (0.45 μm, polypropylene). 50 μL each of the 100 μM standard solution and the filtered equilibrated solubility solutions were placed into a 384 well plate, and the plate was subsequently heat sealed. Each standard and solubility solution was analyzed by UPLC/DAD. The solubility value was calculated by the following equation: Solubility = (Peak area of sample/ Peak area of standard) (Standard concentration) TABLE 1 The following table shows representative data for the compounds of the Examples. In this table, the FXIa Ki is a measure of the ability of the test compound to inhibit the action of the FXIa enzyme. Such results are indicative of the intrinsic activity of the compounds for use as inhibitors of the FXIa enzyme. Additionally Plasma Kallikrein (pKal) inhibition, tissue Kallikrein inhibition (tKal) and Norepenephrine Transporter functional antagonist uptake activity (NET) are provided. E xample FXIa pKal tKal NET I C50 (nM) IC50 (nM) IC50 (nM) IC50 (nM) armaco net c genera procedures: Rat cassette generic procedure: Plasma pharmacokinetic parameters for clearance, volume of distribution, half-life and mean residence time (MRT) were determined in rats from IV cassette administration studies.2 male rats typically weighing 225 – 260 gram, were fasted overnight prior to dosing. Compounds were prepared for IV dosing by addition to a vehicle, depending on the dose used. For a typical preparation, 1 mg per mL (IV) of up to 5 test compounds were added to vehicle comprised of 20% dimethyl sulfoxide (DMSO), 60% polyethylene glycol 400 (PEG400) and 20% water. IV formulation was administered to 2 rats via pre-cannulated jugular vein. Blood was collected by pre-cannulated artery, typically at predose, 2, 8, 15, 30 min, 1, 2, 4, 6, and 8 hr postdose. Samples were collected in K2EDTA tubes, stored on ice, and centrifuged. Plasma was transferred to a micro titer plate and stored at -70 °C until analysis. Plasma samples were extracted using protein precipitation and analyzed by liquid chromatography separation followed by mass spec detection (LCMS/MS), using a standard curve for each compound. Plasma pharmacokinetic parameters were calculated by non-compartmental methods. Rat screening IV/PO Generic procedure: Plasma pharmacokinetic parameters for clearance, volume of distribution, half-life, mean residence time (MRT) and oral bioavailability (%F) were determined in rats from oral administration and IV administration studies.4 male rats, typically weighing 225 – 260 gram, were fasted overnight prior to dosing. Compounds were prepared for oral and IV dosing by addition to a vehicle, depending on the dose used. For a typical preparation, 1 mg per mL (IV) or 1.5 mg per mL (oral) of test compound was added to vehicle comprised of 20% dimethyl sulfoxide (DMSO), 60% polyethylene glycol 400 (PEG400) and 20% water. IV formulation was administered to 2 rats via pre-cannulated jugular vein, and oral dosing was administered to 2 rats via oral gavage. Blood was collected by pre-cannulated artery, typically at predose, 2 min, 8 min, 15 min, 30 min, and 1, 2, 4, 6, and 8 hr postdose for IV, and at predose, 15 min, 30 min, and 1, 2, 4, 6, 8 hr for oral dosing. Samples were collected in K2EDTA tubes, stored on ice, and centrifuged. Plasma was transferred to a micro titer plate and stored at -70 °C until analysis. Plasma samples were extracted using protein precipitation and analyzed by liquid chromatography separation followed by mass spec detection (LCMS/MS), using a standard curve for each compound. Plasma pharmacokinetic parameters were calculated for IV and oral dosing data by non-compartmental methods. Oral bioavailability was determined as the ratio of the dose- normalized plasma area under the curve (AUC) following oral dosing vs. IV dosing. Dog screening IV/PO Generic procedure: Plasma pharmacokinetic parameters for clearance, volume of distribution, half-life, mean residence time (MRT) and oral bioavailability were determined in dogs from oral administration and IV administration studies.4 male dogs, typically weighing 8-12 kilograms, were fasted overnight prior to dosing. Compounds were prepared for oral and IV dosing by addition to a vehicle, depending on the dose used. For a typical preparation, 1 mg per mL (IV) or 1.5 mg per mL (oral) of test compound was added to vehicle comprised of 20% dimethyl sulfoxide (DMSO), 60% polyethylene glycol 400 (PEG400) and 20% water. IV formulation was administered to 2 dogs via the saphenous or cephalic vein, and oral dosing was administered to 2 dogs via oral gavage. Blood was collected by the cephalic or jugular vein, typically at predose, 2 min, 8 min, 15 min, 30 min, and 1, 2, 4, 6, 8 and 24 hr postdose for IV, and at predose, 15 min, 30 min, and 1, 2, 4, 6, 8, and 24 hr for oral dosing. Samples were collected in K2EDTA tubes, stored on ice, and centrifuged. Plasma was transferred to a micro titer plate and stored at -70 °C until analysis. Plasma samples were extracted using protein precipitation and analyzed by liquid chromatography separation followed by mass spec detection (LCMS/MS), using a standard curve for each compound. Plasma pharmacokinetic parameters were calculated for IV and oral dosing data by non-compartmental methods. Oral bioavailability was determined as the ratio of the dose- normalized plasma area under the curve (AUC) following oral dosing vs. IV dosing. Dog cassette generic procedure: Plasma pharmacokinetic parameters for clearance, volume of distribution, half-life, and mean residence time (MRT) were determined in dogs from IV cassette administration studies.2 male dogs, typically weighing 8-12 kilograms, were fasted overnight prior to dosing. Compounds were prepared for IV dosing by addition to a vehicle, depending on the dose used. For a typical preparation, 1 mg per mL (IV) of up to 5 test compounds were added to vehicle comprised of 20% dimethyl sulfoxide (DMSO), 60% polyethylene glycol 400 (PEG400) and 20% water. IV formulation was administered to 2 dogs via the saphenous or cephalic vein. Blood was collected by the cephalic or jugular vein, typically at predose, 2, 8, 15, 30 min, 1, 2, 4, 6, 8 and 24 hr postdose. Samples were collected in K2EDTA tubes, stored on ice, and centrifuged. Plasma was transferred to a micro titer plate and stored at -70 °C until analysis. Plasma samples were extracted using protein precipitation and analyzed by liquid chromatography separation followed by mass spec detection (LCMS/MS), using a standard curve for each compound. Plasma pharmacokinetic parameters were calculated by non-compartmental methods. TABLE 2 The following table shows representative data for the compounds of the Examples. In this table, select pharmacokinetic data (MRT= mean residence time) for both rat and dog are provided. Additionally the high-throughput solubility in pH2 phosphate buffer solution (PBS) and pH6.5 FaSSIF (Fasted State Simulated Intestinal Fluid) is provided. R at M pH 2 solubility pH 6.5 E xample RT Dog MRT (h) (FaSSIF) )