MITOCHONDRIAL TARGETING COMPOUNDS FOR THE TREATMENT OF ASSOCIATED DISEASES

Abstract
Mitochondrial targeting compounds for the treatment of cancer and other disorders associated with mitochondrial function, including diabetes, autoimmune diseases, inflammatory diseases, cardiovascular diseases and neurodegenerative diseases and their preparation. The present invention is also directed to the pharmaceutical compositions and treatment methods, prodrugs based on those compounds and the use thereof.
Description
FIELD OF THE INVENTION

The present invention is directed to the use and preparation of a series of mitochondrial targeted compounds inhibiting oxidative phosphorylation for the treatment of cancer and disorders associated with mitochondrial function. More specifically, the present invention is directed to a series of compounds for the treatment of cancers such as brain cancer, pancreatic cancer, ovarian cancer, renal cancer, breast cancer, prostate cancer, lung cancer, leukemia and lymphoma, as well as other disorders, such as diabetes, autoimmune diseases, inflammatory diseases, cardiovascular diseases and neurodegenerative diseases. The present invention is also directed to the pharmaceutical compositions and treatment methods based on these compounds.


BACKGROUND OF THE INVENTION

Metabolic reprogramming is an emerging hallmark of cancer and draws extensive attention in the field of drug discovery and pathological studies. Ever since the discovery of Warburg effect, researchers have been focusing on the glycolysis pathways, the most effective way to generate energy for cancer cells. However, recent studies highlight that tumors rely on oxidative phosphorylation (OXPHOS) for bioenergetics1-3 and, more importantly, biomass production4, 5, which is essential for enhanced tumor growth.


Consisting of over 90 proteins, OXPHOS complexes form one of the most important machinery in the mitochondria, linking the TCA cycle to the production of ATP. It is comprised of five complexes (Complex I to V). The complexes I-IV, also called the electron transport chain (ETC), transfer electrons from donors generated by the TCA cycle or fatty acid oxidation to oxygen. In the meantime, the energy released from the oxidation of NADH and FADH2 is used to pump protons across the inner membrane of the mitochondrion. This causes protons to build up in the intermembrane space and generates an electrochemical gradient across the membrane. The energy stored in this electrochemical gradient is then used by ATP synthase (complex V) to produce ATP. OXPHOS defect not only causes reduction in ATP production but also decreases the production of aspartate4, a limiting metabolite for cell proliferation.


OXPHOS is required for cancer cells to strive. Subpopulations of tumor cells are dependent on OXPHOS including, for example, glycolysis-deficient cells6 and SWI SNF complex mutated cells in lung cancer7. In addition, exacerbated OXPHOS dependency is frequently characterized by cancer stem cells8 9, as well as KRAS ablation-resistant cells in pancreatic cancers10. Importantly, OXPHOS inhibition is promising in overcoming resistance against chemotherapy11, 12 13 or tyrosine kinase inhibitors14. Thus, inhibition of OXPHOS might be a promising therapeutic strategy to treat various cancers.


Most OXPHOS inhibitors, including biguanides, oligomycin, and other toxins have been known for decades. As the most widely prescribed drug to treat patients with type II diabetes, metformin's mechanism of action remains partly unknown despite its use for over 60 years. Metformin and other biguanides are reported to exert their function as OXPHOS complex I inhibitors, leading to reduce tumorigenesis15-18. However, due to the inadequate potency in cancer cells, the usage of biguanides is largely limited. For other OXPHOS inhibitors, the unspecificity and poor drug-like properties blocked their potential usage in cancer treatment. Gboxin, an OXPHOS inhibitor, was reported to show specific inhibition of mouse and human glioblastoma cells through the depletion of the activity of FOF1ATP synthase19. However, it is still unclear that if this compound can cross blood-brain barrier hindering its path toward clinical trial. Other therapeutic compounds such as ME344, lonidamine and carboxyamidotriazoles are also claimed as OXPHOS inhibitors, however, all of them possess other primary drug targets and are considered as non-specific OXPHOS inhibitors. Recently, a potent and specific OXPHOS inhibitor, IACS-010759 (IACS) was reported and is currently being evaluated in phase I clinical trials. IACS targets OXPHOS complex I and shows significant efficacy in brain cancer and acute myeloid leukemia (AML) mouse xenografts20. Phase I trial of IACS indicated that it is well tolerated with preliminary evidence of antitumor activity. Loss of ENO1 or mutations in SMARCA4, which result in high dependency on OXPHOS, are used as predictive biomarkers of sensitivity to OXPHOS inhibition. In addition, the clinical usage of IACS in ibrutinib-resistant MCL will be evaluated. Overall, the discovery and development of OXPHOS inhibitors is a largely unexplored but promising field in cancer treatment.


Tumor progression is profoundly influenced by the interaction of cancer cells with their surrounding microenvironment, especially the infiltrating immune cells. The differentiation and functions of effector CD8+ T cells, Th1, Th2, and Th17 CD4+ T cells rely on the engagement of aerobic glycolysis21, and antitumor M1 macrophages22 also rely on glycolysis. Inhibition of OXPHOS leads to the upregulation of lactate secretion, leading to upregulation of glycolysis. It might potentially lead to the activation of cytotoxic T cells and M1 macrophages, resulting in improved antitumor engagement of immune cells. On the other hand, immunosuppressive M2 macrophages and other protumor cells, including regulatory T cells and myeloid-derived suppressor cells21-23, depend on various mitochondrial functions including OXPHOS. Thus, in addition to direct targeting of tumor cells, inhibition of OXPHOS might indirectly boost anti-tumor effect through the modulation of tumor microenvironments.


Autoimmune and inflammatory diseases are diverse conditions caused by inappropriate and prolonged activation of immune cells with associated ongoing production of inflammatory mediators that cause tissue damage. Immune cell activation and differentiation occurs concurrently with metabolic reprogramming. This ensures activated cells to generate the energy and substrates necessary to perform their specified functions. It's reported that alternatively activated M2 macrophages and Tregs rely on oxidative phosphorylation to provide their energy24, 25. In a mouse model of bone marrow allotransplant, proliferating bone marrow cells reconstituting the immune system of a lethally irradiated syngeneic host underwent a dramatically different metabolic process than pathogenic cells, with healthy cells relying more on glycolysis than oxidative phosphorylation26. OXPHOS inhibitors have demonstrated promise as a metabolic therapy for graft-versus-host disease (GVHD)26, 27. Similar to pathogenic T cells in GVHD, and in contrast with the lymphocytes activated under normal conditions or in the case of solid organ transplant, CD4 T cells in systemic lupus erythematosus (SLE) meet their energetic needs mostly through oxidative phosphorylation28. Targeting metabolic pathways through inhibition of OXPHOS could lead to selective regulation of immune system to fight various diseases.


Disclosed herein are new compounds selectively inhibiting complex I of the mitochondrial electron transport chain to disrupt OXPHOS. The hit compound is selected from a phenotypical screening of a library of 24,000 compounds. These compounds show profound anti-tumor effect as a single agent and have synergistic effect with radiation and select FDA-approved drugs as well as drugs under clinical trials. Therefore, OXPHOS selective drugs can be used as single agent and in combination to treat various cancers as well as other diseases related to OXPHOS and immune modulation.


SUMMARY OF THE INVENTION

Disclosed herein is a series of novel compounds as mitochondrial modulators that can be used as treatment of diseases associated with mitochondrial functions, including, but not limited to, cancer, inflammatory disease and diabetes.


Accordingly, in one aspect, the present invention features a series of compounds of Formula 1.




embedded image


Such that Q is




embedded image


Such that


n=0-4;


p=0-2;


q=0 or 1;


r=1 or 2;


A is CH, or when V is CR6R7 and p is 2 or q is 1, may also be NR8, O or S;


T is ═O or ═NR6;


U is U1—U2,


B is N or CR8


Wherein


U1 is a bond, C═O or SO2;


U2 is R9, OR10, NHR11, NR11R12, NR11-(4-6-ring azacycloalkyl), 1-piperazinyl, 1-homopiperazinyl, “C”7-10 N-linked-1,x′-diaza-(spiro/fused/bridged)bicycloalkyl, where x′ is 4 or greater, wherein up to four carbon atoms of U2 may be independently substituted with R13, and each of the remaining N atom may be substituted with R14;


V is CR6R7, O, S or NR8.


W1 and W2 are independently selected from CH and N, or the two may be taken together as S or NR8;


X and Y are independently selected from CH and N


Z is selected from a direct bond, —N(R16)—, —N(R16)C(O)—, —O—, —C(O)—, —C(S)—, —S(O)t— (where t is 0, 1 or 2), —S(O)(N(R16))— and P(O);


R1 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R2 and R3 are independently hydrogen, C1-C8 alkyl, C3-C8 carbocyclyl, C3-C8 heterocyclyl, or aryl; or R2 and R3, together with the nitrogen to which they are joined form C3-C8 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R2 and R3 may be independently substituted with R13;


R4 is selected from hydrogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, carboxy, amino, acylamino, aryloxy, heteroaryloxy and C1-C4 alkoxy;


R5 is selected from hydrogen, halogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R6 and R7 are independently hydrogen or lower C1-4 alkyl, or, if R4, is not H, halogen, or OH or lower C1-4 alkoxy, or R6 and R7 taken together may be oxo or lower C1-4 alkylidene;


R8 is selected from hydrogen, cyano, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, aralkyl, alkoxycarbonyl, carbamoyl, acyl and sulfonyl.


R9 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R10 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R11 and R12 are independently hydrogen, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, arylalkyl or heteroarylalkyl; or R11 and R12, together with the nitrogen to which they are joined form C3-C10 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R11 and R12 may be independently substituted with R13;


R13 is selected from hydrogen, halogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R14 is H, lower C1-4 alkyl, C(═O)-lower C1-4 alkyl, C(═O)-lower C1-4 alkoxy, S(═O)2-lower C1-4 alkyl, 5-tetrazyl, 5-oxo-1,24-oxadiazol-3-yl, isoxazol-5-yl, 1,2,4-oxadiazol-5-yl, 1,2,4-oxadiazol-3-yl, phenyl, 2/3/4-pyridinyl, 2/4/5-pyrimidyl, pyrazinyl, 3/4-pyridazinyl, wherein any of the aromatic groups may be substituted with one R15.


R15 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy,


R16 is selected from hydrogen, C1-C8 alkyl, C3-C8 carbocyclyl, C3-C8 heterocyclyl, aryl, heteroaryl, arylalkyl and heteroarylalkyl.


In one embodiment, the invention features compounds of Formula 1-1.




embedded image


Such that


n=0-4;


T1 and T2 are independently ═O or ═NR6;


U2 is R9, OR10, NHR11, NR11R12, NR11-(4-6-ring azacycloalkyl), 1-piperazinyl, 1-homopiperazinyl, “C”7-10 N-linked-1,x′-diaza-(spiro/fused/bridged)bicycloalkyl, where x′ is 4 or greater, wherein up to four carbon atoms of U2 may be independently substituted with R13, and each of the remaining N atom may be substituted with R14;


W1 and W2 are independently selected from CH and N, or the two may be taken together as S or NR8;


X and Y are independently selected from CH and N;


B is N or CR8;


R1 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R2 and R3 are independently hydrogen, C1-C8 alkyl, C3-C8 carbocyclyl, C3-C8 heterocyclyl, or aryl; or R2 and R3, together with the nitrogen to which they are joined form C3-C8 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R2 and R3 may be independently substituted with R13;


R4 is selected from hydrogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, carboxy, amino, acylamino, aryloxy, heteroaryloxy and C1-C4 alkoxy;


R6 is hydrogen or lower C1-4 alkyl or OH or lower C1-4 alkoxy;


R8 is selected from hydrogen, cyano, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, aralkyl, alkoxycarbonyl, carbamoyl, acyl and sulfonyl.


R9 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R10 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R11 and R12 are independently hydrogen, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, arylalkyl or heteroarylalkyl; or R11 and R12, together with the nitrogen to which they are joined form C3-C10 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R11 and R12 may be independently substituted with R13;


R13 is selected from hydrogen, halogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R14 is H, lower C1-4 alkyl, C(═O)-lower C1-4 alkyl, C(═O)-lower C1-4 alkoxy, S(═O)2-lower C1-4 alkyl, 5-tetrazyl, 5-oxo-1,24-oxadiazol-3-yl, isoxazol-5-yl, 1,2,4-oxadiazol-5-yl, 1,2,4-oxadiazol-3-yl, phenyl, 2/3/4-pyridinyl, 2/4/5-pyrimidyl, pyrazinyl, 3/4-pyridazinyl, wherein any of the aromatic groups may be substituted with one R15.


R15 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy.


In a further embodiment, the invention features compounds of Formula 1-1-1.




embedded image


Such that


n=0-4;


T1 and T2 are independently ═O or ═NR6;


U3 is selected from the following groups:




embedded image


embedded image


wherein either side of the can be attached to the carbonyl of the core structure, and the other nitrogen attached to U4, wherein n1, n2, n3 and n4 are independently 0, 1, 2 or 3;


Wherein U4 is selected from the following groups:




embedded image


W1 and W2 are independently selected from CH and N, or the two may be taken together as S or NR8;


X and Y are independently selected from CH and N;


B is N or CR8;


R1 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R2 and R3 are independently hydrogen, C1-C8 alkyl, C3-C8 carbocyclyl, C3-C8 heterocyclyl, or aryl; or R2 and R3, together with the nitrogen to which they are joined form C3-C8 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R2 and R3 may be independently substituted with R13.


R4 is selected from hydrogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, carboxy, amino, acylamino, aryloxy, heteroaryloxy and C1-C4 alkoxy;


R6 is hydrogen or lower C1-4 alkyl or OH or lower C1-4 alkoxy;


R8 is selected from hydrogen, cyano, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, aralkyl, alkoxycarbonyl, carbamoyl, acyl and sulfonyl.


R9 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R10 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R11 and R12 are independently hydrogen, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, arylalkyl or heteroarylalkyl; or R11 and R12, together with the nitrogen to which they are joined form C3-C10 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R11 and R12 may be independently substituted with R13;


R13 is selected from hydrogen, halogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, aryl, heteroaryl, hydroxy, C1-C4 alkoxy, carboxy and amino;


R16 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, hydroxy, carboxy, amino and C1-C4 alkoxy.


In another further embodiment, the invention features compounds of Formula 1-1-2.




embedded image


Such that


n=0-4;


T1 and T2 are independently ═O or ═NR6;


U5 is selected from the following groups:




embedded image


wherein n1, n2, n3, n4 and n7 are independently 0, 1, 2 or 3, n5 and n6 are independently 1, 2 or 3;


W1 and W2 are independently selected from CH and N, or the two may be taken together as S or NR8;


X and Y are independently selected from CH and N;


B is N or CR8;


R1 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R2 and R3 are independently hydrogen, C1-C8 alkyl, C3-C8 carbocyclyl, C3-C8 heterocyclyl, or aryl; or R2 and R3, together with the nitrogen to which they are joined form C3-C8 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R2 and R3 may be independently substituted with R13;


R4 is selected from hydrogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, carboxy, amino, acylamino, aryloxy, heteroaryloxy and C1-C4 alkoxy;


R6 is hydrogen or lower C1-4 alkyl or OH or lower C1-4 alkoxy;


R8 is selected from hydrogen, cyano, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, aralkyl, alkoxycarbonyl, carbamoyl, acyl and sulfonyl.


R10 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R13 is selected from hydrogen, halogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, aryl, heteroaryl, hydroxy, C1-C4 alkoxy, carboxy and amino.


In another embodiment, the invention features compounds of Formula 1-2.




embedded image


Such that


n=0-4;


T is ═O or ═NR6;


U2 is R9, OR10, NHR11, NR11R12, NR11-(4-6-ring azacycloalkyl), 1-piperazinyl, 1-homopiperazinyl, “C”7-10 N-linked-1,x′-diaza-(spiro/fused/bridged)bicycloalkyl, where x′ is 4 or greater, wherein up to four carbon atoms of U2 may be independently substituted with R13, and each of the remaining N atom may be substituted with R14;


W1 and W2 are independently selected from CH and N, or the two may be taken together as S or NR8;


X and Y are independently selected from CH and N;


R1 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R2 and R3 are independently hydrogen, C1-C8 alkyl, C3-C8 carbocyclyl, C3-C8 heterocyclyl, or aryl; or R2 and R3, together with the nitrogen to which they are joined form C3-C8 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R2 and R3 may be independently substituted with R13;


R4 is selected from hydrogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, carboxy, amino, acylamino, aryloxy, heteroaryloxy and C1-C4 alkoxy;


R6 is hydrogen or lower C1-4 alkyl or OH or lower C1-4 alkoxy;


R8 is selected from hydrogen, cyano, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, aralkyl, alkoxycarbonyl, carbamoyl, acyl and sulfonyl.


R9 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R10 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R11 and R12 are independently hydrogen, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, arylalkyl or heteroarylalkyl; or R11 and R12, together with the nitrogen to which they are joined form C3-C10 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R11 and R12 may be independently substituted with R13;


R13 is selected from hydrogen, halogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R14 is H, lower C1-4 alkyl, C(═O)-lower C1-4 alkyl, C(═O)-lower C1-4 alkoxy, S(═O)2-lower C1-4 alkyl, 5-tetrazyl, 5-oxo-1,24-oxadiazol-3-yl, isoxazol-5-yl, 1,2,4-oxadiazol-5-yl, 1,2,4-oxadiazol-3-yl, phenyl, 2/3/4-pyridinyl, 2/4/5-pyrimidyl, pyrazinyl, 3/4-pyridazinyl, wherein any of the aromatic groups may be substituted with one R15.


R15 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy.


In another embodiment, the invention features compounds of Formula 1-3.




embedded image


Such that


n=0-4;


T1 and T2 are independently ═O or ═NR6;


Wherein U6 is selected from the following groups:




embedded image


W1 and W2 are independently selected from CH and N, or the two may be taken together as S or NR8;


X and Y are independently selected from CH and N;


B is N or CR8;


R1 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R2 and R3 are independently hydrogen, C1-C8 alkyl, C3-C8 carbocyclyl, C3-C8 heterocyclyl, or aryl; or R2 and R3, together with the nitrogen to which they are joined form C3-C8 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R2 and R3 may be independently substituted with R13;


R4 is selected from hydrogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, carboxy, amino, acylamino, aryloxy, heteroaryloxy and C1-C4 alkoxy;


R6 is hydrogen or lower C1-4 alkyl or OH or lower C1-4 alkoxy;


R8 is selected from hydrogen, cyano, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, aralkyl, alkoxycarbonyl, carbamoyl, acyl and sulfonyl.


R9 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R11 and R12 are independently hydrogen, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, arylalkyl or heteroarylalkyl; or R11 and R12, together with the nitrogen to which they are joined form C3-C10 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R11 and R12 may be independently substituted with R13;


R13 is selected from hydrogen, halogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R16 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, hydroxy, carboxy, amino and C1-C4 alkoxy.


In another embodiment of the invention, there is provided a compound selected from the compounds as shown in Table 1.


The invention provides the stereochemical mixtures or pure forms for all compounds of invention.


The invention also provides all pharmaceutically acceptable salts, esters, amides, tautomers, geometric isomers, solvates thereof, as well as pharmaceutical composition comprising an effective amount of a compound of the invention and a pharmaceutically acceptable carrier. The composition may further comprise an effective amount of one or more other agents for treating cancer or a disorder associated with mitochondrial function.


The invention further provides prodrugs of all compounds of invention. The term “prodrug” used herein refers to a pharmacologically inactive derivative of a parent “drug” molecule which requires biotransformation within the target physiological system to release, or to convert the prodrug into the active drug. Prodrugs can address the problems associated with solubility, stability, cell permeability or bioavailability. Prodrugs usually comprise an active drug molecule and a chemical masking group. Prodrugs can be readily prepared from the parent compounds with well-known methods.


Furthermore, the invention provides a packaged product comprising a container; an effective amount of a compound of invention.


The invention also provides methods of preparing the compounds of invention.


Definitions

Certain terms employed in the specification, examples, and appended claims are further described here in the present invention. These definitions should be read in light of the entire invention and as would be understood by a person skilled in the art.


“Cycloalkyl” refers to a saturated hydrocarbon ring that is not aromatic. Cycloalkyl rings are monocyclic, or are fused, spiro, or bridged bicyclic or polycyclic ring systems. Monocyclic cycloalkyl rings contain from about 3 to about 12 carbon atoms, preferably from 3 to 7 carbon atoms, in the ring. Bicyclic cycloalkyl rings contain from 7 to 17 carbon atoms, preferably from 7 to 12 carbon atoms, in the ring. Preferred bicyclic cycloalkyl rings comprise 4-, 5-, 6- or 7-membered rings fused to 5-, 6- or 7-membered rings. Cycloalkyl rings may be unsubstituted or substituted with from 1 to 4 substituents on the ring. Cycloalkyl may be substituted with halogen, cyano, nitro, alkyl, heteroalkyl, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, alkoxy, arylalkyl, heteroarylalkyl or any combination thereof. Preferred cycloalkyl rings include cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl and cyclononyl rings.


“Heterocycloalkyl” is a saturated or unsaturated ring containing carbon atoms and from 1 to 4 (preferably 1 to 3) heteroatoms in the ring. Heterocycloalkyl rings are monocyclic, or are fused, spiro, or bridged bicyclic or polycyclic ring systems. Monocyclic heterocycloalkyl rings contain from about 3 to about 9 member atoms (including both carbons and heteroatoms), preferably from 5 to 7 member atoms, in the ring. Bicyclic heterocycloalkyl rings may be fused, spiro, or bridged ring systems. Preferred bicyclic heterocycloalkyl rings comprise 5-, 6- or 7-membered rings fused to 5-, 6- or 7-membered rings. Heterocycloalkyl rings may be unsubstituted (i.e., contain hydrogen) or substituted (on either carbons or heteroatoms or both) with from 1 to 4 substituents selected from halogen, cyano, nitro, alkyl, heteroalkyl, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, alkoxy, arylalkyl, heteroarylalkyl or any combination thereof.


“Aryl” refers to aromatic monocyclic or multicyclic groups containing from 3 to 16 carbon atoms. Aryl may be unsubstituted or substituted with from 1 to 4 substituents. Preferred substituted aryls are mono-, di, or tri-substituted. Aryls may be substituted with halogen, cyano, nitro, alkyl, heteroalkyl, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, alkoxy, arylalkyl, heteroarylalkyl or any combination thereof.


“Heteroaryl” refers to a monocyclic or multicyclic aromatic ring system, of about 5 to about 15 members where one or more of the atoms in the ring system is a heteroatom, that is, an element other than carbon, including but not limited to nitrogen, oxygen or sulfur. The heteroaryl group may be optionally fused to a benzene ring. Heteroaryl may be unsubstituted or substituted with from 1 to 4 substituents. Preferred substituted heteroaryls are mono-, di, or tri-substituted. Heteroaryls may be substituted with halogen, cyano, nitro, alkyl, heteroalkyl, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, alkoxy, arylalkyl, heteroarylalkyl or any combination thereof.


“Halo” or “halogen” is fluoro, chloro, bromo or iodo.


“Alkyl” means a saturated hydrocarbon radical having 1 to 15 carbon atoms, preferably 1 to 10 carbon atoms, more preferably 1 to 5 carbon atoms, most preferably 1 to 3 carbon atoms, that may be branched or unbranched. Non-limiting example of alkyl radicals include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, hexyl, heptyl, octyl and the like, wherein methyl, ethyl, n-propyl and isopropyl represent specifically preferred examples.


“Heteroalkyl” is a saturated or unsaturated chain carbon and at least one heteroatom, wherein no two heteroatoms are adjacent. Heteroalkyl chains contain from 2 to 15 member atoms (carbon and heteroatoms) in the chain, preferably 2 to 10, more preferably 2 to 5. For example, alkoxy (i.e., —O-alkyl or —O-heteroalkyl) radicals are included in heteroalkyl. Heteroalkyl chains may be straight or branched. Preferred branched heteroalkyl have one or two branches, preferably one branch. Preferred heteroalkyl are saturated. Unsaturated heteroalkyl have one or more carbon-carbon double bounds and/or one or more carbon-carbon triple bounds. Preferred unsaturated heteroalkyl have one or two carbon-carbon double bounds or one carbon-carbon triple bound, more preferably one double bound. Heteroalkyl chains may be unsubstituted or substituted with from 1 to 4 substituents. Preferred substituted heteroalkyl are mono-, di, or tri-substituted. Heteroalkyl may be substituted with halogen, cyano, nitro, alkyl, heteroalkyl, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, alkoxy, arylalkyl, heteroarylalkyl or any combination thereof.


“Alkoxy” means an oxygen radical having a hydrocarbon chain substituent, where the hydrocarbon chain is an alkyl or alkenyl (i.e., —O-alkyl or —O-alkenyl). Examples of alkoxy include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, sec-butoxy, tert-butoxy, allyloxy and the like.


“Arylalkyl” alone or in combination, refers to an alkyl radical in which one hydrogen atom is replaced by an aryl radical, for example, benzyl and the like.


“Heteroarylalkyl” refers to an alkyl radical in which one hydrogen atom is replaced by a heteroaryl radical.


“Independently” groups are groups present in the same structure that need not all represent the same substitution.


“Pharmacological composition” refers to a mixture of one or more of the compounds described herein or pharmaceutically acceptable salts thereof, with other chemical components, such as pharmaceutically acceptable carriers and/or excipients. The purpose of a pharmacological composition is to facilitate administration of a compound to an organism.


“Pharmaceutically acceptable salts” is a cationic salt formed at any acidic (e.g., carboxylic acid) group, or an anionic salt formed at any basic (e.g., amino) group.


“Solvate” is a physical association of a compound of the invention with one or more solvent molecules, whether organic or inorganic. This physical association often includes hydrogen bonding. In certain instances, the solvate is capable of isolation, for example, when one or more solvate molecules are incorporated in the crystal lattice of the crystalline solid. “Solvate” encompasses both solution-phase and isolable solvates. Exemplary solvates include hydrates, ethanolates, and methanolates.


“Prodrug” refers to a pharmacologically inactive derivative of a parent “drug” molecule which requires biotransformation within the target physiological system to release, or to convert the prodrug into the active drug. Prodrugs can address the problems associated with solubility, stability, cell permeability or bioavailability. Prodrugs usually comprise an active drug molecule and a chemical masking group. Prodrugs can be readily prepared from the parent compounds with well-known methods.







DETAILED DESCRIPTION OF THE INVENTION

Disclosed herein is a series of novel compounds as mitochondrial modulators that can be used as treatment of diseases associated with mitochondrial functions, including, but not limited to, cancer, inflammatory disease and diabetes.


Accordingly, in one aspect, the present invention features a series of compounds of Formula 1.




embedded image


Such that Q is




embedded image


Such that


n=0-4;


p=0-2;


q=0 or 1;


r=1 or 2;


A is CH, or when V is CR6R7 and p is 2 or q is 1, may also be NR8, O or S;


T is ═O or ═NR6;


U is U1—U2,


Wherein


U1 is a bond, C═O or SO2;


U2 is R9, OR10, NHR11, NR11R12, NR11-(4-6-ring azacycloalkyl), 1-piperazinyl, 1-homopiperazinyl, “C”7-10 N-linked-1,x′-diaza-(spiro/fused/bridged)bicycloalkyl, where x′ is 4 or greater, wherein up to four carbon atoms of U2 may be independently substituted with R13, and each of the remaining N atom may be substituted with R14;


V is CR6R7, O, S or NR8.


W1 and W2 are independently selected from CH and N, or the two may be taken together as S or NR8;


X and Y are independently selected from CH and N


Z is selected from a direct bond, —N(R16)—, —N(R16)C(O)—, —O—, —C(O)—, —C(S)—, —S(O)t— (where t is 0, 1 or 2) and —S(O)(N(R16))—;


B is N or CR8;


R1 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R2 and R3 are independently hydrogen, C1-C8 alkyl, C3-C8 carbocyclyl, C3-C8 heterocyclyl, or aryl; or R2 and R3, together with the nitrogen to which they are joined form C3-C8 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R2 and R3 may be independently substituted with R13;


R4 is selected from hydrogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, carboxy, amino, acylamino, aryloxy, heteroaryloxy and C1-C4 alkoxy;


R5 is selected from hydrogen, halogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R6 and R7 are independently hydrogen or lower C1-4 alkyl, or, if R4, is not H, halogen, or OH or lower C1-4 alkoxy, or R6 and R7 taken together may be oxo or lower C1-4 alkylidene;


R8 is selected from hydrogen, cyano, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, aralkyl, alkoxycarbonyl, carbamoyl, acyl and sulfonyl.


R9 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R10 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R11 and R12 are independently hydrogen, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, arylalkyl or heteroarylalkyl; or R11 and R12, together with the nitrogen to which they are joined form C3-C10 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R11 and R12 may be independently substituted with R13;


R13 is selected from hydrogen, halogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R14 is H, lower C1-4 alkyl, C(═O)-lower C1-4 alkyl, C(═O)-lower C1-4 alkoxy, S(═O)2-lower C1-4 alkyl, 5-tetrazyl, 5-oxo-1,24-oxadiazol-3-yl, isoxazol-5-yl, 1,2,4-oxadiazol-5-yl, 1,2,4-oxadiazol-3-yl, phenyl, 2/3/4-pyridinyl, 2/4/5-pyrimidyl, pyrazinyl, 3/4-pyridazinyl, wherein any of the aromatic groups may be substituted with one R15.


R15 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy,


R16 is selected from hydrogen, C1-C8 alkyl, C3-C8 carbocyclyl, C3-C8 heterocyclyl, aryl, heteroaryl, arylalkyl and heteroarylalkyl.


In one embodiment, the invention features compounds of Formula 1-1.




embedded image


Such that


n=0-4;


T1 and T2 are independently ═O or ═NR6;


U2 is R9, OR10, NHR11, NR11R12, NR11-(4-6-ring azacycloalkyl), 1-piperazinyl, 1-homopiperazinyl, “C”7-10 N-linked-1,x′-diaza-(spiro/fused/bridged)bicycloalkyl, where x′ is 4 or greater, wherein up to four carbon atoms of U2 may be independently substituted with R13, and each of the remaining N atom may be substituted with R14;


W1 and W2 are independently selected from CH and N, or the two may be taken together as S or NR8;


X and Y are independently selected from CH and N;


B is N or CR8;


R1 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R2 and R3 are independently hydrogen, C1-C8 alkyl, C3-C8 carbocyclyl, C3-C8 heterocyclyl, or aryl; or R2 and R3, together with the nitrogen to which they are joined form C3-C8 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R2 and R3 may be independently substituted with R13;


R4 is selected from hydrogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, carboxy, amino, acylamino, aryloxy, heteroaryloxy and C1-C4 alkoxy;


R6 is hydrogen or lower C1-4 alkyl or OH or lower C1-4 alkoxy;


R8 is selected from hydrogen, cyano, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, aralkyl, alkoxycarbonyl, carbamoyl, acyl and sulfonyl.


R9 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R10 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R11 and R12 are independently hydrogen, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, arylalkyl or heteroarylalkyl; or R11 and R12, together with the nitrogen to which they are joined form C3-C10 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R11 and R12 may be independently substituted with R13.


R13 is selected from hydrogen, halogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R14 is H, lower C1-4 alkyl, C(═O)-lower C1-4 alkyl, C(═O)-lower C1-4 alkoxy, S(═O)2-lower C1-4 alkyl, 5-tetrazyl, 5-oxo-1,24-oxadiazol-3-yl, isoxazol-5-yl, 1,2,4-oxadiazol-5-yl, 1,2,4-oxadiazol-3-yl, phenyl, 2/3/4-pyridinyl, 2/4/5-pyrimidyl, pyrazinyl, 3/4-pyridazinyl, wherein any of the aromatic groups may be substituted with one R15.


R15 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy.


In a further embodiment, the invention features compounds of Formula 1-1-1.




embedded image


Such that


n=0-4;


T1 and T2 are independently ═O or ═NR6;


U3 is selected from the following groups:




embedded image


wherein either side of the can be attached to the carbonyl of the core structure, and the other nitrogen attached to U4, wherein n1, n2, n3 and n4 are independently 0, 1, 2 or 3;


Wherein U4 is selected from the following groups:




embedded image


W1 and W2 are independently selected from CH and N, or the two may be taken together as S or NR8;


X and Y are independently selected from CH and N;


B is N or CR8;


R1 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R2 and R3 are independently hydrogen, C1-C8 alkyl, C3-C8 carbocyclyl, C3-C8 heterocyclyl, or aryl; or R2 and R3, together with the nitrogen to which they are joined form C3-C8 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R2 and R3 may be independently substituted with R13.


R4 is selected from hydrogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, carboxy, amino, acylamino, aryloxy, heteroaryloxy and C1-C4 alkoxy;


R6 is hydrogen or lower C1-4 alkyl or OH or lower C1-4 alkoxy;


R8 is selected from hydrogen, cyano, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, aralkyl, alkoxycarbonyl, carbamoyl, acyl and sulfonyl.


R9 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R10 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R11 and R12 are independently hydrogen, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, arylalkyl or heteroarylalkyl; or R11 and R12, together with the nitrogen to which they are joined form C3-C10 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R11 and R12 may be independently substituted with R13;


R13 is selected from hydrogen, halogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, aryl, heteroaryl, hydroxy, C1-C4 alkoxy, carboxy and amino;


R16 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, hydroxy, carboxy, amino and C1-C4 alkoxy.


In another further embodiment, the invention features compounds of Formula 1-1-2.




embedded image


Such that


n=0-4;


T1 and T2 are independently ═O or ═NR6;


U5 is selected from the following groups:




embedded image


wherein n1, n2, n3, n4 and n7 are independently 0, 1, 2 or 3, n5 and n6 are independently 1, 2 or 3;


W1 and W2 are independently selected from CH and N, or the two may be taken together as S or NR8;


X and Y are independently selected from CH and N;


B is N or CR8;


R1 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R2 and R3 are independently hydrogen, C1-C8 alkyl, C3-C8 carbocyclyl, C3-C8 heterocyclyl, or aryl; or R2 and R3, together with the nitrogen to which they are joined form C3-C8 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R2 and R3 may be independently substituted with R13;


R4 is selected from hydrogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, carboxy, amino, acylamino, aryloxy, heteroaryloxy and C1-C4 alkoxy;


R6 is hydrogen or lower C1-4 alkyl or OH or lower C1-4 alkoxy;


R8 is selected from hydrogen, cyano, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, aralkyl, alkoxycarbonyl, carbamoyl, acyl and sulfonyl.


R10 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R13 is selected from hydrogen, halogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, aryl, heteroaryl, hydroxy, C1-C4 alkoxy, carboxy and amino.


In another embodiment, the invention features compounds of Formula 1-2.




embedded image


Such that


n=0-4;


T is ═O or ═NR6;


U2 is R9, OR10, NHR11, NR11R12, NR11-(4-6-ring azacycloalkyl), 1-piperazinyl, 1-homopiperazinyl, “C”7-10 N-linked-1,x′-diaza-(spiro/fused/bridged)bicycloalkyl, where x′ is 4 or greater, wherein up to four carbon atoms of U2 may be independently substituted with R13, and each of the remaining N atom may be substituted with R14;


W1 and W2 are independently selected from CH and N, or the two may be taken together as S or NR8;


X and Y are independently selected from CH and N;


R1 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R2 and R3 are independently hydrogen, C1-C8 alkyl, C3-C8 carbocyclyl, C3-C8 heterocyclyl, or aryl; or R2 and R3, together with the nitrogen to which they are joined form C3-C8 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R2 and R3 may be independently substituted with R13;


R4 is selected from hydrogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, carboxy, amino, acylamino, aryloxy, heteroaryloxy and C1-C4 alkoxy;


R6 is hydrogen or lower C1-4 alkyl or OH or lower C1-4 alkoxy;


R8 is selected from hydrogen, cyano, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, aralkyl, alkoxycarbonyl, carbamoyl, acyl and sulfonyl.


R9 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R10 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R11 and R12 are independently hydrogen, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, arylalkyl or heteroarylalkyl; or R11 and R12, together with the nitrogen to which they are joined form C3-C10 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R11 and R12 may be independently substituted with R13;


R13 is selected from hydrogen, halogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R14 is H, lower C1-4 alkyl, C(═O)-lower C1-4 alkyl, C(═O)-lower C1-4 alkoxy, S(═O)2-lower C1-4 alkyl, 5-tetrazyl, 5-oxo-1,24-oxadiazol-3-yl, isoxazol-5-yl, 1,2,4-oxadiazol-5-yl, 1,2,4-oxadiazol-3-yl, phenyl, 2/3/4-pyridinyl, 2/4/5-pyrimidyl, pyrazinyl, 3/4-pyridazinyl, wherein any of the aromatic groups may be substituted with one R15.


R15 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy.


In another embodiment, the invention features compounds of Formula 1-3.




embedded image


Such that


n=0-4;


T1 and T2 are independently ═O or ═NR6;


Wherein U6 is selected from the following groups:




embedded image


W1 and W2 are independently selected from CH and N, or the two may be taken together as S or NR8;


X and Y are independently selected from CH and N;


B is N or CR8;


R1 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R2 and R3 are independently hydrogen, C1-C8 alkyl, C3-C8 carbocyclyl, C3-C8 heterocyclyl, or aryl; or R2 and R3, together with the nitrogen to which they are joined form C3-C8 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R2 and R3 may be independently substituted with R13;


R4 is selected from hydrogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, carboxy, amino, acylamino, aryloxy, heteroaryloxy and C1-C4 alkoxy;


R6 is hydrogen or lower C1-4 alkyl or OH or lower C1-4 alkoxy;


R8 is selected from hydrogen, cyano, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, aralkyl, alkoxycarbonyl, carbamoyl, acyl and sulfonyl.


R9 is selected from hydrogen, C1-C4 alkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl and aralkyl;


R11 and R12 are independently hydrogen, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, arylalkyl or heteroarylalkyl; or R11 and R12, together with the nitrogen to which they are joined form C3-C10 heterocyclyl, which for the 6 and 7 membered rings may include an extra heteroatom chosen from O, S, SO, SO2 and NR8, and wherein up to four carbon of R11 and R12 may be independently substituted with R13;


R13 is selected from hydrogen, halogen, cyano, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, aryl, heteroaryl, keto, hydroxy, carboxy, amino, acylamino, aryloxy, heteroaryloxy, C1-C4 alkoxy, arylalkyl, and heteroarylalkyl;


R16 is selected from hydrogen, halogen, cyano, nitro, C1-C4 alkyl, C1-C4 heteroalkyl, C1-C4haloalkyl, hydroxy, carboxy, amino and C1-C4 alkoxy.


In another embodiment of the invention, there is provided a compound selected from the compounds as shown in Table 1:













TABLE 1





Code
Structure
IUPAC name
M.W.
cLogP







DX2- 201


embedded image


ethyl (R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxylate
432.55
  2.99





DX2- 202


embedded image


ethyl (S)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxylate
432.55
  2.99





DX2- 208


embedded image


ethyl 1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-4-carboxylate
432.55
  2.17





DX2- 209


embedded image


ethyl 1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-2-carboxylate
432.55
  3.30





DX2- 217


embedded image


ethyl (R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) pyrrolidine-3-carboxylate
418.53
  2.43





DX2- 218


embedded image


ethyl (S)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) pyrrolidine-3-carboxylate
418.53
  2.43





DX2- 219


embedded image


ethyl 1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) azetidine-3-carboxylate
404.50
  2.68





DX2- 221


embedded image


ethyl 1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) azepane-4-carboxylate
446.58
  2.73





DX2- 225


embedded image


ethyl (3R)-1-(4-(N,N- diethylsulfamoyl)phenyl- sulfonimidoyl) piperidine-3-carboxylate
431.57
  3.25





DX2- 226


embedded image


ethyl (R)-1-((2-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxylate
432.55
  2.99





DX2- 229


embedded image


ethyl (R)-1-((3-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxylate
432.55
  2.99





DX2- 230


embedded image


ethyl (R)-1-((4-(4-(N,N- diethylsulfamoyl)phenoxy)phenyl) sulfonyl)piperidine-3-carboxylate
524.65
  5.08





DX2- 235


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxylic acid
404.50
  1.99





DX2- 237


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N,N-diethylpiperidine-3- carboxamide
459.62
  2.93





DX2- 238


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-methylpiperidine-3-carboxamide
417.54
  1.49





DX2- 241


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-(2- (dimethylamino)ethyl)piperidine-3- carboxamide
474.64
  2.13





DX2- 242


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-(oxetan-3-yl)piperidine-3- carboxamide
459.58
  2.05





DX2- 244


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-(4-fluorophenyl)piperidine-3- carboxamide
497.60
  3.85





DX2- 245


embedded image


ethyl (R)-1-((4- (morpholinosulfonyl)phenyl) sulfonyl)piperidine-3- carboxylate
446.54
  1.87





DX2- 246


embedded image


ethyl (R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- 3-methylpiperidine-3-carboxylate
446.58
  3.50





DX2- 248


embedded image


ethyl (R)-1-((4-(piperazin-1- ylsulfonyl)phenyl)sulfonyl) piperidine-3-carboxylate
445.55
  1.86





DX2- 249


embedded image


ethyl (R)-1-((4-(N-(oxetan-3- yl)sulfamoyl)phenyl)sulfonyl) piperidine-3-carboxylate
432.51
  2.18





DX2- 258


embedded image


ethyl (3R)-1-((4-((2,6- dimethylpiperidin-1- yl)sulfonyl)phenyl)sulfonyl) piperidine-3-carboxylate
472.62
  4.16





DX2- 259


embedded image


ethyl (R)-1-((4-(N,N- dimethylsulfamoyl)phenyl) sulfonyl)piperidine-3- carboxylate
404.50
  1.93





DX2- 260


embedded image


1-methylpiperidin-4-yl (R)-1-((4- (N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxylate
501.66
  2.34





DX2- 261


embedded image


isopropyl (R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxylate
446.58
  3.29





DX2- 262


embedded image


oxetan-3-yl (R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxylate
460.56
  2.44





DX2- 263


embedded image


cyclopropyl (R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxylate
444.57
  3.04





DX2- 264


embedded image


ethyl (R)-1-((4-(N,N- dipropylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxylate
460.61
  4.04





DX2- 265


embedded image


ethyl (R)-1-((4-(piperidin-1- ylsulfonyl)phenyl)sulfonyl) piperidine-3-carboxylate
444.57
  3.12





DX2- 266


embedded image


tert-butyl (R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxylate
460.61
  3.69





DX2- 275


embedded image


ethyl (R)-1-((4-(N,N- diethylsulfamoyl)-3- fluorophenyl)sulfonyl)piperidine- 3-carboxylate
450.55
  3.13





DX2- 276


embedded image


ethyl (R)-1-((4-(N,N- diethylsulfamoyl)-2- methylphenyl)sulfonyl)piperidine- 3-carboxylate
446.58
  3.48





DX2- 280


embedded image


ethyl (R)-1-((4-(N,N- diisopropylsulfamoyl)phenyl) sulfonyl)piperidine-3-carboxylate
460.61
  3.60





DX2- 282


embedded image


cyclobutyl (R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxylate
458.59
  3.37





DX2- 291


embedded image


ethyl (R)-1-((6-(N,N- diethylsulfamoyl)pyridin-3- yl)sulfonyl)piperidine-3- carboxylate
433.54
  1.79





DX2- 293


embedded image


ethyl (R)-1-((5-(N,N- diethylsulfamoyl)pyridin-2- yl)sulfonyl)piperidine-3- carboxylate
433.54
  1.79





DX2- 295


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-isopropylpiperidine-3- carboxamide
445.60
  2.32





DX2- 296


embedded image


(R)-N-cyclopropyl-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxamide
443.58
  2.07





DX2- 297


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-isopropyl-N-methylpiperidine-3- carboxamide
459.62
  2.71





DX2- 300


embedded image


ethyl (R)-1-((4- sulfamoylphenyl)sulfonyl) piperidine-3-carboxylate
376.44
  1.34





DX3- 3


embedded image


ethyl 3-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) benzoate
425.51
  3.49





DX3- 13


embedded image


ethyl 3-((4-(N,N- diethylsulfamoyl)phenyl) sulfonamido)benzoate
440.563
  4.00





DX3- 14B- P1


embedded image


(cis)-ethyl 1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- 5-hydroxypiperidine-3-carboxylate
448.55
  2.37





DX3- 14B- P2


embedded image


(trans)-ethyl 1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- 5-hydroxypiperidine-3-carboxylate
448.55
  2.37





DX3- 30


embedded image


ethyl 5-amino-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxylate
447.57
  2.43





DX3- 35B


embedded image


N,N-diethyl-4-(3- (methylsulfonyl)piperidin-1- yl)sulfonyl)benzenesulfonamide
438.57
  1.16





DX3- 37


embedded image


(R)-4-((3-cyanopiperidin-1- yl)sulfony))-N,N- diethylbenzenesulfonamide
385.50
  1.88





DX3- 38


embedded image


(R)-4-((3-(1H-tetrazol-5- yl)piperidin-1-yl)sulfonyl)-N,N- diethylbenzenesulfonamide
428.53
  1.51





DX3- 39


embedded image


1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N,N-dimethylpiperidine-3- sulfonamide
467.61
  2.02





DX3- 39B


embedded image


ethyl 4-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) morpholine-2-carboxylate
434.52
  2.07





DX3- 43


embedded image


(R)-N,N-diethyl-4-((3-(5-oxo-4,5- dihydro-1,2,4-oxadiazol-3- yl)piperidin-1- yl)sulfonyl)benzenesulfonamide
444.52
  1.25





DX3- 43B


embedded image


ethyl 2-(1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidin-3-yl)acetate
446.58
  2.79





DX3- 44B


embedded image


ethyl 4-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperazine-2-carboxylate
433.54
  2.06





DX3- 45


embedded image


ethyl 1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- 4-hydroxy-1,2,5,6- tetrahydropyridine-3-carboxylate
446.53
  2.03





DX3- 46


embedded image


N,N-diethyl-4-(3- hydroxypiperidin-1- yl)sulfonyl)benzenesulfonamide
376.49
  1.55





DX3- 47


embedded image


ethyl 1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- 3-fluoropiperidine-3-carboxylate
450.54
  3.23





DX3- 48B


embedded image


ethyl (R)-4-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- 1-methylpiperazine-2-carboxylate
447.57
  2.66





DX3- 49B


embedded image


N,N-diethyl-4-((3-fluoropiperidin- 1-yl)sulfonyl)benzenesulfonamide
378.48
  2.63





DX3- 50


embedded image


4-((3-cyclohexylpiperidin-1- yl)sulfonyl)-N,N- diethylbenzenesulfonamide
442.63
  5.33





DX3- 52B


embedded image


ethyl 3-benzyl-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxylate
522.68
  5.07





DX3- 69B- P1


embedded image


(trans)-ethyl-8-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- 8-azabicyclo[3.2.1]octane-2- carboxylate
458.59
  3.30





DX3- 69B- P2


embedded image


(cis)-ethyl-8-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- 8-azabicyclo[3.2.1]octane-2- carboxylate
458.59
  3.30





DX3- 78


embedded image


ethyl (R)-1-((4′-(N,N- diethylsulfamoyl)-[1,1′-biphenyl]- 4-yl)sulfonyl)piperidine-3- carboxylate
508.65
  4.87





DX3- 78B


embedded image


ethyl (R)-1-((4- (diethylcarbamoyl)phenyl)sulfonyl) piperidine-3-carboxylate
396.50
  2.39





DX3- 79


embedded image


ethyl (R)-1-(4-(N,N- diethylsulfamoyl)benzoyl) piperidine-3-carboxylate
396.50
  2.56





DX3- 84


embedded image


(R)-N,N-dicyclopropyl-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxamide
483.64
  3.10





DX3- 86B


embedded image


ethyl (R)-1-((4-(N-benzyl-N- methylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxylate
480.59
  3.70





DX3- 90


embedded image


ethyl (3R)-1-((4-(N,N- diethylsulfamidimidoyl)phenyl) sulfonyl)piperidine-3-carboxylate
431.57
  2.64





DX3- 99B


embedded image


ethyl (R)-1-((6-(N,N- diethylsulfamoyl)naphthalen-2- yl)sulfonyl)piperidine-3- carboxylate
482.61
  4.16





DX3- 100


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-methyl-N-(oxetan-3- yl)piperidine-3-carboxamide
473.60
  2.66





DX3- 100B


embedded image


(R)-N-(1-cyanocyclopropyl)-1-((4- (N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxamide
468.59
  1.43





DX3- 101- P1


embedded image


(cis)-ethyl 3-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) cyclohexane-1-carboxylate
431.56
  3.12





DX3- 101- P2


embedded image


(trans)-ethyl 3-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) cyclohexane-1-carboxylate
431.56
  3.12





DX3- 102B


embedded image


ethyl (R)-1-((4- (isopropyl(methyl)carbamoyl) phenyl)sulfonyl)piperidine-3- carboxylate
396.50
  2.17





DX3-103


embedded image


ethyl (R)-1-((4- (ethyl(propyl)carbamoyl)phenyl) sulfonyl)piperidine-3-carboxylate
410.53
  2.92





DX3- 104


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-(oxetan-3-ylmethyl)piperidine-3- carboxamide
473.60
  0.62





DX3- 104B


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-(3-methyloxetan-3-yl)piperidine- 3-carboxamide
473.60
  2.57





DX3- 105


embedded image


ethyl (R)-1-((4-(N-ethyl-N- isopropylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxylate
446.58
  3.29





DX3- 106B


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-(tetrahydro-2H-pyran-4- yl)piperidine-3-carboxamide
487.63
  1.12





DX3- 107


embedded image


tert-butyl (R)-3-(1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxamido)azetidine- 1-carboxylate
558.71
  4.06





DX3- 107B


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-((R)-tetrahydrofuran-3- yl)piperidine-3-carboxamide
473.60
  1.71





DX3- 108


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-((S)-tetrahydrofuran-3- yl)piperidine-3-carboxamide
473.60
  1.71





DX3- 110


embedded image


(R)-N-(azetidin-3-yl)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxamide
458.59
  2.04





DX3- 112B


embedded image


ethyl (S)-3-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-1-carboxylate
432.55
  2.76





DX3- 114


embedded image


ethyl (R)-3-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-1-carboxylate
432.55
  2.76





DX3- 115


embedded image


ethyl 3-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- 3-azabicyclo[3.1.1]heptane-1- carboxylate
444.56
  2.74





DX3- 115B


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-(1-ethylazetidin-3-yl)piperidine- 3-carboxamide
486.65
  3.21





DX3- 116


embedded image


ethyl (R)-3-(1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxamido)azetidine- 1-carboxylate
530.66
  3.35





DX3- 116B


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-(1-pivaloylazetidin-3- yl)piperidine-3-carboxamide
542.71
  3.17





DX3- 117


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-(1-(methylsulfonyl)azetidin-3- yl)piperidine-3-carboxamide
536.68
  2.25





DX3- 117B


embedded image


(R)-N-(1- (cyclopropanecarbonyl)azetidin-3- yl)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxamide
526.67
  2.51





DX3- 118


embedded image


tert-butyl (R)-3-((R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3- carboxamido)pyrrolidine-1- carboxylate
572.74
  3.54





DX3- 118B


embedded image


tert-butyl (R)-1-(1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carbonyl)-1,6- diazaspiro[3.3]heptane-6- carboxylate
584.75
  3.68





DX3- 119


embedded image


tert-butyl (S)-3-((R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3- carboxamido)pyrrolidine-1- carboxylate
572.74
  3.54





DX3- 120


embedded image


tert-butyl (R)-6-(1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carbonyl)-2,6- diazaspiro[3.3]heptane-2- carboxylate
584.75
  3.82





DX3- 121


embedded image


tert-butyl (R)-4-(1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carbonyl)piperazine-1- carboxylate
572.74
  3.93





DX3- 121B


embedded image


(R)-N,N-diethyl-4-((3-(morpholine- 4-carbonyl)piperidin-1- yl)sulfonyl)benzenesulfonamide
473.60
  1.96





DX3- 122


embedded image


(R)-N,N-diethyl-4-((3-(piperidine-1- carbonyl)piperidin-1- yl)sulfonyl)benzenesulfonamide
471.63
  2.75





DX3- 122B


embedded image


(R)-N,N-diethyl-4-((3-(3-methyl- 1,2,4-oxadiazol-5-yl)piperidin-1- yl)sulfonyl)benzenesulfonamide
442.55
  1.73





DX3- 123


embedded image


(R)-N-(1-(tert butylcarbamoyl)azetidin-3-yl)-1- ((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxamide
557.73
  3.36





DX3- 123B


embedded image


(R)-N,N-diethyl-4-((3-(3- methylisoxazol-5-yl)piperidin-1- yl)sulfonyl)benzenesulfonamide
441.56
  2.36





DX3- 124


embedded image


tert-butyl (R)-4-(1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxamido) piperidine-1-carboxylate
586.76
  3.03





DX3- 125


embedded image


(R)-N-((R)-1-benzoylpyrrolidin-3- yl)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carboxamide
576.73
  2.68





DX3- 125B


embedded image


ethyl (R)-3-((R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3- carboxamido)pyrrolidine-1- carboxylate
544.68
  2.84





DX3- 126B


embedded image


tert-butyl (1S,4S)-5-((R)-1-((4- (N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carbonyl)-2,5- diazabicyclo[2.2.1]heptane-2- carboxylate
584.75
  4.24





DX3- 127


embedded image


tert-butyl (R)-2-(1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carbonyl)-2,6- diazaspiro[3.4]octane-6-carboxylate
598.77
  3.55





DX3- 127B


embedded image


tert-butyl 5-((R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3- carbonyl)hexahydropyrrolo[3,4- c]pyrrole-2(1H)-carboxylate
598.77
  3.55





DX3- 128


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-((R)-1-(3,3- dimethylbutanoyl)pyrrolidin-3- yl)piperidine-3-carboxamide
570.76
  2.89





DX3- 128B


embedded image


tert-butyl (1R,4R)-5-((R)-1-((4- (N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carbonyl)-2,5- diazabicyclo[2.2.1]heptane-2- carboxylate
584.75
  4.24





DX3- 130


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-((R)-1-(3-isopropyl-1,2,4- oxadiazol-5-yl)pyrrolidin-3- yl)piperidine-3-carboxamide
582.74
  2.87





DX3- 132B


embedded image


(R)-4-((3-(4-cyanopiperazine-1- carbonyl)piperidin-1-yl)sulfonyl)- N,N-diethylbenzenesulfonamide
497.63
  1.93





DX3- 134


embedded image


(R)-N,N-diethyl-4-((3-(4-(3-methyl- 1,2,4-oxadiazol-5-yl)piperazine-1- carbonyl)piperidin-1- yl)sulfonyl)benzenesulfonamide
554.68
  2.30





DX3- 134B


embedded image


(R)-N,N-diethyl-4-((3-(4-(3- isopropyl-1,2,4-oxadiazol-5- yl)piperazine-1-carbonyl)piperidin- 1-yl)sulfonyl)benzenesulfonamide
582.74
  3.23





DX3- 139B


embedded image


tert-butyl (R)-4-((R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carbonyl)-3- methylpiperazine-1-carboxylate
586.76
  4.45





DX3- 140


embedded image


(R)-N,N-diethyl-4-((3-(4-(3- (trifluoromethyl)-1,2,4-oxadiazol-5- yl)piperazine-1-carbonyl)piperidin- 1-yl)sulfonyl)benzenesulfonamide
608.65
  2.94





DX3- 141


embedded image


tert-butyl (S)-4-((R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carbonyl)-3- methylpiperazine-1-carboxylate
586.76
  4.45





DX3- 141B


embedded image


tert-butyl (R)-4-((R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carbonyl)-2- methylpiperazine-1-carboxylate
586.76
  4.45





DX3- 142


embedded image


tert-butyl (S)-4-((R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carbonyl)-2- methylpiperazine-1-carboxylate
586.76
  4.45





DX3- 142B


embedded image


tert-butyl 8-((R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carbonyl)-3,8- diazabicyclo[3.2.1]octane-3- carboxylate
598.77
  4.24





DX3- 146


embedded image


(R)-N,N-diethyl-4-((3-(4-(5- isopropyl-1,3,4-oxadiazol-2- yl)piperazine-1-carbonyl)piperidin- 1-yl)sulfonyl)benzenesulfonamide
582.74
  2.42





DX3- 149


embedded image


(R)-4-((3-(6-oxa-2- azaspiro[3.4]octane-2- carbonyl)piperidin-1-yl)sulfonyl)- N,N-diethylbenzenesulfonamide
499.64
  1.73





DX3- 149B


embedded image


(R)-4-((3-(2-oxa-6- azaspiro[3.3]heptane-6- carbonyl)piperidin-1-yl)sulfonyl)- N,N-diethylbenzenesulfonamide
485.61
  1.84





DX3- 162B


embedded image


(R)-N,N-diethyl-4-((3-(4-(3- isopropyl-1,2,4-oxadiazol-5- yl)piperazine-1-carbonyl)piperidin- 1-yl)sulfonyl)benzamide
546.69
  2.64





DX3- 164B


embedded image


(R)-(4-(3-isopropyl-1,2,4- oxadiazol-5-yl)piperazin-1-yl)(1- ((4-(pyrrolidin-1- ylsulfonyl)phenyl)sulfonyl) piperidin-3-yl)methanone
580.72
  2.81





DX3- 165B


embedded image


((3R)-1-((4-((3-hydroxypyrrolidin- 1- yl)sulfonyl)phenyl)sulfonyl) piperidin-3-yl)(4-(3-isopropyl- 1,2,4-oxadiazol-5-yl)piperazin- 1-yl)methanone
596.72
  1.58





DX3- 166B


embedded image


(R)-N-isopropyl-4-((3-(4-(3- isopropyl-1,2,4-oxadiazol-5- yl)piperazine-1-carbonyl)piperidin- 1-yl)sulfonyl)benzenesulfonamide
568.71
  3.14





DX3- 167


embedded image


(R)-N-ethyl-4-((3-(4-(3-isopropyl- 1,2,4-oxadiazol-5-yl)piperazine-1- carbonyl)piperidin-1- yl)sulfonyl)benzenesulfonamide
554.68
  2.83





DX3- 167B


embedded image


(R)-N-cyclobutyl-4-((3-(4-(3- isopropyl-1,2,4-oxadiazol-5- yl)piperazine-1-carbonyl)piperidin- 1-yl)sulfonyl)benzenesulfonamide
580.72
  3.21





DX3- 175


embedded image


N,N-diethyl-4-((1-(4-(3-isopropyl- 1,2,4-oxadiazol-5-yl)piperazine-1- carbonyl)-3- azabicyclo[4.1.0]heptan-3- yl)sulfonyl)benzenesulfonamide
594.75
  2.99





DX3- 176


embedded image


N,N-diethyl-4-((1-(4-(3-isopropyl- 1,2,4-oxadiazol-5-yl)piperazine-1- carbonyl)-3- azabicyclo[3.1.1]heptan-3- yl)sulfonyl)benzenesulfonamide
594.75
  2.99





DX3- 177B


embedded image


N,N-diethyl-4-((1-(4-(morpholine- 4-carbonyl)-3- azabicyclo[4.1.0]heptan-3- yl)sulfonyl)benzenesulfonamide
485.61
  1.71





DX3- 178


embedded image


(R)-N-(tert-butyl)-4-((3-(4-(3- isopropyl-1,2,4-oxadiazol-5- yl)piperazine-1-carbonyl)piperidin- 1-yl)sulfonyl)benzenesulfonamide
582.74
  3.54





DX3- 178B


embedded image


N,N-diethyl-4-((1-(morpholine-4- carbonyl)-3- azabicyclo[3.1.1]heptan-3- yl)sulfonyl)benzenesulfonamide
485.61
  1.71





DX3- 179B


embedded image


(R)-N-cyclopropyl-4-((3-(4-(3- isopropyl-1,2,4-oxadiazol-5- yl)piperazine-1-carbonyl)piperidin- 1-yl)sulfonyl)benzenesulfonamide
566.69
  2.88





DX3- 184


embedded image


ethyl (R)-4-(1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl) piperidine-3-carbonyl)piperazine-1- carboxylate
544.68
  3.22





DX3- 184B


embedded image


(R)-4-((3-(4,4-difluoropiperidine-1- carbonyl)piperidin-1-yl)sulfonyl)- N,N-diethylbenzenesulfonamide
507.61
  2.14





DX3- 185


embedded image


(R)-4-(3-(1,1- dioxidothiomorpholine-4- carbonyl)piperidin-1-yl)sulfonyl)- N,N-diethylbenzenesulfonamide
521.66
  0.99





DX3- 185B


embedded image


(R)-N,N-diethyl-4-((3-(4- methoxypiperidine-1- carbonyl)piperidin-1- yl)sulfonyl)benzenesulfonamide
501.66
  1.38





DX3- 186


embedded image


(R)-N,N-diethyl-4-((3-(4- hydroxypiperidine-1- carbonyl)piperidin-1- yl)sulfonyl)benzenesulfonamide
487.63
  0.66





DX3- 186B


embedded image


(R)-N,N-diethyl-4-((3-(4- (methoxymethyl)piperidine-1- carbonyl)piperidin-1- yl)sulfonyl)benzenesulfonamide
515.68
  2.00





DX3- 187B


embedded image


N,N-diethyl-4-(((R)-3-((R)-3- methoxypyrrolidine-1- carbonyl)piperidin-1- yl)sulfonyl)benzenesulfonamide
487.63
  2.07





DX3- 188


embedded image


N,N-diethyl-4-(((R)-3-((S)-3- methoxypyrrolidine-1- carbonyl)piperidin-1- yl)sulfonyl)benzenesulfonamide
487.63
  2.07





DX3- 188B


embedded image


(R)-N,N-diethyl-4-((3-(4-(2- hydroxypropan-2-yl)piperidine-1- carbonyl)piperidin-1- yl)sulfonyl)benzenesulfonamide
529.71
  1.99





DX3- 192


embedded image


(R)-4-((3,3-difluoro-5-(4-(3- isopropyl-1,2,4-oxadiazol-5- yl)piperazine-1-carbonyl)piperidin- 1-yl)sulfony))-N,N- diethylbenzenesulfonamide
618.72
  3.52





DX3- 193


embedded image


(R)-N,N-diethyl-4-((3-(4-(3- isopropyl-1,2,4-oxadiazol-5- yl)piperidine-1-carbonyl)piperidin- 1-yl)sulfonyl)benzenesulfonamide
581.75
  2.48





DX3- 193B


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-((1s,4S)-4- methoxycyclohexyl)piperidine-3- carboxamide
515.68
  2.15





DX3- 194


embedded image


(R)-1-((4-(N,N- diethylsulfamoyl)phenyl)sulfonyl)- N-((1r,4R)-4- methoxycyclohexyl)piperidine-3- carboxamide
515.68
  2.15





DX3- 194B


embedded image


(R)-4-((3,3-difluoro-5-(morpholine- 4-carbonyl)piperidin-1-yl)sulfonyl)- N,N-diethylbenzenesulfonamide
509.58
  2.24





DX3- 195B


embedded image


(R)-4-((3-(4,4-difluoropiperidine-1- carbonyl)piperidin-1-yl)sulfonyl)- N-isopropylbenzenesulfonamide
493.58
  2.04





DX3- 198


embedded image


(R)-4-(3-(4- (difluoromethyl)piperidine-1- carbonyl)piperidin-1-yl)sulfonyl)- N,N-diethylbenzenesulfonamide
521.64
  2.41





DX3- 198B


embedded image


(R)-N,N-diethyl-4-((3-(4- (trifluoromethyl)piperidine-1- carbonyl)piperidin-1- yl)sulfonyl)benzenesulfonamide
539.63
  2.84





DX3- 201B


embedded image


(R)-4-((3-(4,4-difluoropiperidine-1- carbonyl)piperidin-1-yl)sulfonyl)- N,N-dimethylbenzenesulfonamide
479.56
  1.08





DX3- 202B


embedded image


(R)-(4,4-difluoropiperidin-1-yl)(1- ((4-(pentan-3- ylsulfonyl)phenyl)sulfonyl)piperidin- 3-yl)methanone
506.62
  2.36





DX3- 203


embedded image


(R)-4-((3-(4,4-difluoropiperidine-1- carbonyl)piperidin-1-yl)sulfonyl)- N,N-diethylbenzamide
471.56
  1.54





DX3- 205


embedded image


4-(((3R,5S)-3-(4,4- difluoropiperidine-1-carbonyl)-5- hydroxypiperidin-1-yl)sulfonyl)- N,N-diethylbenzenesulfonamide
507.61
  1.95





DX3- 206


embedded image


4-(((3R,5R)-3-(4,4- difluoropiperidine-1-carbonyl)-5- hydroxypiperidin-1-yl)sulfonyl)- N,N-diethylbenzenesulfonamide
507.61
  1.95





DX3- 207


embedded image


(R)-6-((3-(4,4-difluoropiperidine-1- carbonyl)piperidin-1-yl)sulfonyl)- N,N-diethylpyridine-3-sulfonamide
508.60
  0.94





DX3- 208


embedded image


(4,4-difluoropiperidin-1-yl)((R)-1- ((4-(((R)-3-hydroxypyrrolidin-1- yl)sulfonyl)phenyl)sulfonyl) piperidin-3-yl)methanone
521.59
  0.48





DX3- 209


embedded image


(4,4-difluoropiperidin-1-yl)((R)-1- ((4-(((S)-3-hydroxypyrrolidin-1- yl)sulfonyl)phenyl)sulfonyl) piperidin-3-yl)methanone
521.59
  0.48





DX3- 209B


embedded image


(R)-morpholino(1-((4-(pentan-3- ylsulfonyl)phenyl)sulfonyl) piperidin-3-yl)methanone
472.62
  2.18





DX3- 210


embedded image


(R)-(4-(3-isopropyl-1,2,4- oxadiazol-5-yl)piperazin-1-yl)(1- ((4-(pentan-3- ylsulfonyl)phenyl)sulfonyl) piperidin-3-yl)methanone
581.75
  3.46





DX3- 213B


embedded image


(R)-(4,4-difluoropiperidin-1-yl)(1- ((4- (isopropylsulfonyl)phenyl)sulfonyl) piperidin-3-yl)methanone
478.57
  1.30





DX3- 214


embedded image


(R)-(4,4-difluoropiperidin-1-yl)(1- ((4- (dimethylphosphoryl)phenyl) sulfonyl)piperidin-3- yl)methanone
448.47
−0.75





DX3- 215


embedded image


(R)-(1-(4- (isopropylsulfonyl)phenyl)sulfonyl) piperidin-3- yl)(morpholino)methanone
444.56
  1.12





DX3- 216


embedded image


(R)-(1-(4- (diethylphosphoryl)phenyl)sulfonyl) piperidin-3-yl)(4,4- difluoropiperidin-1-yl)methanone
476.52
  0.15





DX3- 218


embedded image


(R)-(4,4-difluoropiperidin-1-yl)(1- ((4- (isobutylsulfonyl)phenyl)sulfonyl) piperidin-3-yl)methanone
492.60
  1.92





DX3- 219B


embedded image


(R)-(4,4-difluoropiperidin-1-yl)(1- ((4-(oxetan-3- ylsulfonyl)phenyl)sulfonyl) piperidin-3-yl)methanone
492.55
  0.78





DX3- 220B


embedded image


(R)-(1-(4- (cyclobutylsulfonyl)phenyl) sulfonyl)piperidin-3-yl)(4,4- difluoropiperidin-1-yl)methanone
490.58
  1.38





DX3- 221


embedded image


(R)-(1-(4- (cyclopropylsulfonyl)phenyl) sulfonyl)piperidin-3-yl)(4,4- difluoropiperidin-1-yl)methanone
476.55
  1.05





DX3- 221B


embedded image


(R)-(4,4-difluoropiperidin-1-yl)(1- ((4-((3-hydroxyazetidin-1- yl)sulfonyl)phenyl)sulfonyl) piperidin-3-yl)methanone
507.6
  0.78





DX3- 224


embedded image


(4-(((R)-3-(4,4-difluoropiperidine- 1-carbonyl)piperidin-1- yl)sulfonyl)phenyl)(imino) (isopropyl)-l6-sulfanone
477.6
  1.04





DX3- 226


embedded image


(4-(((R)-3-(4,4-difluoropiperidine- 1-carbonyl)piperidin-1- yl)sulfonyl)phenyl)(isopropyl) (methylimino)-l6-sulfanone
491.6
  1.80





DX3- 226


embedded image


(R)-(4,4-difluoropiperidin-1-yl)(1- ((4- (methylsulfonyl)phenyl)sulfonyl) piperidin-3-yl)methanone
450.5
  0.47





DX3- 226


embedded image


(R)-(4,4-difluoropiperidin-1-yl)(1- ((5-(isopropylsulfonyl)pyridin-2- yl)sulfonyl)piperidin-3- yl)methanone
479.6
  0.39









The invention provides the stereochemical mixtures or pure forms for all compounds of invention.


The invention also provides all pharmaceutically acceptable salts, esters, amides, tautomers, geometric isomers, solvates thereof, as well as pharmaceutical composition comprising an effective amount of a compound of the invention and a pharmaceutically acceptable carrier. The composition may further comprise an effective amount of one or more other agents for treating cancer or a disorder associated with mitochondrial function.


The invention further provides prodrugs of all compounds of invention. The term “prodrug” used herein refers to a pharmacologically inactive derivative of a parent “drug” molecule which requires biotransformation within the target physiological system to release, or to convert the prodrug into the active drug. Prodrugs can address the problems associated with solubility, stability, cell permeability or bioavailability. Prodrugs usually comprise an active drug molecule and a chemical masking group. Prodrugs can be readily prepared from the parent compounds with well-known methods.


Furthermore, the invention provides a packaged product comprising a container; an effective amount of a compound of invention.


The invention also provides methods of preparing the compounds of invention.


The following schemes can be used to practice the present invention. Additional structural groups, including but not limited to those defined elsewhere in the specification and not shown in the compounds described in the schemes can be incorporated to give various compound disclosed herein, or intermediate compounds which can, after further manipulation using techniques known in the art, be converted to compounds of the present invention.


One route for preparation of compounds of the present invention is depicted in Scheme 1.




embedded image


embedded image


The typical synthesis of the compounds of the invention starts from a condensation reaction between halogen substituted sulfonyl chloride (I) and the appropriate amine to give sulfonamide intermediate I. A Palladium catalyzed coupling or SNAr replacement reaction, depending on the electronic nature of the core structure A, between II and benzyl mercaptan furnish intermediate III, which is then subjected to an oxidative chlorination to give sulfonyl chloride IV. A second condensation reaction between IV and an amine give rise to the final product V. For part of the compounds of the invention, the mono-substituted sulfonyl chloride IV can be obtained directly from the reaction of a commercially available disulfonyl chloride VI and the appropriate amine.


In cases when the starting material I is not readily available, it was synthesized following Scheme 2.




embedded image


Starting from a di-halogenated compound VII, the selective Palladium catalyzed coupling or SNAr replacement reaction, depending on the electronic nature of the core structure A, give rise to intermediate VIII, which was converted to I with a similar oxidative chlorination approach.


For compounds with diverse piperidine-3-carboxylate or piperidine-3-carboxamide structures, exemplified by Formula III, when R1 is —OR or NR1R2, the synthesis is further following the method depicted in Scheme 3 based on Scheme 1.




embedded image


Starting from compound Va with an ethyl piperidine-3-carboxylate structure, the corresponding carboxylic acid IX is obtained through a basic hydrolysis. With either EDCI/DMAP or HATU/DIEA coupling reagent/base combination, the corresponding esters X or amides XI is obtained respectively.


EXAMPLES

The following examples are illustrative, but not limiting, of the compounds, compositions, and methods of present invention. Other suitable modifications and adaptations of the variety of conditions and parameters normally encountered in clinical therapy and which are obvious to those skilled in the art are within the spirit and scope of the invention.


In case the preparative examples are obtained as a mixture of enantiomers, the pure enantiomer can be obtained by methods known to those skilled in the art, such as e.g. chiral chromatography or crystallization.


Example I
Chemistry
General Experimental Methods

All commercial reagents and anhydrous solvents were purchased and used without purification, unless specified. Column chromatography was performed on a Biotage Isolera flash chromatography system on Biotage normal phase silica gel columns. Preparative-HPLC purification was performed by Shimadzu Semi-Prep LC system. Analytical thin layer chromatography was performed on Merck pre-coated plates (silica gel 60 F254). NMR spectra were recorded on a Bruker Ultrashield 300 MHz or Bruker Ascend 400 MHz spectrometer using deuterated CDCl3 or CD3OD as solvents. Chemical shifts for proton magnetic resonance spectra (1H NMR) are quoted in parts per million (ppm) referenced to the appropriate solvent peak or 0.0 ppm for tetramethylsilane (TMS). The following abbreviations are used to describe the peak-splitting patterns when appropriate: br, broad; s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; and dd, doublet of doublets. Coupling constants, J, are reported in hertz (Hz). Mass spectra were recorded on a Shimadzu LCMS-2020 system using the electro spray ionization (ESI) ion source. HPLC was used to determine the purity of biologically tested compounds using Shimadzu LC-2030C 3D system on Kinetex XB-C18 column (2.6 μm, 4.6×75 mm) under the following gradient elution conditions: acetonitrile/water (10-95%) or methanol/water (10-95%), both with 0.1% formic acid as the additive, over 15 minutes at a 0.80 mL/min flow rate at room temperature. The purity was established by integration of the areas of major peaks detected at 254 nm, and all final products have >95% purity.




embedded image


4-Bromo-N,N-diethylbenzenesulfonamide (2a)

To a solution of diethylamine (730 mg, 10.0 mmol) and triethylamine (2.02 g, 20.0 mmol) was added 4-bromobenzenesulfonyl chloride (1a, 2.56 g, 10.0 mmol) portionwise. The mixture was stirred at room temperature overnight. The mixture was concentrated and purified with flash chromatography (10% EtOAc in hexane) to give 2a as a colorless oil. (2.44 g, 83%). 1H NMR (300 MHz, CDCl3) δ 7.73-7.62 (m, 4H), 3.26 (q, J=7.1 Hz, 4H), 1.15 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 291.9, 293.9 [M+H]+.


4-(Benzylthio)-N,N-diethylbenzenesulfonamide (3a)

A solution of 2a, DIEA in dioxane was degassed and flushed with argon for three times. Then Pd2(dba)3, XantPhos, and benzyl mercaptan was added subsequently. Then it was degassed and flushed with argon for three times again before it was heated at reflux overnight. The mixture was cooled and the needle-like crystals generated was filtered off. The filtrate was concentrated and purified with flash chromatography (20% EtOAc in hexane) to give 3a as a yellow solid (520 mg, 91%). 1H NMR (300 MHz, CDCl3) δ 7.68 (d, J=8.1 Hz, 2H), 7.43-7.22 (m, 7H), 4.22 (s, 2H), 3.23 (q, J=7.2 Hz, 4H), 1.13 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 336.0 [M+H]+.


4-(N,N-Diethylsulfamoyl)benzenesulfonyl chloride (4a)

To an ice-cooled solution of 3a (100 mg, 0.30 mmol) in a mixture of CH3CN (2.5 mL), HOAc (0.16 mL) and H2O (0.1 mL) was added 1,3-dichloro-5,5-dimethylimidazolidine-2,4-dione (117 mg, 0.60 mmol) portionwise. The mixture was kept stirring at 0-5° C. for 2 h and concentrated. The residue obtained was taken up by DCM, washed with 5% NaHCO3 solution at 0° C., dried over Na2SO4, filtered and concentrated to give 4a as a white solid (94 mg, 100% crude) and used in the next step without further purification. 1H NMR (300 MHz, CDCl3) δ 8.22-8.04 (m, 4H), 3.32 (q, J=7.2 Hz, 4H), 1.19 (t, J=7.1 Hz, 6H).


Ethyl (R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-201)

To a solution of ethyl (R)-piperidine-3-carboxylate (1.97 g, 12.53 mmol) and triethylamine (2.11 g, 20.88 mmol) in DCM (50 mL) was added 4a (3.25 g, 10.44 mmol). The mixture was stirred at room temperature for 3 h. The mixture was concentrated and purified with flash chromatography (20% EtOAc in hexane) to give 2a (4.12 g, 91%) as a white solid. 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.2 Hz, 2H), 7.90 (d, J=8.3 Hz, 2H), 4.16 (q, J=7.1 Hz, 2H), 3.86 (d, J=7.8 Hz, 1H), 3.64 (d, J=11.6 Hz, 1H), 3.30 (q, J=7.2 Hz, 4H), 2.72-2.54 (m, 2H), 2.50-2.38 (m, 1H), 2.03 (d, J=13.6 Hz, 1H), 1.91-1.76 (m, 2H), 1.72-1.60 (m, 1H), 1.44 (q, J=10.2 Hz, 1H), 1.28 (t, J=7.1 Hz, 3H), 1.17 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 433.0 [M+H]+.


Ethyl (S)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-202)

Using a similar procedure as described for DX2-201 with 4a (47 mg, 0.15 mmol) and ethyl (S)-piperidine-3-carboxylate (26 mg, 0.17 mmol), white solid (44 mg, 68%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.2 Hz, 2H), 7.90 (d, J=8.4 Hz, 2H), 4.16 (q, J=7.1 Hz, 2H), 3.86 (d, J=7.9 Hz, 1H), 3.64 (d, J=11.6 Hz, 1H), 3.30 (q, J=7.1 Hz, 4H), 2.69-2.56 (m, 2H), 2.44 (td, J=11.3, 3.1 Hz, 1H), 2.03 (d, J=13.6 Hz, 1H), 1.91-1.78 (m, 1H), 1.69 (t, J=12.0 Hz, 1H), 1.44 (d, J=10.6 Hz, 1H), 1.28 (t, J=7.1 Hz, 3H), 1.17 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 433.2 [M+H]+.


Ethyl 1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-4-carboxylate (DX2-208)

Using a similar procedure as described for DX2-201 with 4a (47 mg, 0.15 mmol) and ethyl piperidine-4-carboxylate (26 mg, 0.17 mmol), white solid (40 mg, 62%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.4 Hz, 2H), 7.89 (d, J=8.4 Hz, 2H), 4.14 (q, J=7.1 Hz, 2H), 3.72-3.61 (m, 2H), 3.30 (q, J=7.1 Hz, 4H), 2.64-2.52 (m, 2H), 2.37-2.25 (m, 1H), 2.06-1.97 (m, 2H), 1.91-1.77 (m, 2H), 1.24 (t, J=7.1 Hz, 3H), 1.17 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 433.0 [M+H]+.


Ethyl 1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-2-carboxylate (DX2-209)

Using a similar procedure as described for DX2-201 with 4a (47 mg, 0.15 mmol) and ethyl piperidine-2-carboxylate (26 mg, 0.17 mmol), white solid (26 mg, 40%). 1H NMR (300 MHz, CDCl3) δ 7.93 (s, 4H), 4.76 (d, J=5.7 Hz, 1H), 4.12-3.93 (m, 2H), 3.82 (d, J=13.4 Hz, 1H), 3.33-3.17 (m, 5H), 2.20 (d, J=13.7 Hz, 1H), 1.87-1.65 (m, 3H), 1.60-1.42 (m, 1H), 1.40-1.23 (m, 1H), 1.20-1.12 (m, 9H). LC-MS (ESI) m/z 433.0 [M+H]+.


Ethyl (R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)pyrrolidine-3-carboxylate (DX2-217)

Using a similar procedure as described for DX2-201 with 4a (40 mg, 0.13 mmol) and ethyl (R)-pyrrolidine-3-carboxylate (23 mg, 0.13 mmol), white solid (32 mg, 59%). 1H NMR (400 MHz, CDCl3) δ 8.04-7.94 (m, 4H), 4.09 (q, J=7.2 Hz, 2H), 3.66-3.56 (m, 1H), 3.49 (ddd, J=10.3, 6.4, 1.0 Hz, 1H), 3.45-3.33 (m, 2H), 3.37-3.26 (m, 4H), 3.01 (p, J=7.2 Hz, 1H), 2.21-2.03 (m, 2H), 1.23 (t, J=7.1 Hz, 3H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 419.0 [M+H]+.


Ethyl (S)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)pyrrolidine-3-carboxylate (DX2-218)

Using a similar procedure as described for DX2-201 with 4a (40 mg, 0.13 mmol) and ethyl (S)-pyrrolidine-3-carboxylate (23 mg, 0.13 mmol), white solid (32 mg, 59%). 1H NMR (300 MHz, CDCl3) δ 7.98 (s, 4H), 4.08 (q, J=7.1 Hz, 2H), 3.65-3.56 (m, 1H), 3.51-3.44 (m, 1H), 3.42-3.25 (m, 6H), 3.05-2.95 (m, 1H), 2.16-2.06 (m, 2H), 1.25-1.14 (m, 9H). LC-MS (ESI) m/z 419.0 [M+H]+.


Ethyl 1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)azetidine-3-carboxylate (DX2-219)

Using a similar procedure as described for DX2-201 with 4a (40 mg, 0.13 mmol) and ethyl azetidine-3-carboxylate (22 mg, 0.13 mmol), white solid (28 mg, 53%). 1H NMR (300 MHz, CDCl3) δ 8.01 (q, J=8.5 Hz, 4H), 4.15-4.04 (m, 4H), 4.01-3.94 (m, 2H), 3.31 (q, J=7.2 Hz, 5H), 1.20 (dt, J=11.5, 7.1 Hz, 9H). LC-MS (ESI) m/z 405.0 [M+H]+.


Ethyl 1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)azepane-4-carboxylate (DX2-221)

Using a similar procedure as described for DX2-201 with 4a (40 mg, 0.13 mmol) and ethyl azepane-4-carboxylate (22 mg, 0.13 mmol), white solid (30 mg, 52%). 1H NMR (300 MHz, CDCl3) δ 8.00-7.88 (m, 4H), 4.14 (q, J=7.1 Hz, 2H), 3.54-3.43 (m, 1H), 3.38 (q, J=5.0, 4.3 Hz, 2H), 3.29 (q, J=7.1 Hz, 4H), 3.15 (dd, J=13.2, 8.8 Hz, 1H), 2.59 (s, 1H), 2.18-2.03 (m, 2H), 1.95 (dd, J=14.2, 8.0 Hz, 2H), 1.82-1.71 (m, 2H), 1.27 (t, J=7.1 Hz, 3H), 1.17 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 447.0 [M+H]+.


Ethyl (R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)-3-methylpiperidine-3-carboxylate (DX2-246)

Using a similar procedure as described for DX2-201 with 4a (40 mg, 0.13 mmol) and ethyl (R)-3-methylpiperidine-3-carboxylate (24 mg, 0.14 mmol), white solid (17 mg, 29%). 1H NMR (300 MHz, CDCl3) δ 8.01-7.96 (m, 2H), 7.92-7.87 (m, 2H), 4.20 (ddq, J=10.6, 7.1, 3.5 Hz, 2H), 3.64 (d, J=11.6 Hz, 1H), 3.30 (q, J=7.2 Hz, 4H), 3.25-3.16 (m, 1H), 2.78 (dd, J=13.0, 7.4 Hz, 1H), 2.61 (d, J=11.5 Hz, 1H), 2.07-1.95 (m, 1H), 1.81-1.68 (m, 2H), 1.30 (t, J=7.1 Hz, 4H), 1.23 (s, 3H), 1.17 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 447.1 [M+H]+.


Ethyl 3-((4-(N,N-diethylsulfamoyl)phenyl)sulfonamido)benzoate (DX3-13)

To a solution of ethyl 3-aminobenzoate (12 mg, 0.07 mmol) in pyridine (1 mL) was added 4a (20 mg, 0.07 mmol). The mixture was stirred at room temperature for 3 h. The mixture was then concentrated and purified with flash chromatography (40% EtOAc in hexane) to give DX3-13 as a white solid (17 mg, 58%). 1H NMR (300 MHz, CDCl3) δ 7.89 (s, 4H), 7.87-7.83 (m, 1H), 7.70 (td, J=1.8, 0.7 Hz, 1H), 7.46-7.36 (m, 2H), 6.97 (s, 1H), 4.39 (q, J=7.1 Hz, 2H), 3.25 (q, J=7.2 Hz, 4H), 1.40 (t, J=7.1 Hz, 3H), 1.11 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 439.2 [M−H].


(cis)-Ethyl 1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)-5-hydroxypiperidine-3-carboxylate (DX3-14B-P1) and (trans)-ethyl 1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)-5-hydroxypiperidine-3-carboxylate (DX3-14B-P2)

Using a similar procedure as described for DX2-201 with 4a (62 mg, 0.20 mmol) and ethyl 5-hydroxypiperidine-3-carboxylate (38 mg, 0.22 mmol), and purified with flash chromatography (50% EtOAc in hexane) to give DX3-14B-P1 as a white solid (18 mg, 20%) and DX3-14B-P2 as a white solid (8 mg, 9%). DX3-14B-P1 1H NMR (400 MHz, CDCl3) δ 7.99 (d, J=8.6 Hz, 2H), 7.92 (d, J=8.7 Hz, 2H), 4.17 (q, J=7.1 Hz, 2H), 3.89 (tt, J=8.9, 4.3 Hz, 1H), 3.82-3.74 (m, 1H), 3.71 (dd, J=11.4, 4.3 Hz, 1H), 3.30 (q, J=7.2 Hz, 4H), 2.79-2.64 (m, 2H), 2.42 (dd, J=11.3, 8.7 Hz, 1H), 2.32-2.21 (m, 1H), 1.58-1.45 (m, 1H), 1.28 (t, J=7.1 Hz, 3H), 1.17 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 449.2 [M+H]+. DX3-14B-P2 1H NMR (400 MHz, CDCl3) δ 7.99 (d, J=8.7 Hz, 2H), 7.93 (d, J=8.7 Hz, 2H), 4.22-4.14 (m, 2H), 4.13 (tt, J=4.8, 2.1 Hz, 1H), 3.81 (dd, J=12.0, 4.0 Hz, 1H), 3.54 (dd, J=12.0, 4.2 Hz, 1H), 3.31 (q, J=7.2 Hz, 4H), 3.02 (tt, J=10.6, 4.1 Hz, 1H), 2.81 (ddd, J=12.0, 6.0, 3.6 Hz, 2H), 2.08 (dd, J=13.9, 4.5 Hz, 1H), 1.71 (ddd, J=13.9, 11.0, 3.0 Hz, 1H), 1.29 (t, J=7.1 Hz, 3H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 449.0 [M+H]+.


N,N-Diethyl-4-((3-(methylsulfonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-35B)

Using a similar procedure as described for DX2-201 with 4a (47 mg, 0.15 mmol) and 3-(methylsulfonyl)piperidine (30 mg, 0.15 mmol), white solid (53 mg, 80%). 1H NMR (400 MHz, CDCl3) δ 8.01 (d, J=8.7 Hz, 2H), 7.92 (d, J=8.7 Hz, 2H), 4.25 (ddd, J=11.8, 3.9, 1.9 Hz, 1H), 3.86 (d, J=12.2 Hz, 1H), 3.32 (q, J=7.2 Hz, 4H), 3.20 (tt, J=11.4, 3.9 Hz, 1H), 2.94 (s, 3H), 2.63 (t, J=11.4 Hz, 1H), 2.37 (td, J=11.9, 2.8 Hz, 1H), 2.29 (d, J=13.1 Hz, 1H), 2.00 (dt, J=13.5, 3.4 Hz, 1H), 1.76 (qt, J=12.7, 3.9 Hz, 1H), 1.65 (td, J=12.4, 3.8 Hz, 1H), 1.19 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 439.0 [M+H]+.


(R)-4-((3-Cyanopiperidin-1-yl)sulfonyl)-N,N-diethylbenzenesulfonamide (DX3-37)

Using a similar procedure as described for DX2-201 with 4a (221 mg, 0.71 mmol) and (R)-piperidine-3-carbonitrile (78 mg, 0.71 mmol), white solid (223 mg, 81%). 1H NMR (300 MHz, CDCl3) δ 8.00 (d, J=8.6 Hz, 2H), 7.91 (d, J=8.5 Hz, 2H), 3.59 (dd, J=11.9, 3.6 Hz, 1H), 3.40-3.25 (q, J=7.1 Hz, 5H), 3.04 (dd, J=11.8, 8.2 Hz, 1H), 2.94-2.79 (m, 2H), 2.05-1.86 (m, 2H), 1.81-1.66 (m, 2H), 1.17 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 386.1 [M+H]+.


1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N,N-dimethylpiperidine-3-sulfonamide (DX3-39)

Using a similar procedure as described for DX2-201 with 4a (40 mg, 0.13 mmol) and N,N-dimethylpiperidine-3-sulfonamide (32 mg, 0.14 mmol), white solid (50 mg, 82%). 1H NMR (300 MHz, CDCl3) δ 8.03 (d, J=8.6 Hz, 2H), 7.92 (d, J=8.5 Hz, 2H), 4.20 (d, J=11.8 Hz, 1H), 3.88 (d, J=12.0 Hz, 1H), 3.33 (q, J=7.1 Hz, 4H), 3.30-3.23 (m, 1H), 2.97 (s, 6H), 2.57 (t, J=11.4 Hz, 1H), 2.38-2.26 (m, 1H), 2.23-2.12 (m, 1H), 2.03-1.89 (m, 1H), 1.84-1.64 (m, 2H), 1.21 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 468.3 [M+H]+.


Ethyl 4-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)morpholine-2-carboxylate (DX3-39B)

Using a similar procedure as described for DX2-201 with 4a (40 mg, 0.13 mmol) and ethyl morpholine-2-carboxylate (22 mg, 0.14 mmol), white solid (27 mg, 48%). 1H NMR (300 MHz, CDCl3) δ 8.01 (d, J=8.7 Hz, 2H), 7.90 (d, J=8.6 Hz, 2H), 4.32-4.21 (m, 3H), 4.11 (dt, J=11.8, 3.2 Hz, 1H), 3.85-3.70 (m, 2H), 3.48 (d, J=12.0 Hz, 1H), 3.31 (q, J=7.1 Hz, 4H), 2.74-2.61 (m, 2H), 1.33 (t, J=7.1 Hz, 3H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 435.0 [M+H]+.


Ethyl 2-(1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidin-3-yl)acetate (DX3-43B)

Using a similar procedure as described for DX2-201 with 4a (40 mg, 0.13 mmol) and ethyl 2-(piperidin-3-yl)acetate (29 mg, 0.14 mmol), white solid (30 mg, 52%). 1H NMR (300 MHz, CDCl3) δ 7.98 (d, J=8.6 Hz, 2H), 7.89 (d, J=8.5 Hz, 2H), 4.16 (q, J=7.1 Hz, 2H), 3.67-3.49 (m, 2H), 3.30 (q, J=7.1 Hz, 4H), 2.59-2.47 (m, 1H), 2.37-2.09 (m, 4H), 1.84-1.63 (m, 4H), 1.28 (t, J=7.1 Hz, 3H), 1.16 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 447.2 [M+H]+.


Ethyl 1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)-4-hydroxy-1,2,5,6-tetrahydropyridine-3-carboxylate (DX3-45)

Using a similar procedure as described for DX2-201 with 4a (150 mg, 0.48 mmol) and ethyl 4-oxopiperidine-3-carboxylate (120 mg, 0.58 mmol), white solid (135 mg, 63%). 1H NMR (400 MHz, CDCl3) δ 12.06 (s, 1H), 8.03-7.93 (m, 4H), 4.27 (q, J=7.1 Hz, 2H), 3.85 (t, J=1.7 Hz, 2H), 3.37 (t, J=6.0 Hz, 2H), 3.31 (q, J=7.2 Hz, 4H), 2.49 (tt, J=6.0, 1.6 Hz, 2H), 1.34 (t, J=7.1 Hz, 3H), 1.18 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 475.1 [M+CH3CN+H]+.


N,N-Diethyl-4-((3-hydroxypiperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-46)

Using a similar procedure as described for DX2-201 with 4a (40 mg, 0.13 mmol) and ethyl piperidin-3-ol (16 mg, 0.16 mmol), white solid (30 mg, 61%). 1H NMR (400 MHz, CDCl3) δ 8.02-7.97 (m, 2H), 7.91 (d, J=8.6 Hz, 2H), 3.90 (s, 1H), 3.41 (dd, J=11.5, 3.6 Hz, 1H), 3.31 (q, J=7.2 Hz, 4H), 3.21 (dt, J=11.0, 5.1 Hz, 1H), 2.86 (ddd, J=11.8, 8.4, 3.4 Hz, 1H), 2.77 (dd, J=11.5, 7.3 Hz, 1H), 1.94-1.78 (m, 3H), 1.66 (dq, J=12.8, 4.3 Hz, 1H), 1.45 (dtd, J=12.6, 8.3, 3.7 Hz, 1H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 377.0 [M+H]+.


Ethyl 1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)-3-fluoropiperidine-3-carboxylate (DX3-47)

Using a similar procedure as described for DX2-201 with 4a (45 mg, 0.15 mmol) and ethyl 3-fluoropiperidine-3-carboxylate (28 mg, 0.16 mmol), white solid (41 mg, 60%). 1H NMR (400 MHz, CDCl3) δ 8.01-7.92 (m, 4H), 4.33-4.24 (m, 2H), 4.00 (ddt, J=13.5, 9.8, 1.9 Hz, 1H), 3.77 (d, J=12.5 Hz, 1H), 3.31 (q, J=7.1 Hz, 4H), 3.14 (dd, J=30.6, 13.4 Hz, 1H), 2.79-2.70 (m, 1H), 2.05 (d, J=11.7 Hz, 1H), 1.92-1.77 (m, 2H), 1.70 (dt, J=12.3, 3.3 Hz, 1H), 1.34 (t, J=7.1 Hz, 3H), 1.17 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 451.1 [M+H]+.


N,N-Diethyl-4-((3-fluoropiperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-49B)

Using a similar procedure as described for DX2-201 with 4a (40 mg, 0.13 mmol) and 3-fluoropiperidine (22 mg, 0.16 mmol), white solid (30 mg, 61%). 1H NMR (400 MHz, CDCl3) δ 7.99 (d, J=8.7 Hz, 2H), 7.93 (d, J=8.8 Hz, 2H), 4.80-4.61 (m, 1H), 3.40-3.34 (m, 1H), 3.34-3.22 (m, 5H), 3.14 (dq, J=9.9, 5.2, 4.5 Hz, 2H), 1.94-1.72 (m, 3H), 1.68-1.59 (m, 1H), 1.17 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 420.1 [M+CH3CN+H]+.


4-((3-Cyclohexylpiperidin-1-yl)sulfonyl)-N,N-diethylbenzenesulfonamide (DX3-50)

Using a similar procedure as described for DX2-201 with 4a (40 mg, 0.13 mmol) and 3-cyclohexylpiperidine (26 mg, 0.16 mmol), white solid (20 mg, 34%). 1H NMR (400 MHz, CDCl3) δ 8.01-7.96 (m, 2H), 7.89 (d, J=8.6 Hz, 2H), 3.76-3.66 (m, 2H), 3.31 (q, J=7.1 Hz, 4H), 2.31 (td, J=11.6, 2.8 Hz, 1H), 2.11 (t, J=11.0 Hz, 1H), 1.83-1.59 (m, 8H), 1.42 (dtt, J=10.4, 6.9, 3.4 Hz, 1H), 1.17 (t, J=7.1 Hz, 10H), 1.04-0.88 (m, 3H). LC-MS (ESI) m/z 484.3 [M+CH3CN+H]+.


Ethyl 3-benzyl-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX3-52B)

Using a similar procedure as described for DX2-201 with 4a (40 mg, 0.13 mmol) and ethyl 3-benzylpiperidine-3-carboxylate (38 mg, 0.16 mmol), white solid (40 mg, 59%). 1H NMR (400 MHz, CDCl3) δ 7.99 (d, J=8.7 Hz, 2H), 7.90 (d, J=8.5 Hz, 2H), 7.34-7.21 (m, 3H), 7.14-7.07 (m, 2H), 4.12 (qd, J=7.1, 3.2 Hz, 2H), 3.61 (d, J=11.6 Hz, 1H), 3.31 (q, J=7.2 Hz, 4H), 3.22-3.13 (m, 1H), 3.04 (d, J=13.4 Hz, 1H), 2.78 (d, J=13.1 Hz, 3H), 1.95 (dt, J=12.9, 5.3 Hz, 1H), 1.79 (qt, J=9.0, 4.4 Hz, 2H), 1.43 (ddd, J=13.4, 8.7, 4.7 Hz, 1H), 1.22 (t, J=7.2 Hz, 3H), 1.18 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 523.2 [M+H]+.


(trans)-Ethyl-8-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)-8-azabicyclo[3.2.1]octane-2-carboxylate (DX3-69B-P1) and (cis)-ethyl-8-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)-8-azabicyclo[3.2.1]octane-2-carboxylate (DX3-69B-P2)

Using a similar procedure as described for DX2-201 with 4a (34 mg, 0.11 mmol) and ethyl-8-azabicyclo[3.2.1]octane-2-carboxylate (38 mg, 0.16 mmol), and purified with preparative-HPLC to give the two diastereoisomers. DX3-69B-P1 (20 mg, 40%). 1H NMR (400 MHz, CDCl3) δ 7.99 (d, J=8.1 Hz, 2H), 7.93 (d, J=8.7 Hz, 2H), 4.71 (dd, J=7.2, 2.8 Hz, 1H), 4.33-4.23 (m, 1H), 4.19-4.04 (m, 1H), 4.01-3.88 (m, 1H), 3.28 (q, J=7.1 Hz, 4H), 2.52 (dd, J=6.4, 2.9 Hz, 1H), 2.11-1.97 (m, 2H), 1.88-1.58 (m, 5H), 1.48 (ddd, J=13.0, 6.3, 3.4 Hz, 1H), 1.23 (t, J=7.1 Hz, 3H), 1.15 (t, J=7.0 Hz, 6H). LC-MS (ESI) m/z 459.1 [M+H]+. DX3-69B-P2 (27 mg, 53%). 1H NMR (400 MHz, CDCl3) δ 8.02 (d, J=8.5 Hz, 2H), 7.94 (d, J=8.4 Hz, 2H), 4.51 (dd, J=6.0, 2.8 Hz, 1H), 4.27 (d, J=3.1 Hz, 1H), 4.15 (q, J=7.1 Hz, 2H), 3.29 (q, J=7.2 Hz, 4H), 2.83 (ddd, J=12.2, 5.2, 2.8 Hz, 1H), 1.98-1.51 (m, 8H), 1.27 (t, J=7.1 Hz, 3H), 1.14 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 459.1 [M+H]+.


Ethyl 3-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)-3-azabicyclo[3.1.1]heptane-1-carboxylate (DX3-115)

Using a similar procedure as described for DX2-201 with 4a (20 mg, 0.064 mmol) and ethyl 3-azabicyclo[3.1.1]heptane-1-carboxylate hydrochloride (11 mg, 0.064 mmol), white solid (21 mg, 75%). 1H NMR (300 MHz, CDCl3) δ 7.99 (s, 4H), 4.15 (q, J=7.1 Hz, 2H), 3.68 (s, 2H), 3.52 (d, J=2.5 Hz, 2H), 3.31 (q, J=7.2 Hz, 4H), 2.50-2.35 (m, 3H), 1.42 (dd, J=7.0, 2.7 Hz, 2H), 1.28 (t, J=7.1 Hz, 3H), 1.16 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 445.1 [M+H]+.


Ethyl (R)-1-((4-bromophenyl)sulfonyl)piperidine-3-carboxylate (2b)

Using a similar procedure as described for 2a with 4-bromobenzenesulfonyl chloride (1a, 1.50 g, 5.86 mmol) and ethyl (R)-piperidine-3-carboxylate (0.92 g, 5.86 mmol), white solid (2.16 g, 98%). 1H NMR (300 MHz, CDCl3) δ 7.67 (q, J=8.6 Hz, 4H), 4.16 (q, J=7.2 Hz, 2H), 3.85 (d, J=11.0 Hz, 1H), 3.63 (d, J=11.8 Hz, 1H), 2.68-2.49 (m, 2H), 2.38 (td, J=11.3, 3.2 Hz, 1H), 2.02 (d, J=12.6 Hz, 1H), 1.88-1.77 (m, 1H), 1.77-1.61 (m, 1H), 1.50-1.34 (m, 1H), 1.28 (t, J=7.1 Hz, 3H).


Ethyl (R)-1-((4-(benzylthio)phenyl)sulfonyl)piperidine-3-carboxylate (3b)

Using a similar procedure as described for 4a with 2b (1.20 g, 3.19 mmol) and benzyl mercaptan (0.40 g, 3.19 mmol), white solid (1.17 g, 87%). 1H NMR (300 MHz, CDCl3) δ 7.63 (d, J=8.5 Hz, 2H), 7.43-7.26 (m, 7H), 4.24 (s, 2H), 4.15 (q, J=7.1 Hz, 2H), 3.84 (d, J=11.3 Hz, 1H), 3.61 (d, J=11.2 Hz, 1H), 2.69-2.56 (m, 1H), 2.50 (t, J=10.8 Hz, 1H), 2.34 (td, J=11.2, 3.0 Hz, 1H), 2.00 (d, J=13.8 Hz, 1H), 1.87-1.75 (m, 1H), 1.73-1.57 (m, 1H), 1.47-1.34 (m, 1H), 1.28 (t, J=7.1 Hz, 3H).


Ethyl (R)-1-((4-(chlorosulfonyl)phenyl)sulfonyl)piperidine-3-carboxylate (4b)

Using a similar procedure as described for 4a with 3b (600 mg, 1.43 mmol), white solid (520 mg, 92%). 1H NMR (300 MHz, CDCl3) δ 8.23 (d, J=8.5 Hz, 2H), 8.03 (d, J=8.5 Hz, 2H), 4.16 (q, J=7.1 Hz, 2H), 3.86 (t, J=8.7 Hz, 1H), 3.67 (d, J=11.7 Hz, 1H), 2.73-2.57 (m, 2H), 2.50 (td, J=11.3, 3.1 Hz, 1H), 2.10-1.98 (m, 1H), 1.92-1.80 (m, 1H), 1.77-1.60 (m, 1H), 1.47 (s, 1H), 1.28 (t, J=7.1 Hz, 3H).


Ethyl (R)-1-((4-(morpholinosulfonyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-245)

Using a similar procedure as described for DX2-201 with 4b (40 mg, 0.10 mmol) and morpholine (11 mg, 0.12 mmol), white solid (36 mg, 81%). 1H NMR (300 MHz, CDCl3) δ 8.02-7.89 (m, 4H), 4.17 (q, J=7.1 Hz, 2H), 3.88 (d, J=7.4 Hz, 1H), 3.82-3.74 (m, 4H), 3.66 (d, J=11.7 Hz, 1H), 3.08 (dd, J=5.8, 3.6 Hz, 4H), 2.65 (d, J=8.8 Hz, 2H), 2.53-2.40 (m, 1H), 2.04 (d, J=10.1 Hz, 2H), 1.92-1.59 (m, 2H), 1.48 (s, 1H), 1.28 (t, J=7.1 Hz, 3H). LC-MS (ESI) m/z 447.0 [M+H]+.


Ethyl (R)-1-((4-(piperazin-1-ylsulfonyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-248)

Using a similar procedure as described for DX2-201 with 4b (40 mg, 0.10 mmol) and piperazine (11 mg, 0.12 mmol), white solid (4 mg, 9%). 1H NMR (400 MHz, CDCl3) δ 8.00-7.90 (m, 4H), 4.17 (q, J=7.1 Hz, 2H), 3.88 (d, J=8.1 Hz, 1H), 3.70-3.62 (m, 1H), 3.17 (t, J=4.9 Hz, 4H), 3.05 (t, J=5.0 Hz, 4H), 2.70-2.63 (m, 2H), 2.48 (td, J=11.3, 3.2 Hz, 1H), 2.07-2.00 (m, 1H), 1.85 (dt, J=13.7, 3.8 Hz, 1H), 1.74-1.63 (m, 1H), 1.54-1.42 (m, 1H), 1.29 (t, J=7.1 Hz, 3H). LC-MS (ESI) m/z 446.0 [M+H]+.


Ethyl (R)-1-((4-(N-(oxetan-3-yl)sulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-249)

Using a similar procedure as described for DX2-201 with 4b (40 mg, 0.10 mmol) and oxetan-3-amine hydrochloride (13 mg, 0.12 mmol), white solid (25 mg, 58%). 1H NMR (400 MHz, CDCl3) δ 8.03 (d, J=8.5 Hz, 2H), 7.94 (d, J=8.5 Hz, 2H), 5.48 (d, J=9.0 Hz, 1H), 4.80 (t, J=7.1 Hz, 2H), 4.66-4.53 (m, 1H), 4.49-4.41 (m, 2H), 4.17 (q, J=7.1 Hz, 2H), 3.87 (d, J=8.0 Hz, 1H), 3.66 (d, J=11.6 Hz, 1H), 2.64 (dd, J=9.2, 4.5 Hz, 2H), 2.45 (td, J=11.3, 3.1 Hz, 1H), 2.05 (dd, J=13.0, 4.3 Hz, 1H), 1.85 (dt, J=13.5, 3.8 Hz, 1H), 1.70 (td, J=10.7, 10.1, 5.1 Hz, 1H), 1.52-1.38 (m, 1H), 1.29 (t, J=7.1 Hz, 3H). LC-MS (ESI) m/z 433.0 [M+H]+.


Ethyl (3R)-1-((4-((2,6-dimethylpiperidin-1-yl)sulfonyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-258)

To a solution of 4b (20 mg, 0.05 mmol) and 2,6-dimethylpiperidine (7 mg, 0.06 mmol) in CH3CN was added Cs2CO3 (32 mg, 0.10 mmol). The mixture was heated at 60° C. for 1 h and diluted with EtOAc, washed with water, brine, dried over anhydrous Na2SO4, filtered and purified with flash chromatography (20% EtOAc in hexane) to give DX2-258 as a white solid (13 mg, 52%). 1H NMR (300 MHz, CDCl3) δ 8.00 (d, J=8.2 Hz, 2H), 7.89 (t, J=7.2 Hz, 2H), 4.26-4.04 (m, 4H), 3.91-3.80 (m, 1H), 3.71-3.57 (m, 1H), 2.68-2.56 (m, 2H), 2.43 (td, J=11.6, 2.2 Hz, 1H), 2.09-1.97 (m, 1H), 1.89-1.60 (m, 7H), 1.56-1.45 (m, 2H), 1.39 (d, J=7.1 Hz, 3H), 1.28 (t, J=6.6 Hz, 6H). LC-MS (ESI) m/z 473.1 [M+H]+.


Ethyl (R)-1-((4-(N,N-dimethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-259)

Using a similar procedure as described for DX2-201 with 4b (40 mg, 0.10 mmol) and dimethylamine (5.4 mg, 0.12 mmol), white solid (16 mg, 40%). 1H NMR (300 MHz, CDCl3) δ 7.96 (s, 4H), 4.17 (q, J=7.2 Hz, 2H), 3.88 (d, J=7.4 Hz, 1H), 3.67 (d, J=11.9 Hz, 1H), 2.79 (s, 6H), 2.64 (d, J=8.4 Hz, 2H), 2.46 (td, J=11.6, 3.3 Hz, 1H), 2.06 (s, 2H), 1.90-1.80 (m, 1H), 1.77-1.62 (m, 1H), 1.54-1.40 (m, 1H), 1.28 (t, J=7.1 Hz, 3H). LC-MS (ESI) m/z 405.0 [M+H]+.


Ethyl (R)-1-((4-(N,N-dipropylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-264)

Using a similar procedure as described for DX2-201 with 4b (40 mg, 0.10 mmol) and dipropylamine (12 mg, 0.12 mmol), white solid (20 mg, 43%). 1H NMR (400 MHz, CDCl3) δ 7.99 (d, J=8.6 Hz, 2H), 7.91 (d, J=8.5 Hz, 2H), 4.17 (q, J=7.1 Hz, 2H), 3.87 (d, J=7.4 Hz, 1H), 3.65 (d, J=11.7 Hz, 1H), 3.20-3.11 (m, 4H), 2.68-2.59 (m, 2H), 2.45 (td, J=11.4, 3.1 Hz, 1H), 2.08-2.00 (m, 1H), 1.84 (dt, J=13.7, 3.7 Hz, 1H), 1.71 (dq, J=11.2, 3.6, 3.2 Hz, 1H), 1.64-1.53 (m, 4H), 1.46 (dd, J=12.6, 9.0 Hz, 1H), 1.29 (t, J=7.1 Hz, 3H), 0.90 (t, J=7.4 Hz, 6H). LC-MS (ESI) m/z 461.0 [M+H]+.


Ethyl (R)-1-((4-(piperidin-1-ylsulfonyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-265)

Using a similar procedure as described for DX2-201 with 4b (40 mg, 0.10 mmol) and piperidine (10 mg, 0.12 mmol), white solid (20 mg, 45%). 1H NMR (400 MHz, CDCl3) δ 7.94 (s, 4H), 4.17 (q, J=7.1 Hz, 2H), 3.88 (d, J=8.0 Hz, 1H), 3.67 (d, J=11.7 Hz, 1H), 3.15-3.03 (m, 4H), 2.71-2.60 (m, 2H), 2.48 (td, J=11.4, 3.1 Hz, 1H), 2.04 (dd, J=12.8, 3.9 Hz, 1H), 1.85 (dt, J=13.7, 3.8 Hz, 1H), 1.69 (hept, J=7.6, 6.8 Hz, 5H), 1.50 (q, J=6.3, 5.8 Hz, 3H), 1.29 (t, J=7.1 Hz, 3H). LC-MS (ESI) m/z 445.0 [M+H]+.


Ethyl (R)-1-((4-(N,N-diisopropylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-280)

Using a similar procedure as described for DX2-258 with 4b (60 mg, 0.15 mmol) and diisopropylamine (11 mg, 0.30 mmol) and purified with flash chromatography (20% EtOAc in hexane), white solid (11 mg, 16%). 1H NMR (300 MHz, CDCl3) δ 8.04 (d, J=8.2 Hz, 2H), 7.89 (d, J=8.4 Hz, 2H), 4.16 (q, J=7.2 Hz, 2H), 3.93-3.81 (m, 1H), 3.77 (p, J=6.8 Hz, 2H), 3.63 (d, J=11.6 Hz, 1H), 2.62 (d, J=8.8 Hz, 2H), 2.52-2.35 (m, 1H), 2.10-1.96 (m, 1H), 1.91-1.75 (m, 1H), 1.73-1.57 (m, 1H), 1.57-1.38 (m, 1H), 1.35-1.21 (m, 15H). LC-MS (ESI) m/z 461.1 [M+H]+.


Ethyl (R)-1-((4-sulfamoylphenyl)sulfonyl)piperidine-3-carboxylate (DX2-300)

4b (40 mg, 0.10 mmol) was dissolved in NH3 in dioxane solution (1 mL, 4M) and stirred at room temperature for 3 h. The mixture was concentrated and purified with flash chromatography (10% MeOH in DCM) to give DX-300 as a white solid (30 mg, 80%). 1H NMR (300 MHz, DMSO) (8.26 (d, J=8.5 Hz, 2H), 8.16 (d, J=8.6 Hz, 2H), 4.27 (q, J=7.1 Hz, 2H), 3.56-3.45 (m, 1H), 2.99-2.70 (m, 4H), 2.04-1.86 (m, 2H), 1.78-1.55 (m, 2H), 1.38 (t, J=7.1 Hz, 3H). LC-MS (ESI) m/z 377.0 [M+H]+.


Ethyl (R)-1-((4-(N-benzyl-N-methylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX3-86B)

Using a similar procedure as described for DX2-201 with 4b (20 mg, 0.05 mmol) and N-methyl-1-phenylmethanamine (6 mg, 0.05 mmol), white solid (13 mg, 54%). 1H NMR (300 MHz, CDCl3) δ 8.05-7.92 (m, 4H), 7.42-7.30 (m, 5H), 4.24 (s, 2H), 4.17 (q, J=7.1 Hz, 2H), 3.88 (t, J=8.7 Hz, 1H), 3.68 (d, J=11.6 Hz, 1H), 2.70 (s, 3H), 2.65 (dd, J=8.8, 2.2 Hz, 2H), 2.47 (td, J=11.3, 3.1 Hz, 1H), 2.12-2.00 (m, 1H), 1.86 (dt, J=13.3, 3.7 Hz, 1H), 1.78-1.63 (m, 1H), 1.53-1.38 (m, 1H), 1.29 (t, J=7.1 Hz, 3H). LC-MS (ESI) m/z 481.0 [M+H]+.


Ethyl (R)-1-((4-(N-ethyl-N-isopropylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX3-105)

Using a similar procedure as described for DX2-201 with 4b (20 mg, 0.05 mmol) and N-ethylpropan-2-amine (4.4 mg, 0.05 mmol), white solid (11 mg, 50%). 1H NMR (300 MHz, CDCl3) δ 8.01 (d, J=8.5 Hz, 2H), 7.90 (d, J=8.5 Hz, 2H), 4.14 (dq, J=11.8, 6.9 Hz, 3H), 3.86 (d, J=7.9 Hz, 1H), 3.64 (d, J=11.6 Hz, 1H), 3.25 (q, J=7.1 Hz, 2H), 2.69-2.55 (m, 2H), 2.43 (td, J=11.3, 3.1 Hz, 1H), 2.09-1.97 (m, 1H), 1.84 (dt, J=13.3, 3.9 Hz, 1H), 1.75-1.61 (m, 1H), 1.51-1.36 (m, 1H), 1.29 (td, J=7.1, 6.2 Hz, 6H), 1.11 (d, J=6.8 Hz, 6H). LC-MS (ESI) m/z 447.1 [M+H]+.


4-Bromo-N,N-diethyl-2-fluorobenzenesulfonamide (2c)

Using a similar procedure as described for 2a with 1c (1.00 g, 3.65 mmol) and diethylamine (320 mg, 4.38 mmol), white solid (861 mg, 76%). 1H NMR (400 MHz, CDCl3) δ 7.84-7.75 (m, 1H), 7.46-7.36 (m, 2H), 3.36 (q, J=7.1 Hz, 4H), 1.17 (t, J=7.2 Hz, 6H).


4-(Benzylthio)-N,N-diethyl-2-fluorobenzenesulfonamide (3c)

Using a similar procedure as described for 3a with 2c (861 mg, 2.78 mmol) and benzyl mercaptan (344 mg, 2.78 mmol), yellow solid (950 mg, 97%). 1H NMR (400 MHz, CDCl3) δ 7.76 (dd, J=8.3, 7.5 Hz, 1H), 7.41-7.29 (m, 5H), 7.10 (dd, J=8.3, 1.8 Hz, 1H), 7.05 (dd, J=10.8, 1.8 Hz, 1H), 4.22 (s, 2H), 3.34 (q, J=7.1 Hz, 4H), 1.15 (t, J=7.2 Hz, 6H).


4-(N,N-Diethylsulfamoyl)-3-fluorobenzenesulfonyl chloride (4c)

Using a similar procedure as described for 4a with 3c (500 mg, 1.42 mmol), yellow solid (403 mg, 86). 1H NMR (400 MHz, CDCl3) δ 8.21 (dd, J=8.3, 6.6 Hz, 1H), 7.98-7.93 (m, 1H), 7.88 (dd, J=8.6, 1.8 Hz, 1H), 3.46-3.38 (m, 4H), 1.20 (t, J=7.1 Hz, 6H).


Ethyl (R)-1-((4-(N,N-diethylsulfamoyl)-3-fluorophenyl)sulfonyl)piperidine-3-carboxylate (DX2-275)

Using a similar procedure as described for DX2-201 with 4c (60 mg, 0.18 mmol) and ethyl (R)-piperidine-3-carboxylate (31 mg, 0.20 mmol), white solid (60 mg, 74%). 1H NMR (300 MHz, CDCl3) δ 8.15-8.05 (m, 1H), 7.62 (dd, J=15.2, 8.6 Hz, 2H), 4.17 (q, J=7.1 Hz, 2H), 3.85 (d, J=8.5 Hz, 1H), 3.63 (dd, J=12.8, 5.4 Hz, 1H), 3.40 (q, J=7.1 Hz, 4H), 2.67 (q, J=9.6 Hz, 2H), 2.50 (t, J=9.8 Hz, 1H), 2.11-1.98 (m, 1H), 1.93-1.80 (m, 1H), 1.75-1.59 (m, 1H), 1.57-1.39 (m, 1H), 1.29 (t, J=7.1 Hz, 3H), 1.18 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 451.0 [M+H]+.


4-Bromo-N,N-diethyl-3-methylbenzenesulfonamide (2d)

Using a similar procedure as described for 2a with 1d (1.00 g, 3.70 mmol) and diethylamine (324 mg, 4.44 mmol), white solid (950 mg, 84%). 1H NMR (400 MHz, CDCl3) δ 7.68-7.62 (m, 2H), 7.50-7.43 (m, 1H), 3.23 (q, J=7.2 Hz, 4H), 2.45 (s, 3H), 1.13 (t, J=7.2 Hz, 6H).


4-(Benzylthio)-N,N-diethyl-3-methylbenzenesulfonamide (3d)

Using a similar procedure as described for 3a with 2d (950 mg, 3.10 mmol) and benzyl mercaptan (384 mg, 3.10 mmol), yellow solid (900 mg, 83%). 1H NMR (400 MHz, CDCl3) δ 7.61-7.53 (m, 2H), 7.41-7.26 (m, 6H), 4.21 (s, 2H), 3.24 (q, J=7.1 Hz, 4H), 2.37 (s, 3H), 1.15 (t, J=7.2 Hz, 6H).


4-(N,N-Diethylsulfamoyl)-2-methylbenzenesulfonyl chloride (4d)

Using a similar procedure as described for 4a with 3d (500 mg, 1.43 mmol), yellow solid (mg, %). 1H NMR (400 MHz, CDCl3) δ 7.97 (d, J=8.2 Hz, 1H), 7.65 (s, 1H), 7.61 (d, J=8.1 Hz, 1H), 3.22 (q, J=7.2 Hz, 4H), 2.61 (s, 3H), 1.14 (t, J=7.1 Hz, 6H).


Ethyl (R)-1-((4-(N,N-diethylsulfamoyl)-2-methylphenyl)sulfonyl)piperidine-3-carboxylate (DX2-276)

Using a similar procedure as described for DX2-201 with 4d (60 mg, 0.18 mmol) and ethyl (R)-piperidine-3-carboxylate (31 mg, 0.20 mmol), white solid (41 mg, 51%). 1H NMR (300 MHz, CDCl3) δ 8.04 (d, J=8.1 Hz, 1H), 7.79-7.70 (m, 2H), 4.21-4.03 (m, 2H), 3.81 (ddt, J=12.5, 4.0, 1.5 Hz, 1H), 3.62 (d, J=12.5 Hz, 1H), 3.29 (q, J=7.1 Hz, 4H), 2.95 (dd, J=12.5, 10.0 Hz, 1H), 2.86-2.75 (m, 1H), 2.68 (s, 3H), 2.60 (ddd, J=10.2, 6.2, 4.0 Hz, 1H), 2.12-2.02 (m, 1H), 1.88-1.80 (m, 1H), 1.73-1.54 (m, 2H), 1.25 (t, J=7.1 Hz, 3H), 1.17 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 447.1 [M+H]+.




embedded image


2-(Benzylthio)-5-bromopyridine (6)

To a solution of 5 (2.00 g, 10.42 mmol) and benzyl mercaptan (1.30 g, 10.42 mmol) in DMF (40 mL) was added Cs2CO3 (6.80 g, 20.84 mmol). The mixture was stirred at room temperature for 3 h, then diluted with EtOAc, washed with water, brine, dried over anhydrous Na2SO4, filtered and purified with flash chromatography to give 6 as a colorless oil (1.96 g, 67%). 1H NMR (300 MHz, CDCl3) δ 8.57-8.49 (m, 1H), 7.59 (dd, J=8.6, 2.4 Hz, 1H), 7.42 (d, J=7.4 Hz, 2H), 7.37-7.22 (m, 3H), 7.08 (d, J=8.5 Hz, 1H), 4.43 (s, 2H). LC-MS (ESI) m/z 279.8, 281.7 [M+H]+.


5-Bromopyridine-2-sulfonyl chloride (7)

Using a similar procedure as described for 4a with 6 (1.86 g, 6.64 mmol), white solid (1.61 g, 94%). 1H NMR (300 MHz, CDCl3) δ 8.92-8.86 (m, 1H), 8.21 (dd, J=9.0, 1.6 Hz, 1H), 8.02 (d, J=8.4 Hz, 1H).


5-Bromo-N,N-diethylpyridine-2-sulfonamide (8a)

Using a similar procedure as described for DX2-201 with 7 (800 mg, 3.11 mmol) and diethylamine (250 mg, 3.42 mmol), white solid (422 mg, 74%). 1H NMR (300 MHz, CDCl3) δ 8.74 (d, J=2.2 Hz, 1H), 8.03 (dd, J=8.3, 2.3 Hz, 1H), 7.87 (d, J=8.3 Hz, 1H), 3.41 (q, J=7.2 Hz, 4H), 1.18 (t, J=7.1 Hz, 6H).


5-(Benzylthio)-N,N-diethylpyridine-2-sulfonamide (9a)

Using a similar procedure as described for 3a with 8a (280 mg, 0.95 mmol) and benzyl mercaptan (119 mg, 0.95 mmol), white solid (287 mg, 90%). 1H NMR (400 MHz, CDCl3) δ 8.52 (dd, J=2.4, 0.8 Hz, 1H), 7.83-7.78 (m, 1H), 7.67 (dd, J=8.2, 2.3 Hz, 1H), 7.36-7.29 (m, 5H), 4.22 (s, 2H), 3.39 (q, J=7.1 Hz, 4H), 1.15 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 337.0 [M+H]+.


6-(N,N-Diethylsulfamoyl)pyridine-3-sulfonyl chloride (10a)

Using a similar procedure as described for 4a with 9a (80 mg, 0.24 mmol), white solid (75 mg, 75%). 1H NMR (300 MHz, CDCl3) δ 9.27 (d, J=2.4 Hz, 1H), 8.51 (dd, J=8.4, 2.3 Hz, 1H), 8.22 (d, J=8.3 Hz, 1H), 3.47 (q, J=7.2 Hz, 4H), 1.22 (t, J=7.2 Hz, 6H).


Ethyl (R)-1-((6-(N,N-diethylsulfamoyl)pyridin-3-yl)sulfonyl)piperidine-3-carboxylate (DX2-291)

Using a similar procedure as described for DX2-201 with 10a (72 mg, 0.23 mmol) and ethyl (R)-piperidine-3-carboxylate (40 mg, 0.25 mmol), white solid (40 mg, 40%). 1H NMR (300 MHz, CDCl3) δ 9.02 (d, J=1.7 Hz, 1H), 8.28-8.21 (m, 1H), 8.13 (d, J=8.2 Hz, 1H), 4.17 (q, J=7.1 Hz, 2H), 3.87 (d, J=9.8 Hz, 1H), 3.71-3.61 (m, 1H), 3.45 (q, J=7.1 Hz, 4H), 2.69 (q, J=10.1 Hz, 2H), 2.58-2.47 (m, 1H), 2.11-2.00 (m, 1H), 1.92-1.80 (m, 1H), 1.75-1.59 (m, 1H), 1.56-1.42 (m, 1H), 1.28 (t, J=7.1 Hz, 3H), 1.20 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 434.0 [M+H]+.


Ethyl (R)-1-((5-bromopyridin-2-yl)sulfonyl)piperidine-3-carboxylate (8b)

Using a similar procedure as described for DX2-201 with 7 (660 mg, 2.57 mmol) and ethyl (R)-piperidine-3-carboxylate (444 mg, 2.82 mmol), white solid (120 mg, 12%). 1H NMR (300 MHz, CDCl3) δ 8.74 (d, J=2.3 Hz, 1H), 8.04 (dd, J=8.3, 2.3 Hz, 1H), 7.83 (d, J=8.3 Hz, 1H), 4.11 (dt, J=7.2, 4.5 Hz, 2H), 4.00 (dd, J=12.3, 4.0 Hz, 1H), 3.77 (d, J=12.5 Hz, 1H), 3.56-3.44 (m, 1H), 3.00-2.89 (m, 1H), 2.81 (dd, J=11.6, 3.0 Hz, 1H), 2.60 (tt, J=10.6, 3.9 Hz, 1H), 1.86-1.73 (m, 1H), 1.70-1.58 (m, 2H), 1.34-1.16 (m, 3H). LC-MS (ESI) m/z 376.9, 378.9 [M+H]+.


Ethyl (R)-1-((5-(benzylthio)pyridin-2-yl)sulfonyl)piperidine-3-carboxylate (9b)

Using a similar procedure as described for 3a with 8b (120 mg, 0.32 mmol) and benzyl mercaptan (39 mg, 0.32 mmol), white solid (81 mg, 60%). 1H NMR (300 MHz, CDCl3) δ 8.53 (d, J=2.3 Hz, 1H), 7.78 (d, J=8.2 Hz, 1H), 7.69 (dd, J=8.2, 2.3 Hz, 1H), 7.41-7.30 (m, 5H), 4.14 (qd, J=7.1, 2.2 Hz, 2H), 4.00 (d, J=10.5 Hz, 1H), 3.78 (d, J=12.6 Hz, 1H), 2.93-2.82 (m, 1H), 2.74 (td, J=11.7, 3.0 Hz, 1H), 2.67-2.54 (m, 1H), 2.06 (s, 1H), 1.80 (dt, J=12.5, 3.6 Hz, 1H), 1.70-1.57 (m, 1H), 1.57-1.41 (m, 1H), 1.34-1.22 (m, 3H). LC-MS (ESI) m/z 421.1 [M+H]+.


Ethyl (R)-1-((5-(chlorosulfonyl)pyridin-2-yl)sulfonyl)piperidine-3-carboxylate (10b)

Using a similar procedure as described for 4a with 9b (50 mg, 0.12 mmol), white solid (44 mg, 92%). 1H NMR (300 MHz, CDCl3) δ 9.30 (d, J=2.3 Hz, 1H), 8.54 (dd, J=8.4, 2.3 Hz, 1H), 8.21 (d, J=8.4 Hz, 1H), 4.16 (q, J=7.1 Hz, 2H), 4.06 (d, J=11.0 Hz, 1H), 3.85 (d, J=12.3 Hz, 1H), 3.17-3.06 (m, 1H), 2.98 (t, J=11.5 Hz, 1H), 2.72-2.61 (m, 1H), 2.14-2.04 (m, 1H), 1.93-1.82 (m, 1H), 1.74-1.56 (m, 2H), 1.28 (t, J=7.1 Hz, 3H).


Ethyl (R)-1-((5-(N,N-diethylsulfamoyl)pyridin-2-yl)sulfonyl)piperidine-3-carboxylate (DX2-293)

Using a similar procedure as described for DX2-201 with 10b (44 mg, 0.12 mmol) and diethylamine (10 mg, 0.13 mmol), white solid (24 mg, 46%). 1H NMR (300 MHz, CDCl3) δ 9.08 (d, J=2.1 Hz, 1H), 8.31 (dd, J=8.2, 2.3 Hz, 1H), 8.08 (d, J=8.2 Hz, 1H), 4.16 (q, J=7.1 Hz, 2H), 4.04 (dd, J=9.4, 6.2 Hz, 1H), 3.83 (dt, J=12.1, 3.9 Hz, 1H), 3.32 (q, J=7.1 Hz, 4H), 3.10-2.99 (m, 1H), 2.88 (t, J=11.7 Hz, 1H), 2.71-2.58 (m, 1H), 2.14-2.03 (m, 1H), 1.91-1.79 (m, 1H), 1.71-1.49 (m, 3H), 1.28 (t, J=7.1 Hz, 3H), 1.20 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 434.0 [M+H]+.




embedded image


2-(N,N-Diethylsulfamoyl)benzenesulfonyl chloride (12a)

To a solution of diethylamine (21 mg, 0.29 mmol) and triethylamine (88 mg, 0.84 mmol) in DCM (2 mL) was added benzene-1,2-disulfonyl dichloride (11a, 80 mg, 0.29 mmol) at 0° C. The mixture was stirred at room temperature overnight. The mixture was concentrated and purified with flash chromatography (30% EtOAc in hexane) to give 12a as a white solid (15 mg, 17%). 1H NMR (300 MHz, CDCl3) δ 8.40 (dd, J=7.7, 1.6 Hz, 1H), 8.17 (dd, J=7.6, 1.7 Hz, 1H), 7.94-7.75 (m, 2H), 3.46 (q, J=7.3 Hz, 4H), 1.19 (t, J=6.9 Hz, 6H).


Ethyl (R)-1-((2-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-226)

Using a similar procedure as described for DX2-201 with 12a (15 mg, 0.048 mmol) and ethyl (R)-piperidine-3-carboxylate (8.3 mg, 0.053 mmol), white solid (5 mg, 24%). 1H NMR (300 MHz, CDCl3) δ 8.05 (d, J=8.2 Hz, 2H), 7.97 (d, J=8.3 Hz, 2H), 4.17 (qd, J=7.1, 3.0 Hz, 2H), 3.69 (s, 1H), 3.45 (s, 1H), 3.36-3.22 (m, 4H), 2.72-2.55 (m, 2H), 2.40 (t, J=12.5 Hz, 1H), 2.04-1.41 (m, 4H), 1.28 (t, J=7.1 Hz, 3H), 1.16 (t, J=7.0 Hz, 6H). LC-MS (ESI) m/z 433.1 [M+H]+.


3-(N,N-Diethylsulfamoyl)benzenesulfonyl chloride (12b)

Using a similar procedure as described for 12a with benzene-1,3-disulfonyl dichloride (11b, 275 mg, 1.00 mmol) and diethylamine (73 mg, 1.00 mmol), white solid (97 mg, 31%). 1H NMR (300 MHz, CDCl3) δ 8.47 (d, J=1.9 Hz, 1H), 8.21 (ddt, J=13.0, 7.9, 1.5 Hz, 2H), 7.82 (t, J=7.9 Hz, 1H), 3.32 (q, J=7.2 Hz, 4H), 1.19 (t, J=7.1 Hz, 6H).


Ethyl (R)-1-((3-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-229)

Using a similar procedure as described for DX2-201 with 12b (47 mg, 0.15 mmol) and ethyl (R)-piperidine-3-carboxylate (27 mg, 0.18 mmol), white solid (22 mg, 34%). 1H NMR (300 MHz, CDCl3) δ 8.20 (d, J=1.8 Hz, 1H), 8.05 (dd, J=7.9, 1.6 Hz, 1H), 7.96 (d, J=8.0 Hz, 1H), 7.71 (t, J=7.8 Hz, 1H), 4.16 (q, J=7.1 Hz, 2H), 3.83 (d, J=7.5 Hz, 1H), 3.63 (d, J=11.7 Hz, 1H), 3.29 (q, J=7.1 Hz, 4H), 2.70-2.56 (m, 2H), 2.46 (td, J=11.2, 3.2 Hz, 1H), 2.00 (d, J=13.6 Hz, 1H), 1.84 (dt, J=13.3, 3.9 Hz, 1H), 1.75-1.57 (m, 1H), 1.44 (q, J=15.3, 13.1 Hz, 1H), 1.28 (t, J=7.1 Hz, 3H), 1.16 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 433.0 [M+H]+.


4-(4-(N,N-Diethylsulfamoyl)phenoxy)benzenesulfonyl chloride (12c)

Using a similar procedure as described for 12a with 4,4′-oxydibenzenesulfonyl chloride (11c, 200 mg, 0.54 mmol) and diethylamine (40 mg, 0.54 mmol), white solid (72 mg, 33%). 1H NMR (300 MHz, CDCl3) δ 8.06 (d, J=9.0 Hz, 2H), 7.90 (d, J=8.7 Hz, 2H), 7.20 (dd, J=8.8, 6.7 Hz, 4H), 3.30 (q, J=7.1 Hz, 4H), 1.18 (t, J=7.1 Hz, 6H).


Ethyl (R)-1-((4-(4-(N,N-diethylsulfamoyl)phenoxy)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-230)

Using a similar procedure as described for DX2-201 with 12c (14 mg, 0.039 mmol) and ethyl (R)-piperidine-3-carboxylate (7 mg, 0.042 mmol), white solid (5 mg, 25%). 1H NMR (300 MHz, CDCl3) δ 7.83 (dd, J=20.6, 8.3 Hz, 4H), 7.15 (d, J=8.3 Hz, 4H), 4.16 (q, J=7.1 Hz, 2H), 3.88 (d, J=11.3 Hz, 1H), 3.67 (d, J=11.2 Hz, 1H), 3.29 (q, J=7.2 Hz, 4H), 2.60 (dt, J=33.2, 10.8 Hz, 2H), 2.39 (t, J=11.1 Hz, 1H), 2.03 (d, J=13.2 Hz, 1H), 1.91-1.78 (m, 1H), 1.69 (d, J=12.4 Hz, 1H), 1.51-1.34 (m, 1H), 1.28 (t, J=7.1 Hz, 3H), 1.18 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 525.1 [M+H]+.


4′-(N,N-Diethylsulfamoyl)-[1,1′-biphenyl]-4-sulfonyl chloride (12d)

Using a similar procedure as described for 12a with [1,1′-biphenyl]-4,4′-disulfonyl dichloride (11d, 200 mg, 0.57 mmol) and diethylamine (42 mg, 0.57 mmol), white solid (47 mg, 21%). 1H NMR (300 MHz, CDCl3) δ 8.21-8.11 (m, 2H), 7.99-7.93 (m, 2H), 7.85 (d, J=8.7 Hz, 2H), 7.76 (d, J=8.0 Hz, 2H), 3.31 (q, J=7.1 Hz, 4H), 1.18 (t, J=7.1 Hz, 6H).


Ethyl (R)-1-((4′-(N,N-diethylsulfamoyl)-[1,1′-biphenyl]-4-yl)sulfonyl)piperidine-3-carboxylate (DX3-78)

Using a similar procedure as described for DX2-201 with 12d (45 mg, 0.12 mmol) and ethyl (R)-piperidine-3-carboxylate (18 mg, 0.12 mmol), white solid (39 mg, 64%). 1H NMR (300 MHz, CDCl3) δ 7.92 (dd, J=16.5, 8.5 Hz, 4H), 7.76 (t, J=8.6 Hz, 4H), 4.16 (q, J=7.1 Hz, 2H), 3.92 (d, J=10.9 Hz, 1H), 3.70 (d, J=11.7 Hz, 1H), 3.31 (q, J=7.1 Hz, 4H), 2.72-2.53 (m, 2H), 2.42 (td, J=11.3, 3.1 Hz, 1H), 2.09-1.98 (m, 1H), 1.85 (dt, J=13.6, 3.6 Hz, 1H), 1.78-1.65 (m, 1H), 1.48-1.36 (m, 1H), 1.28 (t, J=7.1 Hz, 3H), 1.19 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 509.1 [M+H]+.


6-(N,N-Diethylsulfamoyl)naphthalene-2-sulfonyl chloride (12e)

Using a similar procedure as described for 12a with naphthalene-2,6-disulfonyl dichloride (11e, 200 mg, 0.62 mmol) and diethylamine (36 mg, 0.49 mmol), white solid (42 mg, 19%). 1H NMR (400 MHz, CDCl3) δ 8.68 (s, 1H), 8.52 (s, 1H), 8.28-8.13 (m, 3H), 8.02 (dd, J=8.6, 1.8 Hz, 1H), 3.36 (q, J=7.1 Hz, 4H), 1.19 (td, J=7.2, 1.0 Hz, 6H).


Ethyl (R)-1-((6-(N,N-diethylsulfamoyl)naphthalen-2-yl)sulfonyl)piperidine-3-carboxylate (DX3-99B)

Using a similar procedure as described for DX2-201 with 12e (21 mg, 0.058 mmol) and ethyl (R)-piperidine-3-carboxylate (18 mg, 0.12 mmol), white solid (4 mg, 14%). 1H NMR (300 MHz, CDCl3) δ 8.47 (s, 1H), 8.41 (s, 1H), 8.12 (dd, J=8.6, 1.9 Hz, 2H), 7.92 (ddd, J=15.0, 8.6, 1.8 Hz, 2H), 4.15 (q, J=7.1 Hz, 2H), 3.95 (d, J=8.2 Hz, 1H), 3.74 (d, J=11.6 Hz, 1H), 3.35 (q, J=7.1 Hz, 4H), 2.72-2.58 (m, 2H), 2.47 (td, J=11.4, 3.2 Hz, 1H), 2.01 (d, J=13.4 Hz, 1H), 1.89-1.78 (m, 1H), 1.78-1.61 (m, 1H), 1.49-1.33 (m, 2H), 1.27 (t, J=7.1 Hz, 3H), 1.18 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 483.0 [M+H]+.




embedded image


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylic acid (DX2-235)

To a solution of DX2-201 (230 mg, 0.53 mmol) in THF (1 mL) and H2O (1 mL) was added LiOH·H2O (186 mg, 2.66 mmol) at 0° C. and stirred at room temperature for 5 h. The mixture was then diluted with H2O, and the pH was adjusted to 3 by 1N HCl solution. It was extracted with EtOAc, and the organic layer was washed with brine, dried over anhydrous Na2SO4, filtered and concentrated to give DX2-235 as a white solid (187 mg, 87%). 1H NMR (300 MHz, MeOD) δ 8.07 (d, J=8.1 Hz, 2H), 7.99 (d, J=7.9 Hz, 2H), 3.82-3.70 (m, 1H), 3.60-3.48 (m, 1H), 3.36-3.24 (m, 4H), 2.69 (t, J=10.7 Hz, 1H), 2.65-2.47 (m, 2H), 2.05-1.91 (m, 1H), 1.89-1.76 (m, 1H), 1.71-1.57 (m, 1H), 1.56-1.38 (m, 1H), 1.15 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 405.0 [M+H]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N,N-diethylpiperidine-3-carboxamide (DX2-237)

To a solution of DX2-235 (30 mg, 0.074 mmol) and HATU (42 mg, 0.11 mmol) in DCM (1 mL) was added diethylamine (65 mg, 0.089 mmol) and DIEA (29 mg, 0.22 mmol). The mixture was stirred at room temperature overnight. The mixture was then diluted with EtOAc, washed with brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified with flash chromatography (10% MeOH in DCM) to give DX2-237 as a white solid (21 mg, 62%). 1H NMR (300 MHz, CDCl3) δ 7.97 (d, J=8.3 Hz, 2H), 7.89 (d, J=8.3 Hz, 2H), 3.87 (d, J=11.9 Hz, 2H), 3.43-3.23 (m, 8H), 2.84-2.73 (m, 1H), 2.58 (t, J=11.3 Hz, 1H), 2.34-2.22 (m, 1H), 1.91-1.66 (m, 3H), 1.56-1.46 (m, 1H), 1.30-1.06 (m, 12H). LC-MS (ESI) m/z 460.1 [M+H]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N-methylpiperidine-3-carboxamide (DX2-238)

Using a similar procedure as described for DX2-237 with DX2-235 (30 mg, 0.074 mmol) and methylamine (2.3 mg, 0.074 mmol), white solid (26 mg, 84%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.3 Hz, 2H), 7.88 (d, J=8.4 Hz, 2H), 5.79 (s, 1H), 3.72 (d, J=11.5 Hz, 1H), 3.61 (d, J=11.6 Hz, 1H), 3.30 (q, J=7.1 Hz, 4H), 2.84 (d, J=4.6 Hz, 3H), 2.76-2.66 (m, 1H), 2.57-2.37 (m, 2H), 1.89-1.78 (m, 2H), 1.76-1.59 (m, 2H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 418.1 [M+H]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N-(2-(dimethylamino)ethyl)piperidine-3-carboxamide (DX2-241)

Using a similar procedure as described for DX2-237 with DX2-235 (30 mg, 0.074 mmol) and N,N-dimethylethane-1,2-diamine (7 mg, 0.074 mmol) in DMF (1 mL), white solid (27 mg, 77%). 1H NMR (300 MHz, CDCl3) δ 8.03-7.90 (m, 4H), 3.88-3.64 (m, 2H), 3.61-3.37 (m, 3H), 3.30 (q, J=7.1 Hz, 4H), 3.08 (s, 2H), 2.79 (s, 6H), 2.67-2.48 (m, 2H), 1.96-1.84 (m, 2H), 1.76-1.48 (m, 3H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 475.1 [M+H]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N-(oxetan-3-yl)piperidine-3-carboxamide (DX2-242)

Using a similar procedure as described for DX2-237 with DX2-235 (30 mg, 0.074 mmol) and oxetan-3-amine (5.4 mg, 0.074 mmol) in DMF (1 mL), white solid (24 mg, 71%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.4 Hz, 2H), 7.89 (d, J=8.5 Hz, 2H), 6.56 (d, J=7.2 Hz, 1H), 5.09-4.87 (m, 3H), 4.54 (q, J=6.5 Hz, 2H), 3.73-3.48 (m, 1H), 3.31 (q, J=7.2 Hz, 4H), 2.84-2.70 (m, 1H), 2.56 (d, J=23.6 Hz, 2H), 1.90-1.57 (m, 4H), 1.18 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 460.1 [M+H]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N-(4-fluorophenyl)piperidine-3-carboxamide (DX2-244)

Using a similar procedure as described for DX2-237 with DX2-235 (30 mg, 0.074 mmol) and 4-fluoroaniline (8.2 mg, 0.074 mmol) in DMF (1 mL), white solid (32 mg, 86%). 1H NMR (300 MHz, CDCl3) δ 8.01 (d, J=8.5 Hz, 2H), 7.91 (d, J=8.4 Hz, 2H), 7.75 (s, 1H), 7.53 (dd, J=9.0, 4.8 Hz, 2H), 7.04 (t, J=8.7 Hz, 2H), 3.70 (dd, J=10.2, 2.9 Hz, 1H), 3.60-3.50 (m, 1H), 3.31 (q, J=7.1 Hz, 4H), 2.98-2.87 (m, 1H), 2.77-2.59 (m, 2H), 2.00-1.72 (m, 4H), 1.19 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 498.1 [M+H]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N-isopropylpiperidine-3-carboxamide (DX2-295)

Using a similar procedure as described for DX2-237 with DX2-235 (30 mg, 0.074 mmol) and propan-2-amine (5.2 mg, 0.079 mmol) in DMF (1 mL), white solid (29 mg, 88%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.0 Hz, 2H), 7.89 (d, J=7.9 Hz, 2H), 5.61 (s, 1H), 4.18-4.05 (m, 1H), 3.74-3.56 (m, 2H), 3.31 (q, J=7.3 Hz, 4H), 2.80-2.67 (m, 1H), 2.60-2.47 (m, 1H), 2.42-2.32 (m, 1H), 1.89-1.77 (m, 2H), 1.75-1.62 (m, 2H), 1.18 (t, J=6.9 Hz, 12H). LC-MS (ESI) m/z 446.0 [M+H]+.


(R)—N-Cyclopropyl-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxamide (DX2-296)

Using a similar procedure as described for DX2-237 with DX2-235 (30 mg, 0.074 mmol) and cyclopropanamine (5.0 mg, 0.089 mmol) in DMF (1 mL), white solid (17 mg, 52%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.3 Hz, 2H), 7.88 (d, J=8.2 Hz, 2H), 5.86 (s, 1H), 3.73-3.53 (m, 2H), 3.31 (q, J=7.2 Hz, 4H), 2.79-2.63 (m, 2H), 2.58-2.47 (m, 1H), 2.37 (d, J=11.2 Hz, 1H), 1.90-1.76 (m, 2H), 1.74-1.60 (m, 2H), 1.18 (t, J=7.2 Hz, 6H), 0.81 (d, J=6.7 Hz, 2H), 0.53 (t, J=5.6 Hz, 2H). LC-MS (ESI) m/z 444.1 [M+H]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N-isopropyl-N-methylpiperidine-3-carboxamide (DX2-297)

Using a similar procedure as described for DX2-237 with DX2-235 (30 mg, 0.074 mmol) and N-methylpropan-2-amine (6.5 mg, 0.089 mmol) in DMF (1 mL), white solid (25 mg, 74%). 1H NMR (400 MHz, CDCl3) δ 8.01-7.95 (m, 2H), 7.89 (d, J=8.7 Hz, 2H), 3.86 (d, J=11.5 Hz, 2H), 3.31 (q, J=7.1 Hz, 5H), 2.66-2.57 (m, 1H), 2.57-2.44 (m, 2H), 2.29 (td, J=12.0, 2.8 Hz, 1H), 1.85 (dd, J=12.6, 5.7 Hz, 2H), 1.73 (dtd, J=17.9, 8.9, 4.8 Hz, 1H), 1.46 (ddd, J=12.2, 10.3, 3.8 Hz, 1H), 1.19 (t, J=7.1 Hz, 6H), 1.06-0.94 (m, 2H), 0.94-0.81 (m, 4H), 0.63 (s, 2H). LC-MS (ESI) m/z 460.1 [M+H]+.


(R)—N,N-dicyclopropyl-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxamide (DX3-84)

Using a similar procedure as described for DX2-237 with DX2-235 (30 mg, 0.074 mmol) and dicyclopropylamine (10 mg, 0.074 mmol) in DMF (1 mL), white solid (25 mg, 74%). 1H NMR (400 MHz, CDCl3) δ 8.01-7.95 (m, 2H), 7.89 (d, J=8.7 Hz, 2H), 3.86 (d, J=11.5 Hz, 2H), 3.31 (q, J=7.1 Hz, 5H), 2.66-2.57 (m, 1H), 2.57-2.44 (m, 2H), 2.29 (td, J=12.0, 2.8 Hz, 1H), 1.85 (dd, J=12.6, 5.7 Hz, 2H), 1.73 (dtd, J=17.9, 8.9, 4.8 Hz, 1H), 1.46 (ddd, J=12.2, 10.3, 3.8 Hz, 1H), 1.19 (t, J=7.1 Hz, 6H), 1.06-0.94 (m, 2H), 0.94-0.81 (m, 4H), 0.63 (s, 2H). LC-MS (ESI) m/z 484.1 [M+H]+.


(R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)-N-methyl-N-(oxetan-3-yl)piperidine-3-carboxamide (DX3-100)

Using a similar procedure as described for DX2-237 with DX2-235 (30 mg, 0.074 mmol) and N-methyloxetan-3-amine (6.5 mg, 0.074 mmol) in DMF (1 mL), white solid (30 mg, 86%). 1H NMR (300 MHz, CDCl3) δ 8.01-7.94 (m, 2H), 7.92-7.85 (m, 2H), 5.41 (p, J=7.1 Hz, 1H), 4.89-4.79 (m, 2H), 4.66 (q, J=7.0 Hz, 2H), 3.88 (d, J=11.4 Hz, 2H), 3.30 (q, J=7.2 Hz, 4H), 3.21 (s, 3H), 2.53 (q, J=11.1 Hz, 1H), 2.28 (td, J=12.0, 3.0 Hz, 1H), 1.87 (q, J=12.5, 10.7 Hz, 2H), 1.80-1.60 (m, 2H), 1.45 (td, J=12.5, 4.0 Hz, 1H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 474.1 [M+H]+.


(R)—N-(1-Cyanocyclopropyl)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxamide (DX3-100B)

Using a similar procedure as described for DX2-237 with DX2-235 (30 mg, 0.074 mmol) and 1-aminocyclopropane-1-carbonitrile hydrochloride (8.8 mg, 0.074 mmol) in DMF (1 mL), white solid (30 mg, 86%). 1H NMR (300 MHz, CDCl3) δ 8.01 (d, J=8.4 Hz, 2H), 7.92-7.86 (m, 2H), 6.60 (s, 1H), 3.59 (dd, J=12.1, 3.7 Hz, 1H), 3.50 (d, J=11.5 Hz, 1H), 3.31 (q, J=7.2 Hz, 4H), 2.82 (dd, J=12.0, 9.0 Hz, 1H), 2.73-2.61 (m, 1H), 2.48 (s, 1H), 1.89-1.77 (m, 2H), 1.77-1.65 (m, 2H), 1.59 (dd, J=8.0, 5.3 Hz, 3H), 1.31-1.23 (m, 3H), 1.19 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 469.1 [M+H]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N-(oxetan-3-ylmethyl)piperidine-3-carboxamide (DX3-104)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and oxetan-3-ylmethanamine (4.4 mg, 0.05 mmol) in DMF (1 mL), white solid (19 mg, 80%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.7 Hz, 2H), 7.89 (d, J=8.7 Hz, 2H), 6.10 (t, J=6.5 Hz, 1H), 4.83 (ddd, J=7.7, 6.3, 2.3 Hz, 2H), 4.42 (td, J=6.1, 2.3 Hz, 2H), 3.59 (td, J=6.4, 6.0, 2.6 Hz, 3H), 3.50 (d, J=11.3 Hz, 1H), 3.31 (q, J=7.2 Hz, 4H), 3.19 (p, J=6.4 Hz, 1H), 2.84 (dd, J=11.9, 9.1 Hz, 1H), 2.63 (d, J=10.6 Hz, 1H), 2.48 (dt, J=9.1, 4.8 Hz, 1H), 1.89-1.65 (m, 4H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 474.1 [M+H]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N-(3-methyloxetan-3-yl)piperidine-3-carboxamide (DX3-104B)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and 3-methyloxetan-3-amine (4.4 mg, 0.05 mmol) in DMF (1 mL), white solid (22 mg, 93%). 1H NMR (300 MHz, CDCl3) δ 8.00 (d, J=8.6 Hz, 2H), 7.89 (d, J=8.7 Hz, 2H), 6.22 (s, 1H), 4.74 (dd, J=6.5, 2.9 Hz, 2H), 4.50 (d, J=6.5 Hz, 2H), 3.62 (dd, J=11.9, 3.8 Hz, 1H), 3.51 (d, J=11.3 Hz, 1H), 3.31 (q, J=7.2 Hz, 4H), 2.82 (dd, J=11.7, 9.3 Hz, 1H), 2.65 (t, J=9.9 Hz, 1H), 2.46 (s, 1H), 1.89-1.68 (m, 4H), 1.66 (s, 3H), 1.18 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 474.0 [M+H]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N-(tetrahydro-2H-pyran-4-yl)piperidine-3-carboxamide (DX3-106B)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and tetrahydro-2H-pyran-4-amine (6.9 mg, 0.05 mmol) in DMF (1 mL), white solid (21 mg, 86%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.7 Hz, 2H), 7.89 (d, J=8.7 Hz, 2H), 5.70 (d, J=8.0 Hz, 1H), 3.98 (dt, J=11.2, 3.5 Hz, 3H), 3.66 (d, J=11.0 Hz, 1H), 3.61-3.42 (m, 3H), 3.31 (q, J=7.2 Hz, 4H), 2.84-2.73 (m, 1H), 2.60 (d, J=10.3 Hz, 1H), 2.49-2.36 (m, 1H), 1.97-1.63 (m, 6H), 1.51 (ddd, J=12.9, 6.5, 4.4 Hz, 2H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 488.2 [M+H]+.


tert-Butyl (R)-3-(1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxamido)azetidine-1-carboxylate (DX3-107)

Using a similar procedure as described for DX2-237 with DX2-235 (40 mg, 0.10 mmol) and tert-butyl 3-aminoazetidine-1-carboxylate (17.2 mg, 0.10 mmol) in DMF (2 mL), white solid (54 mg, 96%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.7 Hz, 2H), 7.89 (d, J=8.7 Hz, 2H), 6.40 (d, J=7.1 Hz, 1H), 4.69-4.54 (m, 1H), 4.27 (td, J=8.5, 7.9, 4.4 Hz, 2H), 3.77 (dt, J=9.3, 5.8 Hz, 2H), 3.67 (d, J=10.4 Hz, 1H), 3.57 (d, J=11.7 Hz, 1H), 3.31 (q, J=7.1 Hz, 4H), 2.83-2.69 (m, 1H), 2.65-2.42 (m, 2H), 1.84 (d, J=12.1 Hz, 2H), 1.68 (t, J=9.2 Hz, 2H), 1.46 (s, 9H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 559.2 [M+H]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N—((R)-tetrahydrofuran-3-yl)piperidine-3-carboxamide (DX3-107B)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and (R)-tetrahydrofuran-3-amine hydrochloride (6.2 mg, 0.05 mmol) in DMF (1 mL), white solid (15 mg, 63%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.5 Hz, 2H), 7.89 (d, J=8.6 Hz, 2H), 6.01 (d, J=7.4 Hz, 1H), 4.50 (dt, J=7.6, 5.8 Hz, 1H), 3.96 (q, J=7.6 Hz, 1H), 3.83 (qd, J=9.1, 5.5 Hz, 2H), 3.73-3.54 (m, 3H), 3.31 (q, J=7.1 Hz, 4H), 2.74 (dd, J=11.8, 9.6 Hz, 1H), 2.55 (t, J=10.3 Hz, 1H), 2.44 (dd, J=10.0, 6.2 Hz, 1H), 2.37-2.20 (m, 1H), 1.91-1.63 (m, 5H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 474.1 [M+H]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N—((S)-tetrahydrofuran-3-yl)piperidine-3-carboxamide (DX3-108)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and (S)-tetrahydrofuran-3-amine hydrochloride (6.2 mg, 0.05 mmol) in DMF (1 mL), white solid (23 mg, 98%). 1H NMR (300 MHz, CDCl3) δ 8.03-7.95 (m, 2H), 7.89 (d, J=8.7 Hz, 2H), 6.01 (d, J=7.4 Hz, 1H), 4.57-4.45 (m, 1H), 3.99 (q, J=7.9, 7.5 Hz, 1H), 3.83 (ddd, J=9.6, 5.5, 2.4 Hz, 2H), 3.66 (dd, J=9.6, 2.8 Hz, 2H), 3.57 (d, J=11.7 Hz, 1H), 3.31 (q, J=7.2 Hz, 4H), 2.84-2.71 (m, 1H), 2.57 (t, J=10.0 Hz, 1H), 2.43 (s, 1H), 2.37-2.23 (m, 1H), 1.90-1.63 (m, 5H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 474.0 [M+H]+.


tert-Butyl (R)-3-((R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxamido)pyrrolidine-1-carboxylate (DX3-118)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and tert-butyl (R)-3-aminopyrrolidine-1-carboxylate (9.3 mg, 0.05 mmol) in DMF (1 mL), white solid (19 mg, 66%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.8 Hz, 2H), 7.89 (d, J=8.6 Hz, 2H), 5.98 (s, 1H), 4.44 (q, J=5.8 Hz, 1H), 3.65 (dd, J=11.6, 5.3 Hz, 2H), 3.60-3.52 (m, 1H), 3.52-3.40 (m, 2H), 3.31 (q, J=7.2 Hz, 4H), 3.24-3.13 (m, 1H), 2.81-2.70 (m, 1H), 2.67-2.53 (m, 1H), 2.49-2.39 (m, 1H), 2.16 (dq, J=13.4, 6.9 Hz, 1H), 1.91-1.75 (m, 3H), 1.75-1.62 (m, 2H), 1.50 (s, 9H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 595.3 [M+Na]+.


tert-Butyl (R)-1-(1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carbonyl)-1,6-diazaspiro[3.3]heptane-6-carboxylate (DX3-118B)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and tert-butyl 1,6-diazaspiro[3.3]heptane-6-carboxylate hemioxylate (12 mg, 0.05 mmol) in DMF (1 mL), white solid (22 mg, 76%). 1H NMR (300 MHz, CDCl3) δ 7.98 (d, J=8.5 Hz, 2H), 7.88 (d, J=8.6 Hz, 2H), 4.64 (t, J=9.5 Hz, 2H), 4.22-3.99 (m, 2H), 3.97-3.78 (m, 4H), 3.30 (q, J=7.1 Hz, 4H), 2.60-2.35 (m, 4H), 2.25 (t, J=11.5 Hz, 1H), 1.85 (d, J=12.5 Hz, 2H), 1.75-1.60 (m, 2H), 1.44 (s, 9H), 1.19 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 607.2 [M+Na]+.


tert-Butyl (S)-3-((R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxamido)pyrrolidine-1-carboxylate (DX3-119)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and tert-butyl (S)-3-aminopyrrolidine-1-carboxylate (9.3 mg, 0.05 mmol) in DMF (1 mL), white solid (18 mg, 62%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.3 Hz, 2H), 7.88 (d, J=8.1 Hz, 2H), 6.03 (d, J=7.2 Hz, 1H), 4.45 (s, 1H), 3.71-3.40 (m, 5H), 3.31 (q, J=7.1 Hz, 4H), 3.17 (s, 1H), 2.87-2.75 (m, 1H), 2.68-2.54 (m, 1H), 2.52-2.39 (m, 1H), 2.19 (dq, J=12.8, 6.9, 6.2 Hz, 1H), 1.95-1.66 (m, 5H), 1.49 (s, 9H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 595.1 [M+Na]+.


tert-Butyl (R)-6-(1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carbonyl)-2,6-diazaspiro[3.3]heptane-2-carboxylate (DX3-120)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and tert-butyl 2,6-diazaspiro[3.3]heptane-2-carboxylate (10 mg, 0.05 mmol) in DMF (1 mL), white solid (28 mg, 96%). 1H NMR (300 MHz, CDCl3) δ 7.98 (d, J=8.5 Hz, 2H), 7.87 (d, J=8.5 Hz, 2H), 4.42-4.24 (m, 2H), 4.10 (d, J=7.4 Hz, 6H), 3.86 (dt, J=8.7, 5.0 Hz, 2H), 3.30 (q, J=7.1 Hz, 4H), 2.58-2.42 (m, 2H), 2.32-2.20 (m, 1H), 1.91-1.79 (m, 2H), 1.75-1.60 (m, 2H), 1.46 (s, 9H), 1.18 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 585.1 [M+H]+.


tert-Butyl (R)-4-(1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carbonyl)piperazine-1-carboxylate (DX3-121)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and tert-butyl piperazine-1-carboxylate (9.3 mg, 0.05 mmol) in DMF (1 mL), white solid (26 mg, 91%). 1H NMR (300 MHz, CDCl3) δ 7.98 (d, J=8.7 Hz, 2H), 7.89 (d, J=8.7 Hz, 2H), 3.88 (d, J=11.7 Hz, 2H), 3.61-3.40 (m, 8H), 3.30 (q, J=7.2 Hz, 4H), 2.92-2.80 (m, 1H), 2.58 (t, J=11.4 Hz, 1H), 2.35-2.22 (m, 1H), 1.94-1.79 (m, 2H), 1.78-1.68 (m, 1H), 1.63-1.53 (m, 1H), 1.50 (s, 9H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 595.2 [M+H]+.


(R)—N,N-Diethyl-4-((3-(morpholine-4-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-121B)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and morpholine (4.4 mg, 0.05 mmol) in DMF (1 mL), white solid (23 mg, 97%). 1H NMR (300 MHz, CDCl3) δ 7.98 (d, J=8.7 Hz, 2H), 7.89 (d, J=8.7 Hz, 2H), 3.88 (d, J=11.6 Hz, 2H), 3.81-3.49 (m, 8H), 3.30 (q, J=7.1 Hz, 4H), 2.91-2.78 (m, 1H), 2.64-2.53 (m, 1H), 2.36-2.24 (m, 1H), 1.92-1.81 (m, 2H), 1.79-1.64 (m, 1H), 1.55-1.39 (m, 1H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 474.1 [M+H]+.


(R)—N,N-Diethyl-4-((3-(piperidine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-122)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and piperidine (4.3 mg, 0.05 mmol) in DMF (1 mL), white solid (21 mg, 89%). 1H NMR (300 MHz, CDCl3) δ 7.97 (d, J=7.8 Hz, 2H), 7.88 (d, J=8.0 Hz, 2H), 3.87 (d, J=11.5 Hz, 2H), 3.49 (dt, J=24.8, 5.4 Hz, 5H), 3.29 (q, J=7.2 Hz, 4H), 3.16 (d, J=9.1 Hz, 1H), 2.94-2.78 (m, 1H), 2.56 (t, J=11.3 Hz, 1H), 2.34-2.19 (m, 2H), 1.94-1.78 (m, 2H), 1.77-1.38 (m, 5H), 1.17 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 472.1 [M+H]+.


tert-Butyl (R)-4-(1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxamido)piperidine-1-carboxylate (DX3-124)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and tert-butyl 4-aminopiperidine-1-carboxylate (10 mg, 0.05 mmol) in DMF (1 mL), white solid (28 mg, 95%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.7 Hz, 2H), 7.88 (d, J=8.7 Hz, 2H), 5.77 (d, J=7.9 Hz, 1H), 4.16-4.00 (m, 2H), 3.91 (ddd, J=11.1, 7.5, 3.9 Hz, 1H), 3.68-3.58 (m, 1H), 3.53 (d, J=11.2 Hz, 1H), 3.31 (q, J=7.1 Hz, 4H), 2.97-2.73 (m, 3H), 2.67-2.54 (m, 1H), 2.50-2.35 (m, 1H), 1.99-1.63 (m, 6H), 1.48 (s, 9H), 1.42-1.26 (m, 2H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 609.2 [M+Na]+.


tert-Butyl (1S,4S)-5-((R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carbonyl)-2,5-diazabicyclo[2.2.1]heptane-2-carboxylate (DX3-126B)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and tert-butyl (1S,4S)-2,5-diazabicyclo[2.2.1]heptane-2-carboxylate (10 mg, 0.05 mmol) in DMF (1 mL), white solid (26 mg, 90%). 1H NMR (300 MHz, CDCl3) δ 7.98 (dd, J=8.6, 1.6 Hz, 2H), 7.93-7.83 (m, 2H), 4.93-4.46 (m, 2H), 3.99-3.79 (m, 2H), 3.64-3.35 (m, 4H), 3.30 (q, J=7.2, 6.6 Hz, 4H), 2.75-2.46 (m, 2H), 2.27 (t, J=11.6 Hz, 1H), 2.02-1.64 (m, 5H), 1.50 (s, 9H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 585.2 [M+H]+.


tert-Butyl (R)-2-(1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carbonyl)-2,6-diazaspiro[3.4]octane-6-carboxylate (DX3-127)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and tert-butyl 2,6-diazaspiro[3.4]octane-6-carboxylate (11 mg, 0.05 mmol) in DMF (1 mL), white solid (27 mg, 90%). 1H NMR (300 MHz, CDCl3) δ 7.97 (d, J=8.6 Hz, 2H), 7.87 (d, J=8.8 Hz, 2H), 4.25-4.01 (m, 2H), 3.89 (dt, J=15.6, 9.5 Hz, 4H), 3.62-3.37 (m, 4H), 3.30 (q, J=7.2 Hz, 4H), 2.60-2.40 (m, 2H), 2.26 (t, J=11.6 Hz, 1H), 2.09 (d, J=8.7 Hz, 2H), 1.84 (d, J=12.3 Hz, 2H), 1.68 (d, J=13.2 Hz, 2H), 1.48 (s, 9H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 621.2 [M+Na]+.


tert-Butyl 5-((R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carbonyl)hexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate (DX3-127B)

Using a similar procedure as described for DX2-237 with DX2-235 (40 mg, 0.10 mmol) and tert-butyl hexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate (22 mg, 0.10 mmol) in DMF (1 mL), white solid (44 mg, 73%). 1H NMR (300 MHz, CDCl3) δ 7.98 (dd, J=8.6, 1.6 Hz, 2H), 7.93-7.84 (m, 2H), 3.96-3.56 (m, 6H), 3.53-3.36 (m, 2H), 3.30 (q, J=7.2 Hz, 6H), 3.06-2.95 (m, 1H), 2.96-2.84 (m, 1H), 2.69 (t, J=11.7 Hz, 1H), 2.54 (t, J=11.3 Hz, 1H), 2.28 (t, J=11.8 Hz, 1H), 1.88 (t, J=12.5 Hz, 2H), 1.82-1.62 (m, 2H), 1.49 (d, J=8.6 Hz, 9H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 621.2 [M+Na]+.


tert-Butyl (1R,4R)-5-((R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carbonyl)-2,5-diazabicyclo[2.2.1]heptane-2-carboxylate (DX3-128B)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and tert-butyl (1R,4R)-2,5-diazabicyclo[2.2.1]heptane-2-carboxylate (10 mg, 0.05 mmol) in DMF (1 mL), white solid (24 mg, 83%). 1H NMR (300 MHz, CDCl3) δ 7.97 (dd, J=8.5, 1.8 Hz, 2H), 7.88 (dd, J=8.5, 2.7 Hz, 2H), 4.91-4.43 (m, 2H), 3.96-3.78 (m, 2H), 3.66-3.34 (m, 4H), 3.29 (qd, J=7.2, 1.7 Hz, 4H), 2.76-2.44 (m, 2H), 2.36-2.19 (m, 1H), 2.08-1.64 (m, 5H), 1.51 (s, 9H), 1.17 (t, J=7.3 Hz, 6H). LC-MS (ESI) m/z 585.2 [M+H]+.


tert-butyl (R)-4-((R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carbonyl)-3-methylpiperazine-1-carboxylate (DX3-139B)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and tert-butyl (R)-3-methylpiperazine-1-carboxylate (10 mg, 0.05 mmol) in DMF (1 mL), white solid (29 mg, 99%). 1H NMR (300 MHz, CDCl3) δ 7.98 (d, J=8.2 Hz, 2H), 7.89 (d, J=8.3 Hz, 2H), 4.80-4.66 (m, 0.5H), 4.34 (d, J=12.6 Hz, 0.5H), 4.24-3.80 (m, 5H), 3.63 (d, J=12.8 Hz, 1H), 3.30 (q, J=7.2 Hz, 4H), 3.07-2.73 (m, 4H), 2.68-2.55 (m, 1H), 2.28 (t, J=11.7 Hz, 1H), 1.93-1.81 (m, 2H), 1.79-1.69 (m, 1H), 1.49 (s, 9H), 1.33 (d, J=6.6 Hz, 1.5H), 1.18 (t, J=7.1 Hz, 6H), 1.13 (d, J=7.0 Hz, 1.5H). LC-MS (ESI) m/z 587.2 [M+H]+.


tert-butyl (S)-4-((R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carbonyl)-3-methylpiperazine-1-carboxylate (DX3-141)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and tert-butyl (S)-3-methylpiperazine-1-carboxylate (10 mg, 0.05 mmol) in DMF (1 mL), white solid (26 mg, 89%). 1H NMR (300 MHz, CDCl3) δ 7.98 (d, J=8.3 Hz, 2H), 7.89 (d, J=8.2 Hz, 2H), 4.81-4.67 (m, 0.5H), 4.41-4.30 (m, 0.5H), 4.25-3.78 (m, 5H), 3.59 (d, J=11.8 Hz, 0.5H), 3.43 (dd, J=10.6, 3.9 Hz, 0.5H), 3.30 (q, J=7.1 Hz, 4H), 3.16-2.73 (m, 4H), 2.57 (t, J=11.3 Hz, 1H), 2.28 (t, J=11.6 Hz, 1H), 1.86 (d, J=12.6 Hz, 2H), 1.76-1.69 (m, 1H), 1.50 (s, 9H), 1.30 (d, J=7.1 Hz, 1.5H), 1.18 (t, J=7.2 Hz, 7.5H). LC-MS (ESI) m/z 587.2 [M+H]+.


tert-Butyl (R)-4-((R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carbonyl)-2-methylpiperazine-1-carboxylate (DX3-141B)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and tert-butyl (R)-2-methylpiperazine-1-carboxylate (10 mg, 0.05 mmol) in DMF (1 mL), white solid (26 mg, 89%). 1H NMR (300 MHz, CDCl3) δ 7.98 (d, J=8.0 Hz, 2H), 7.89 (t, J=7.5 Hz, 2H), 4.49-4.23 (m, 2H), 4.04-3.76 (m, 4H), 3.66 (d, J=13.3 Hz, 0.5H), 3.41 (d, J=14.4 Hz, 0.5H), 3.30 (q, J=7.1 Hz, 4H), 3.20-3.10 (m, 1H), 3.09-2.68 (m, 3H), 2.69-2.44 (m, 1H), 2.29 (t, J=11.9 Hz, 1H), 1.95-1.81 (m, 2H), 1.79-1.67 (m, 1H), 1.50 (s, 9H), 1.25-1.08 (m, 9H). LC-MS (ESI) m/z 587.2 [M+H]+.


tert-Butyl (S)-4-((R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carbonyl)-2-methylpiperazine-1-carboxylate (DX3-142)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and tert-butyl (S)-2-methylpiperazine-1-carboxylate (10 mg, 0.05 mmol) in DMF (1 mL), white solid (27 mg, 92%). 1H NMR (300 MHz, CDCl3) δ 7.98 (d, J=8.2 Hz, 2H), 7.88 (d, J=8.2 Hz, 2H), 4.51-4.24 (m, 2H), 4.04-3.76 (m, 4H), 3.64 (d, J=13.1 Hz, 0.5H), 3.43-3.32 (m, 0.5H), 3.30 (q, J=7.2 Hz, 4H), 3.27-3.17 (m, 0.5H), 3.15-2.96 (m, 1.5H), 2.94-2.53 (m, 4H), 2.28 (t, J=11.8 Hz, 1H), 1.96-1.79 (m, 2H), 1.77-1.67 (m, 1H), 1.49 (s, 9H), 1.24 (d, J=6.8 Hz, 1.5H), 1.18 (t, J=7.1 Hz, 6H), 1.06 (d, J=6.7 Hz, 1.5H). LC-MS (ESI) m/z 587.2 [M+H]+.


tert-Butyl 8-((R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carbonyl)-3,8-diazabicyclo[3.2.1]octane-3-carboxylate (DX3-142B)

Using a similar procedure as described for DX2-237 with DX2-235 (20 mg, 0.05 mmol) and tert-butyl (1R,5S)-3,8-diazabicyclo[3.2.1]octane-3-carboxylate (11 mg, 0.05 mmol) in DMF (1 mL), white solid (29 mg, 97%). 1H NMR (300 MHz, CDCl3) δ 7.98 (dd, J=8.5, 2.0 Hz, 2H), 7.89 (dd, J=8.3, 5.2 Hz, 2H), 4.75-4.59 (m, 1H), 4.32-4.13 (m, 1H), 4.10-3.73 (m, 4H), 3.30 (q, J=7.1 Hz, 4H), 3.14-2.88 (m, 2H), 2.81-2.68 (m, 1H), 2.68-2.48 (m, 1H), 2.36-2.21 (m, 1H), 2.07-1.70 (m, 8H), 1.48 (d, J=4.4 Hz, 9H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 599.2 [M+H]+.


(R)-4-((3-(6-Oxa-2-azaspiro[3.4]octane-2-carbonyl)piperidin-1-yl)sulfonyl)-N,N-diethylbenzenesulfonamide (DX3-149)

Using a similar procedure as described for DX2-237 with DX2-235 (15 mg, 0.037 mmol) and 6-oxa-2-azaspiro[3.4]octane hemioxalate (5.9 mg, 0.037 mmol) in DMF (1 mL), white solid (18 mg, 97%). 1H NMR (300 MHz, CDCl3) δ 7.98 (d, J=8.5 Hz, 2H), 7.88 (d, J=8.4 Hz, 2H), 4.28-4.09 (m, 2H), 4.02-3.80 (m, 8H), 3.30 (q, J=7.1 Hz, 4H), 2.59-2.44 (m, 2H), 2.34-2.12 (m, 3H), 1.85 (d, J=12.8 Hz, 2H), 1.78-1.68 (m, 1H), 1.54-1.41 (m, 1H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 500.1 [M+H]+.


(R)-4-((3-(2-Oxa-6-azaspiro[3.3]heptane-6-carbonyl)piperidin-1-yl)sulfonyl)-N,N-diethylbenzenesulfonamide (DX3-149B)

Using a similar procedure as described for DX2-237 with DX2-235 (15 mg, 0.037 mmol) and 2-oxa-6-azaspiro[3.3]heptane hemioxalate (5.3 mg, 0.037 mmol) in DMF (1 mL), white solid (11 mg, 61%). 1H NMR (300 MHz, CDCl3) δ 7.97 (d, J=8.4 Hz, 2H), 7.87 (d, J=8.5 Hz, 2H), 4.90-4.77 (m, 4H), 4.48-4.30 (m, 2H), 4.15 (s, 2H), 3.91-3.75 (m, 2H), 3.30 (q, J=7.2 Hz, 4H), 2.55-2.42 (m, 2H), 2.32-2.20 (m, 1H), 1.91-1.70 (m, 3H), 1.50-1.39 (m, 1H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 486.1 [M+H]+.


(R)-4-((3-(4,4-Difluoropiperidine-1-carbonyl)piperidin-1-yl)sulfonyl)-N,N-diethylbenzenesulfonamide (DX3-184B)

Using a similar procedure as described for DX2-237 with DX2-235 (15 mg, 0.037 mmol) and 4,4-difluoropiperidine (4.5 mg, 0.037 mmol) in DMF (1 mL), white solid (11 mg, 59%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.2 Hz, 2H), 7.89 (d, J=8.2 Hz, 2H), 3.89 (d, J=11.7 Hz, 2H), 3.79-3.58 (m, 4H), 3.30 (q, J=7.1 Hz, 4H), 2.88 (t, J=12.3 Hz, 1H), 2.58 (t, J=11.4 Hz, 1H), 2.30 (t, J=11.7 Hz, 1H), 2.14-1.94 (m, 4H), 1.93-1.82 (m, 2H), 1.80-1.67 (m, 1H), 1.56-1.42 (m, 1H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 508.2 [M+H]+.


(R)-4-((3-(1,1-Dioxidothiomorpholine-4-carbonyl)piperidin-1-yl)sulfonyl)-N,N-diethylbenzenesulfonamide (DX3-185)

Using a similar procedure as described for DX2-237 with DX2-235 (15 mg, 0.037 mmol) and thiomorpholine 1,1-dioxide (5.0 mg, 0.037 mmol) in DMF (1 mL), white solid (14 mg, 73%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.2 Hz, 2H), 7.89 (d, J=8.2 Hz, 2H), 4.26-3.98 (m, 4H), 3.88 (d, J=11.8 Hz, 2H), 3.31 (q, J=7.1 Hz, 4H), 3.18-3.01 (m, 4H), 2.89 (t, J=11.5 Hz, 1H), 2.59 (t, J=11.4 Hz, 1H), 2.33 (t, J=11.8 Hz, 1H), 1.89 (d, J=12.6 Hz, 2H), 1.81-1.68 (m, 1H), 1.51 (dd, J=14.3, 5.2 Hz, 1H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 544.1 [M+Na]+.


(R)—N,N-Diethyl-4-((3-(4-methoxypiperidine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-185B)

Using a similar procedure as described for DX2-237 with DX2-235 (15 mg, 0.037 mmol) and 4-methoxypiperidine (4.3 mg, 0.037 mmol) in DMF (1 mL), white solid (13 mg, 70%). 1H NMR (300 MHz, CDCl3) δ 7.98 (d, J=8.2 Hz, 2H), 7.89 (d, J=8.2 Hz, 2H), 3.95-3.77 (m, 3H), 3.77-3.65 (m, 1H), 3.52-3.43 (m, 1H), 3.42-3.25 (m, 9H), 2.87 (t, J=11.4 Hz, 1H), 2.57 (dt, J=15.4, 7.7 Hz, 1H), 2.28 (t, J=11.8 Hz, 1H), 1.96-1.79 (m, 4H), 1.76-1.60 (m, 3H), 1.52-1.39 (m, 1H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 502.3 [M+H]+.


(R)—N,N-Diethyl-4-((3-(4-hydroxypiperidine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-186)

Using a similar procedure as described for DX2-237 with DX2-235 (15 mg, 0.037 mmol) and 4-hydroxypiperidine (3.7 mg, 0.037 mmol) in DMF (1 mL), white solid (15 mg, 83%). 1H NMR (300 MHz, CDCl3) δ 7.98 (d, J=8.2 Hz, 2H), 7.89 (d, J=8.2 Hz, 2H), 4.09-3.95 (m, 2H), 3.93-3.73 (m, 3H), 3.29 (p, J=7.2 Hz, 6H), 2.95-2.81 (m, 1H), 2.57 (t, J=11.3 Hz, 1H), 2.29 (t, J=11.8 Hz, 1H), 2.05-1.66 (m, 5H), 1.64-1.38 (m, 3H), 1.18 (t, J=7.1 Hz, 6H).


(R)—N,N-Diethyl-4-((3-(4-(methoxymethyl)piperidine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-186B)

Using a similar procedure as described for DX2-237 with DX2-235 (15 mg, 0.037 mmol) and 4-(methoxymethyl)piperidine (4.8 mg, 0.037 mmol) in DMF (1 mL), white solid (16 mg, 84%). 1H NMR (300 MHz, CDCl3) δ 7.97 (d, J=8.0 Hz, 2H), 7.89 (d, J=8.2 Hz, 2H), 4.59 (d, J=13.2 Hz, 1H), 3.97-3.81 (m, 3H), 3.41-3.19 (m, 10H), 3.07 (q, J=13.2 Hz, 1H), 2.93-2.77 (m, 1H), 2.66-2.48 (m, 2H), 2.34-2.21 (m, 1H), 1.94-1.68 (m, 7H), 1.52-1.36 (m, 1H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 516.3 [M+H]+.


N,N-Diethyl-4-(((R)-3-((R)-3-methoxypyrrolidine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-187B)

Using a similar procedure as described for DX2-237 with DX2-235 (15 mg, 0.037 mmol) and (R)-3-methoxypyrrolidine hydrochloride (5.1 mg, 0.037 mmol) in DMF (1 mL), white solid (14 mg, 78%). 1H NMR (300 MHz, CDCl3) δ 7.97 (d, J=8.2 Hz, 2H), 7.88 (d, J=8.3 Hz, 2H), 4.10-3.82 (m, 3H), 3.71-3.41 (m, 4H), 3.39-3.22 (m, 7H), 2.80-2.64 (m, 1H), 2.53 (t, J=11.3 Hz, 1H), 2.34-2.00 (m, 1H), 1.98-1.80 (m, 2H), 1.77-1.66 (m, 1H), 1.56-1.42 (m, 1H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 488.2 [M+H]+.


N,N-Diethyl-4-(((R)-3-((S)-3-methoxypyrrolidine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-188)

Using a similar procedure as described for DX2-237 with DX2-235 (15 mg, 0.037 mmol) and (S)-3-methoxypyrrolidine hydrochloride (5.1 mg, 0.037 mmol) in DMF (1 mL), white solid (14 mg, 78%). 1H NMR (300 MHz, CDCl3) δ 7.97 (d, J=8.2 Hz, 2H), 7.88 (d, J=8.3 Hz, 2H), 4.11-3.81 (m, 3H), 3.74-3.39 (m, 4H), 3.38-3.22 (m, 7H), 2.78-2.62 (m, 1H), 2.53 (td, J=11.3, 4.4 Hz, 1H), 2.35-2.13 (m, 1H), 2.08-1.78 (m, 4H), 1.74-1.57 (m, 1H), 1.56-1.39 (m, 1H), 1.17 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 488.2 [M+H]+.


(R)—N,N-Diethyl-4-((3-(4-(3-isopropyl-1,2,4-oxadiazol-5-yl)piperidine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-193)

Using a similar procedure as described for DX2-237 with DX2-235 (15 mg, 0.037 mmol) and 3-isopropyl-5-(piperidin-4-yl)-1,2,4-oxadiazole (7.2 mg, 0.037 mmol) in DMF (1 mL), white solid (12 mg, 55%). 1H NMR (300 MHz, CDCl3) δ 7.98 (d, J=8.2 Hz, 2H), 7.89 (d, J=8.2 Hz, 2H), 4.51-4.38 (m, 1H), 4.01-3.84 (m, 3H), 3.40-3.17 (m, 6H), 3.17-3.04 (m, 1H), 3.02-2.81 (m, 2H), 2.58 (t, J=11.4 Hz, 1H), 2.35-2.05 (m, 4H), 1.97-1.74 (m, 5H), 1.36 (t, J=6.1 Hz, 6H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 582.4 [M+H]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N-(cis-4-methoxycyclohexyl)piperidine-3-carboxamide (DX3-193B)

Using a similar procedure as described for DX2-237 with DX2-235 (15 mg, 0.037 mmol) and cis-4-methoxycyclohexan-1-amine (6.1 mg, 0.037 mmol) in DMF (1 mL), white solid (12 mg, 63%). 1H NMR (300 MHz, CDCl3) δ 7.98 (d, J=8.2 Hz, 2H), 7.89 (d, J=8.2 Hz, 2H), 5.60 (d, J=8.0 Hz, 1H), 3.89-3.69 (m, 2H), 3.63 (d, J=11.5 Hz, 1H), 3.44-3.34 (m, 1H), 3.35-3.25 (m, 7H), 2.68 (t, J=10.8 Hz, 1H), 2.55-2.34 (m, 2H), 1.95-1.77 (m, 4H), 1.77-1.61 (m, 4H), 1.59-1.48 (m, 4H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 538.2 [M+Na]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N-(trans-4-methoxycyclohexyl)piperidine-3-carboxamide (DX3-194)

Using a similar procedure as described for DX2-237 with DX2-235 (15 mg, 0.037 mmol) and trans-4-methoxycyclohexan-1-amine (6.1 mg, 0.037 mmol) in DMF (1 mL), white solid (10 mg, 53%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.2 Hz, 2H), 7.88 (d, J=8.2 Hz, 2H), 5.62 (d, J=7.8 Hz, 1H), 3.84-3.70 (m, 1H), 3.67-3.47 (m, 2H), 3.40-3.26 (m, 7H), 3.23-3.11 (m, 1H), 2.79 (t, J=10.6 Hz, 1H), 2.66-2.55 (m, 1H), 2.47-2.33 (m, 1H), 2.13-1.96 (m, 4H), 1.85-1.63 (m, 4H), 1.44-1.23 (m, 4H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 516.3 [M+H]+.


(R)-4-((3-(4-(Difluoromethyl)piperidine-1-carbonyl)piperidin-1-yl)sulfonyl)-N,N-diethylbenzenesulfonamide (DX3-198)

Using a similar procedure as described for DX2-237 with DX2-235 (15 mg, 0.037 mmol) and 4-(difluoromethyl)piperidine (6.4 mg, 0.037 mmol) in DMF (1 mL), white solid (22 mg, 63%). 1H NMR (300 MHz, CDCl3) δ 7.98 (d, J=8.2 Hz, 2H), 7.89 (d, J=8.2 Hz, 2H), 5.64 (t, J=56.5 Hz, 2H), 4.69 (d, J=13.4 Hz, 1H), 3.99 (d, J=13.7 Hz, 1H), 3.88 (d, J=11.6 Hz, 2H), 3.30 (q, J=7.1 Hz, 4H), 3.10 (q, J=13.5 Hz, 1H), 2.85 (d, J=12.0 Hz, 1H), 2.58 (q, J=12.2 Hz, 2H), 2.29 (t, J=11.7 Hz, 1H), 2.12-1.70 (m, 7H), 1.54-1.31 (m, 4H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 522.2 [M+H]+.


(R)—N,N-Diethyl-4-((3-(4-(trifluoromethyl)piperidine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-198B)

Using a similar procedure as described for DX2-237 with DX2-235 (15 mg, 0.037 mmol) and 4-(trifluoromethyl)piperidine (5.7 mg, 0.037 mmol) in DMF (1 mL), white solid (10 mg, 50%). 1H NMR (300 MHz, CDCl3) δ 7.98 (d, J=8.2 Hz, 2H), 7.89 (d, J=8.2 Hz, 2H), 4.71 (d, J=13.6 Hz, 1H), 4.02 (d, J=13.7 Hz, 1H), 3.88 (d, J=11.6 Hz, 2H), 3.30 (q, J=7.1 Hz, 2H), 3.17-3.00 (m, 1H), 2.86 (t, J=11.8 Hz, 1H), 2.56 (t, J=12.3 Hz, 2H), 2.29 (t, J=11.6 Hz, 2H), 2.09-1.83 (m, 4H), 1.74 (d, J=12.6 Hz, 1H), 1.53-1.41 (m, 2H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 540.2 [M+H]+.


1-Methylpiperidin-4-yl (R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-260)

To a solution of DX2-235 (40 mg, 0.10 mmol) and EDCI (22 mg, 0.12 mmol) in DCM (2 mL) was added 1-methylpiperidin-4-ol (14 mg, 0.12 mmol) followed by DMAP (6 mg, 0.05 mmol).


The mixture was stirred at room temperature overnight. The mixture was then diluted with EtOAc, washed with brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified with flash chromatography (30% EtOAc in hexane) to give DX2-260 as a white solid (43 mg, 71%). 1H NMR (300 MHz, CDCl3) δ 8.03 (d, J=8.4 Hz, 2H), 7.88 (d, J=8.5 Hz, 2H), 4.16-4.00 (m, 1H), 3.60-3.39 (m, 4H), 3.32 (q, J=7.1 Hz, 5H), 2.88 (d, J=3.3 Hz, 3H), 2.80-2.76 (m, 1H), 2.68-2.61 (m, 2H), 2.45-2.35 (m, 2H), 2.27 (d, J=9.1 Hz, 2H), 2.08-1.95 (m, 1H), 1.54-1.38 (m, 2H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 502.2 [M+H]+.


Isopropyl (R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-261)

Using a similar procedure as described for DX2-260 with DX2-235 (35 mg, 0.09 mmol) and propan-2-ol (6.2 mg, 0.10 mmol), white solid (30 mg, 75%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.5 Hz, 2H), 7.90 (d, J=8.4 Hz, 2H), 5.02 (dt, J=12.5, 6.3 Hz, 1H), 3.83 (d, J=8.4 Hz, 1H), 3.68-3.57 (m, 1H), 3.31 (q, J=7.1 Hz, 4H), 2.67-2.53 (m, 2H), 2.50-2.39 (m, 1H), 2.01 (dd, J=17.0, 4.3 Hz, 1H), 1.90-1.78 (m, 1H), 1.73-1.62 (m, 1H), 1.51-1.37 (m, 1H), 1.25 (t, J=5.9 Hz, 6H), 1.17 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 447.0 [M+H]+.


Oxetan-3-yl (R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-262)

Using a similar procedure as described for DX2-260 with DX2-235 (35 mg, 0.09 mmol) and oxetan-3-ol (7.4 mg, 0.10 mmol), white solid (32 mg, 78%). 1H NMR (300 MHz, CDCl3) δ 8.00 (d, J=8.3 Hz, 2H), 7.91 (d, J=8.4 Hz, 2H), 5.45 (q, J=5.8 Hz, 1H), 4.91 (t, J=7.0 Hz, 2H), 4.72-4.56 (m, 2H), 3.87-3.75 (m, 1H), 3.65-3.51 (m, 1H), 3.31 (q, J=7.1 Hz, 4H), 2.78-2.66 (m, 2H), 2.58-2.46 (m, 1H), 2.10-1.96 (m, 1H), 1.93-1.81 (m, 1H), 1.78-1.62 (m, 1H), 1.58-1.44 (m, 1H), 1.17 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 461.0 [M+H]+.


Cyclopropyl (R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-263)

Using a similar procedure as described for DX2-260 with DX2-235 (35 mg, 0.09 mmol) and cyclopropanol (5.8 mg, 0.10 mmol), white solid (36 mg, 90%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.6 Hz, 2H), 7.90 (d, J=8.7 Hz, 2H), 4.15 (tt, J=6.6, 3.3 Hz, 1H), 3.80 (d, J=8.5 Hz, 1H), 3.60 (d, J=11.7 Hz, 1H), 3.30 (q, J=7.1 Hz, 4H), 2.69-2.55 (m, 2H), 2.47 (td, J=11.1, 3.2 Hz, 1H), 1.97 (d, J=13.2 Hz, 1H), 1.83 (dt, J=13.7, 3.8 Hz, 1H), 1.73-1.63 (m, 1H), 1.44 (q, J=14.4, 12.2 Hz, 1H), 1.17 (t, J=7.1 Hz, 6H), 0.81-0.67 (m, 4H). LC-MS (ESI) m/z 445.1 [M+H]+.


tert-Butyl (R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-266)

Using a similar procedure as described for DX2-260 with DX2-235 (35 mg, 0.09 mmol) and tert-butanol (7.4 mg, 0.10 mmol), white solid (19 mg, 46%). 1H NMR (300 MHz, CDCl3) δ 8.03-7.96 (m, 2H), 7.94-7.87 (m, 2H), 3.78 (d, J=10.9 Hz, 1H), 3.59 (d, J=11.7 Hz, 1H), 3.30 (q, J=7.1 Hz, 4H), 2.67-2.39 (m, 3H), 2.04-1.92 (m, 1H), 1.80 (s, 1H), 1.72-1.58 (m, 1H), 1.46 (s, 9H), 1.44-1.32 (m, 1H), 1.17 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 461.1 [M+H]+.


Cyclobutyl (R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX2-282)

Using a similar procedure as described for DX2-260 with DX2-235 (35 mg, 0.09 mmol) and cyclobutanol (7.2 mg, 0.10 mmol), white solid (30 mg, 73%). 1H NMR (400 MHz, CDCl3) δ 8.02-7.96 (m, 2H), 7.94-7.88 (m, 2H), 5.06-4.93 (m, 1H), 3.91-3.80 (m, 1H), 3.68-3.59 (m, 1H), 3.31 (q, J=7.2 Hz, 4H), 2.67-2.55 (m, 2H), 2.45 (td, J=11.3, 3.1 Hz, 1H), 2.41-2.31 (m, 2H), 2.14-1.98 (m, 3H), 1.91-1.77 (m, 2H), 1.74-1.61 (m, 2H), 1.50-1.39 (m, 1H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 459.1 [M+H]+.




embedded image


Ethyl (3R)-1-(4-(N,N-diethylsulfamoyl)phenylsulfonimidoyl)piperidine-3-carboxylate (DX2-225)

To a solution of 2a (100 mg, 0.34 mmol) in THF (4 mL) was added nBuLi (0.14 mL, 0.34 mmol, 2.5 M in hexane) dropwise and the mixture was stirred at the same temperature for 1 h. N-Sulfinyltriphenylmethylamine (104 mg, 0.34 mmol) was then added as a THF solution dropwise and the mixture was stirred at the same temperature for 25 min before warmed to 0° C. in an ice bath. After stirred for 5 min, BuOCl (39 mg, 0.36 mmol) was added in a darkened hood with aluminum foil covering the flask and stirred for 25 min. Then ethyl (R)-piperidine-3-carboxylate (80 mg, 0.51 mmol) and Et3N (34 mg, 0.34 mmol) was added, and the mixture was stirred at room temperature overnight and quenched with MsOH (326 mg, 3.40 mmol), diluted with DCM, washed with sat. aq. NaHCO3, brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified with preparative HPLC to give DX2-225 as a white solid (57 mg, 39%). 1H NMR (300 MHz, CDCl3) δ 8.20 (dd, J=6.0, 3.4 Hz, 1H), 8.05 (dd, J=5.9, 3.5 Hz, 1H), 7.68 (dd, J=5.9, 3.4 Hz, 2H), 4.13 (q, J=7.1 Hz, 2H), 3.95 (d, J=13.9 Hz, 1H), 3.83 (d, J=13.4 Hz, 1H), 3.53-3.34 (m, 4H), 2.98 (dd, J=13.0, 10.6 Hz, 1H), 2.91-2.80 (m, 1H), 2.72-2.61 (m, 1H), 2.22-2.03 (m, 1H), 1.85-1.47 (m, 3H), 1.25 (t, J=7.1 Hz, 3H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 432.0 [M+H]+.




embedded image


Ethyl (R)-1-((4-(N-(tert-butyldimethylsilyl)sulfamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (13)

To a solution of DX2-300 (200 mg, 0.54 mmol) in THF was added NaH (32 mg, 0.82 mmol, 60% in mineral oil) at 0° C. portionwise. The resulting mixture was stirred at the same temperature for 15 min and tert-butyldimethylsilyl chloride (100 mg, 0.64 mmol) was added. The mixture was stirred at 0° C. for 5 min and allowed to warm to room temperature and stirred for 3 h. The mixture was quenched by ice-cooled water, extracted by EtOAc, washed by brine, dried over anhydrous Na2SO4, filtered and purified with flash chromatography (20% EtOAc in hexane) to give 13 as a white solid (165 mg, 62%). 1H NMR (300 MHz, CDCl3) δ 8.09-8.01 (m, 2H), 7.90 (dd, J=8.5, 1.6 Hz, 2H), 4.53 (s, 1H), 4.16 (q, J=7.1 Hz, 2H), 3.86 (d, J=8.0 Hz, 1H), 3.64 (d, J=11.8 Hz, 1H), 2.60 (dd, J=12.8, 8.0 Hz, 2H), 2.43 (t, J=11.2 Hz, 1H), 2.03 (d, J=13.4 Hz, 1H), 1.84 (d, J=13.3 Hz, 1H), 1.77-1.60 (m, 1H), 1.53-1.36 (m, 1H), 1.34-1.23 (m, 3H), 0.93 (s, 9H), 0.25 (s, 6H). LC-MS (ESI) m/z 489.3 [M−H].


Ethyl (3R)-1-((4-(N′-(tert-butyldimethylsilyl)-N,N-diethylsulfamidimidoyl)phenyl)sulfonyl)piperidine-3-carboxylate (14)

To a suspension of Ph3PCl2 (30 mg, 0.09 mmol) in CHCl3 (0.3 mL) under argon was added Et3N (12.4 mg, 0.12 mmol). The mixture was stirred at room temperature for 10 min and light yellow suspension was formed. The mixture was then cooled to 0° C., and 13 (40 mg, 0.08 mmol) in CHCl3 was added. It was stirred at the same temperature for 20 min before a solution of diethylamine (18 mg, 0.24 mmol) in CHCl3 (0.8 mL) was added in one portion. The resulting mixture was stirred at 0° C. for 30 min then room temperature for 1 h. The mixture was concentrated and directly used in the next step without further purification. LC-MS (ESI) m/z 546.4 [M+H]+.


Ethyl (3R)-1-((4-(N,N-diethylsulfamidimidoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX3-90)

14 (43 mg, 0.08 mmol) was dissolved in CH3CN (0.5 mL), and formic acid (37 mg 0.80 mmol) and H2O (0.03 mL) was added. The mixture was stirred at room temperature for 5 h. It was purified with preparative HPLC to give DX3-90 as a white solid (10 mg, 46%). 1H NMR (300 MHz, MeOD) (8.17 (d, J=8.6 Hz, 2H), 7.98 (d, J=8.6 Hz, 2H), 4.16 (q, J=7.1 Hz, 2H), 3.66 (dd, J=11.6, 3.9 Hz, 1H), 3.51-3.33 (m, 4H), 3.32-3.20 (m, 1H), 2.80 (t, J=10.5 Hz, 1H), 2.69-2.56 (m, 2H), 1.98-1.77 (m, 2H), 1.71-1.45 (m, 2H), 1.27 (t, J=7.1 Hz, 3H), 1.14 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 432.1 [M+H]+.




embedded image


Sodium 4-(N,N-diethylsulfamoyl)benzoate (15)

To a solution of 2a (100 mg, 0.34 mmol) in THF (2 mL) was added nBuLi (136 μL, 0.34 mmol) dropwise under argon at −78° C. The color turned yellow-brown. The mixture was stirred at the same temperature for 20 min and a suspension of DABSO (82 mg, 0.34 mmol) in THF (1 mL) was added dropwise. The mixture was stirred at the same temperature for another 15 min and stirred at room temperature for 3 h. The mixture was partitioned between Et3O and 10% Na2CO3, and the aqueous layer was acidified with 1N HCl and extracted with Et2O. The organic layer was then extracted with 10% Na2CO3, and the aqueous layer was concentrated and boiled with EtOH (5 mL) for 1 h, filtered and concentrated to give 15 as a white solid (60 mg, 59%). 1H NMR (300 MHz, MeOD) (7.87 (q, J=8.3 Hz, 4H), 3.25 (q, J=7.1 Hz, 4H), 1.13 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 276.2 [M−H].


Ethyl 3-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)benzoate (DX3-3)

A solution of 15 (50 mg, 0.17 mmol), (3-(ethoxycarbonyl)phenyl)boronic acid (32 mg, 0.17 mmol), Cu(OAc)2 (36 mg, 0.20 mmol) and K2CO3 (46 mg, 0.33 mmol) in DMSO (1.5 mL) was stirred at room temperature for 3 h. The mixture was then diluted with EtOAc, washed with water, brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified with flash chromatography (10% EtOAc in hexane) to give DX3-3 as a white solid (24 mg, 33%). 1H NMR (400 MHz, CDCl3) δ 8.62 (d, J=1.9 Hz, 1H), 8.30 (dq, J=7.5, 1.2 Hz, 1H), 8.15 (ddt, J=7.9, 2.0, 1.0 Hz, 1H), 8.12-8.06 (m, 2H), 7.99-7.92 (m, 2H), 7.66 (t, J=7.8 Hz, 1H), 4.44 (qd, J=7.1, 0.8 Hz, 2H), 3.31-3.22 (m, 4H), 1.43 (td, J=7.1, 0.8 Hz, 3H), 1.16 (td, J=7.2, 0.8 Hz, 6H). LC-MS (ESI) m/z 426.0 [M+H]+.




embedded image


(R)-1-(4-(N,N-Diethylsulfamoyl)phenyl)-N′-hydroxypiperidine-3-carboximidamide (16)

To a solution of DX3-37 (100 mg, 0.26 mmol) in EtOH (0.5 mL) was added NH2OH aqueous solution (17 mg, 0.52 mmol) and heated under microwave irradiation at 75° C. for 5 h. The mixture was concentrated to give 16 as a white solid (54 mg, 50%) which was used directly in the next step. 1H NMR (400 MHz, CDCl3) δ 8.00 (d, J=7.9 Hz, 2H), 7.95-7.87 (m, 2H), 3.70 (d, J=11.3 Hz, 1H), 3.57 (s, 1H), 3.32 (q, J=7.2 Hz, 4H), 2.79 (t, J=10.8 Hz, 1H), 2.66-2.51 (m, 2H), 2.00-1.62 (m, 4H), 1.19 (t, J=7.2 Hz, 6H).


(R)—N,N-Diethyl-4-((3-(5-oxo-4,5-dihydro-1,2,4-oxadiazol-3-yl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-43)

To a solution of 16 (54 mg, 0.13 mmol) and pyridine (12 mg, 0.15 mmol) in DMF (1 mL) was added isobutyl chloroformate (41 mg, 0.13 mmol) dropwise at 0° C. The mixture was stirred for 16 h while slowly warming to room temperature. The reaction was diluted with H2O and extracted with EtOAc. The combined organic extracts were washed with H2O, brine, dried over Na2SO4, filtered, and concentrated. The residue was suspended in toluene (1.0 mL) in a microwave vial and stirred at 120° C. for 2 h, then at 140° C. for 5 h. After cooling to room temperature, the mixture was concentrated. Purification by flash chromatography (10% MeOH in DCM) gave DX3-43 as a white solid (19 mg, 38%). 1H NMR (300 MHz, CDCl3) δ 8.01 (d, J=8.7 Hz, 2H), 7.91 (d, J=8.8 Hz, 2H), 3.82-3.70 (m, 1H), 3.52 (d, J=11.9 Hz, 1H), 3.31 (q, J=7.1 Hz, 4H), 3.04 (dt, J=9.3, 5.2 Hz, 1H), 2.87 (dd, J=11.8, 8.9 Hz, 1H), 2.76 (t, J=10.1 Hz, 1H), 2.09-1.87 (m, 2H), 1.85-1.63 (m, 2H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 443.3 [M−H].


(R)-4-(3-(1H-Tetrazol-5-yl)piperidin-1-yl)-N,N-diethylbenzenesulfonamide (DX3-38)

To a solution of DX3-37 (55 mg, 0.14 mmol) in toluene (4 mL) was added TMSN3 (95 mg, 0.83 mmol) and AlMe3 (0.42 mL, 0.83 mmol, 2 M in toluene). The mixture was heated at 80° C. overnight, diluted with EtOAc, washed with water, brine, dried over anhydrous Na2SO4, filtered, concentrated and purified with preparative HPLC to give DX3-38 as a white solid (6 mg, 10%). 1H NMR (400 MHz, CDCl3) δ 8.02 (d, J=8.3 Hz, 2H), 7.93 (d, J=8.3 Hz, 2H), 3.57-3.39 (m, 3H), 3.33 (q, J=7.1 Hz, 4H), 3.28-3.19 (m, 2H), 2.12-2.02 (m, 2H), 1.83-1.72 (m, 2H), 1.20 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 429.1 [M+H]+.




embedded image


(R)-1-(tert-Butoxycarbonyl)-4-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperazine-2-carboxylic acid (17)

To a solution of 4a (120 mg, 0.39 mmol) in dioxane (4 mL) and H2O (2 mL) was added Na2CO3 (82 mg, 0.77 mmol). The mixture was stirred at room temperature overnight, diluted with EtOAc, washed with water, brine, dried over anhydrous Na2SO4, filtered, concentrated to give 17 as a white solid (176 mg, 90%). 1H NMR (400 MHz, MeOD) (8.07 (d, J=8.5 Hz, 2H), 7.99 (d, J=8.5 Hz, 2H), 4.73 (d, J=23.2 Hz, 1H), 4.37 (s, 1H), 4.30 (t, J=14.2 Hz, 1H), 3.93 (d, J=13.3 Hz, 1H), 3.74 (dd, J=21.8, 11.8 Hz, 1H), 3.32-3.27 (m, 4H), 3.24-3.14 (m, 1H), 2.64-2.56 (m, 1H), 2.37 (t, J=12.0 Hz, 1H), 1.44 (d, J=9.2 Hz, 9H), 1.16 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 504.3 [M−H].


Ethyl (R)-4-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperazine-2-carboxylate (DX3-44B)

To a solution of 17 (44 mg, 0.087 mmol) in EtOH (2 mL) was added conc. H2SO4 (50 μL). The mixture was heated at reflux overnight, concentrated and partitioned between EtOAc and sat. NaHCO3. The organic layer was washed with brine, dried over anhydrous Na2SO4, filtered, concentrated. The residue was purified with flash chromatography (5% MeOH in DCM) to give DX3-44B as a white solid (16 mg, 42%). 1H NMR (300 MHz, CDCl3) δ 8.02-7.97 (m, 2H), 7.90 (d, J=8.8 Hz, 2H), 4.24 (q, J=7.2 Hz, 2H), 3.75-3.68 (m, 1H), 3.59 (dd, J=8.4, 3.4 Hz, 1H), 3.47-3.34 (m, 1H), 3.30 (q, J=7.2 Hz, 4H), 3.19-3.10 (m, 1H), 2.98-2.87 (m, 1H), 2.81 (dd, J=11.2, 8.4 Hz, 1H), 2.75-2.63 (m, 1H), 1.32 (t, J=7.1 Hz, 3H), 1.18 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 475.1 [M+CH3CN+H]+.


Ethyl (R)-4-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)-1-methylpiperazine-2-carboxylate (DX3-48B)

To a solution of DX3-44B (20 mg, 0.046 mmol) and K2CO3 (9.5 mg, 0.069 mmol) in CH3CN (1 mL) was added CH3I (13 mg, 0.092 mmol). The mixture was heated at 50° C. under microwave for 2 h. The mixture was diluted with EtOAc, washed with water, brine, dried over anhydrous Na2SO4, filtered, concentrated. The residue was purified with flash chromatography (50% EtOAc in hexane) to give DX3-48B as a white solid (mg, %). 1H NMR (400 MHz, CDCl3) δ 8.02 (d, J=8.6 Hz, 2H), 7.88 (d, J=8.6 Hz, 2H), 4.44-4.26 (m, 2H), 3.92 (s, 1H), 3.83 (s, 1H), 3.79-3.68 (m, 1H), 3.62-3.50 (m, 1H), 3.41 (d, J=12.4 Hz, 1H), 3.29 (q, J=7.1 Hz, 4H), 3.26-3.17 (m, 1H), 3.05 (d, J=12.1 Hz, 1H), 2.84 (s, 3H), 1.39 (t, J=7.2 Hz, 3H), 1.19 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 489.2 [M+CH3CN+H]+.




embedded image


Ethyl (R)-1-((4-(diethylcarbamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX3-78B)

To a solution of ethyl (R)-piperidine-3-carboxylate (35 mg, 0.22 mmol) and triethylamine (36 mg, 0.36 mmol) in DCM (2 mL) was added 4-(diethylcarbamoyl)benzenesulfonyl chloride (18, 50 mg, 0.18 mmol). The mixture was stirred at room temperature for 3 h. The mixture was concentrated and purified with flash chromatography (20% EtOAc in hexane) to give DX3-78B as a colorless gel (59 mg, 83%). 1H NMR (300 MHz, CDCl3) δ 7.83 (d, J=8.6 Hz, 2H), 7.60-7.51 (m, 2H), 4.16 (q, J=7.1 Hz, 2H), 3.86 (d, J=8.8 Hz, 1H), 3.59 (q, J=9.8, 7.0 Hz, 3H), 3.23 (q, J=6.8 Hz, 2H), 2.70-2.54 (m, 2H), 2.41 (td, J=11.2, 3.1 Hz, 1H), 2.06-1.96 (m, 1H), 1.83 (dt, J=12.9, 3.8 Hz, 1H), 1.74-1.64 (m, 1H), 1.43 (t, J=10.9 Hz, 1H), 1.28 (t, J=7.1 Hz, 6H), 1.14 (t, J=7.0 Hz, 3H). LC-MS (ESI) m/z 397.1 [M+H]+.




embedded image


(R)-4-((3-(Ethoxycarbonyl)piperidin-1-yl)sulfonyl)benzoic acid (20a)

To a solution of ethyl (R)-piperidine-3-carboxylate (426 mg, 2.71 mmol) and Et3N (457 mg, 4.52 mmol) in THF (10 mL) was added 4-(chlorosulfonyl)benzoic acid (19, 500 mg, 2.26 mmol). The mixture was stirred at room temperature overnight. The mixture was then concentrated, diluted with EtOAc, washed with 1N HCl, brine, dried over anhydrous Na2SO4, filtered and concentrated to give 20a as a white solid (604 mg, 78%). 1H NMR (400 MHz, CDCl3) δ 12.19 (d, J=8.4 Hz, 2H), 11.85 (d, J=8.4 Hz, 2H), 7.69 (d, J=11.8 Hz, 1H), 7.51-7.41 (m, 1H), 6.63 (dd, J=11.6, 9.8 Hz, 1H), 6.58-6.42 (m, 2H), 5.91 (d, J=13.9 Hz, 1H), 5.83-5.72 (m, 1H), 5.56 (dd, J=13.7, 10.1 Hz, 1H), 5.41 (q, J=13.1, 12.5 Hz, 1H). LC-MS (ESI) m/z 342.1 [M+H]+.


Ethyl (R)-1-((4-(isopropyl(methyl)carbamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX3-102B)

To a solution of 20a (25 mg, 0.073 mmol) and HATU (42 mg, 0.11 mmol) in DMF (1 mL) was added N-methylpropan-2-amine (5.3 mg, 0.073 mmol) and DIEA (28 mg, 0.22 mmol). The mixture was stirred at room temperature overnight. The mixture was then diluted with EtOAc, washed with brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified with flash chromatography (20% EtOAc in hexane) to give DX3-102B as a colorless gel (25 mg, 86%). 1H NMR (300 MHz, CDCl3) δ 7.82 (d, J=8.3 Hz, 2H), 7.60-7.47 (m, 2H), 4.15 (q, J=7.1 Hz, 2H), 3.83 (t, J=8.1 Hz, 2H), 3.62 (d, J=11.7 Hz, 1H), 2.98 (s, 2H), 2.76 (s, 1H), 2.71-2.51 (m, 2H), 2.48-2.33 (m, 1H), 2.01 (dd, J=14.0, 3.9 Hz, 1H), 1.82 (dp, J=11.3, 3.8 Hz, 1H), 1.76-1.57 (m, 1H), 1.51-1.31 (m, 1H), 1.32-1.12 (m, 9H). LC-MS (ESI) m/z 397.1 [M+H]+.


Ethyl (R)-1-((4-(ethyl(propyl)carbamoyl)phenyl)sulfonyl)piperidine-3-carboxylate (DX3-103)

To a solution of 20a (25 mg, 0.073 mmol) and HATU (42 mg, 0.11 mmol) in DMF (1 mL) was added N-ethylpropan-1-amine (6.4 mg, 0.073 mmol) and DIEA (28 mg, 0.22 mmol). The mixture was stirred at room temperature overnight. The mixture was then diluted with EtOAc, washed with brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified with flash chromatography (20% EtOAc in hexane) to give DX3-103 as a colorless gel (20 mg, 67%). 1H NMR (300 MHz, CDCl3) δ 7.82 (d, J=8.3 Hz, 2H), 7.53 (d, J=7.9 Hz, 2H), 4.15 (q, J=7.1 Hz, 2H), 3.84 (d, J=8.5 Hz, 1H), 3.60 (t, J=9.8 Hz, 2H), 3.48 (t, J=7.6 Hz, 1H), 3.22 (q, J=7.1 Hz, 1H), 3.12 (t, J=7.6 Hz, 1H), 2.70-2.52 (m, 2H), 2.40 (td, J=11.3, 3.1 Hz, 1H), 2.06-1.94 (m, 1H), 1.82 (dt, J=13.2, 3.8 Hz, 1H), 1.76-1.62 (m, 2H), 1.55 (q, J=8.7 Hz, 1H), 1.49-1.35 (m, 1H), 1.27 (t, J=7.2 Hz, 4.3H), 1.11 (t, J=7.1 Hz, 1.3H), 1.00 (t, J=7.4 Hz, 1.7H), 0.77 (t, J=7.3 Hz, 1.7H). LC-MS (ESI) m/z 433.1 [M+Na]+.


Ethyl (R)-1-(4-(N,N-diethylsulfamoyl)benzoyl)piperidine-3-carboxylate (DX3-79)

To a solution of commercially available 4-(N,N-diethylsulfamoyl)benzoic acid (20b, 50 mg, 0.19 mmol) and HATU (108 mg, 0.29 mmol) in DMF (2 mL) was added ethyl (R)-piperidine-3-carboxylate (37 mg, 0.23 mmol) and DIEA (49 mg, 0.38 mmol). The mixture was stirred at room temperature overnight. The mixture was then diluted with EtOAc, washed with brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified with flash chromatography (20% EtOAc in hexane) to give DX3-79 as a colorless gel (72 mg, 96%). 1H NMR (300 MHz, CDCl3) δ 7.88 (d, J=8.3 Hz, 2H), 7.53 (d, J=7.9 Hz, 2H), 4.74-4.27 (m, 1H), 4.17 (s, 2H), 3.58 (d, J=46.6 Hz, 1H), 3.26 (q, J=7.1 Hz, 6H), 2.54 (dt, J=48.8, 14.7 Hz, 1H), 2.15 (dd, J=15.1, 6.1 Hz, 1H), 1.90-1.71 (m, 2H), 1.35-1.22 (m, 3H), 1.17 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 397.1 [M+H]+.




embedded image


N,N-Diethyl-4-fluorobenzenesulfonamide (22)

To a solution of diethylamine (360 mg, 4.93 mmol) and triethylamine (996 mg, 9.86 mmol) was added 4-fluorobenzenesulfonyl chloride (21, 962 mg, 4.93 mmol) portionwise. The mixture was stirred at room temperature overnight. The mixture was concentrated and purified with flash chromatography (10% EtOAc in hexane) to give 22 as a colorless oil. (940 mg, 83%). 1H NMR (400 MHz, CDCl3) δ 7.89-7.80 (m, 2H), 7.24-7.14 (m, 2H), 3.27 (q, J=7.1 Hz, 4H), 1.16 (t, J=7.2 Hz, 6H).


N,N-Diethyl-4-mercaptobenzenesulfonamide (23)

22 (400 mg, 1.74 mmol) and NaSMe (484 mg, 6.92 mmol) was heated in DMF (2 mL) under microwave for 1 h. Et2O was added and it was extracted with 1N NaOH. The aqueous layer was acidified to pH<4 with 1N HCl and extracted with Et2O. The organic layer was dried with Na2SO4, filtered and concentrated to give 23 as a white solid (410 mg, 96%). 1H NMR (400 MHz, CDCl3) δ 7.68 (d, J=8.8 Hz, 2H), 7.36 (d, J=8.7 Hz, 2H), 3.25 (q, J=7.1 Hz, 4H), 1.15 (t, J=7.1 Hz, 6H).


Ethyl 3-((methylsulfonyl)oxy)cyclohexane-1-carboxylate (25)

To a solution of ethyl 3-hydroxycyclohexane-1-carboxylate (24, 500 mg, 2.91 mmol) and triethylamine (588 mg, 5.82 mmol) was added methanesulfonyl chloride (401 mg, 3.49 mmol) portionwise. The mixture was stirred at room temperature for 3 h. The mixture was concentrated and purified with flash chromatography (10% EtOAc in hexane) to give 25 as a. (652 mg, 90%). 1H NMR (400 MHz, CDCl3) δ 5.08 (s, 0.4H), 4.65 (td, J=10.9, 5.2 Hz, 0.6H), 4.16 (q, J=7.1 Hz, 2H), 3.05 (d, J=3.0 Hz, 3H), 2.82-2.72 (m, 0.4H), 2.44 (d, J=11.1 Hz, 1H), 2.19 (d, J=12.8 Hz, 1H), 1.96 (d, J=12.7 Hz, 2.6H), 1.78-1.65 (m, 2H), 1.56 (d, J=9.0 Hz, 1H), 1.40 (dd, J=11.9, 9.5 Hz, 1H), 1.28 (t, J=7.2 Hz, 3H).


Ethyl 3-((4-(N,N-diethylsulfamoyl)phenyl)thio)cyclohexane-1-carboxylate (26)

To a solution of 23 (52 mg, 0.21 mmol) and 25 (53 mg, 0.21 mmol) was added Cs2CO3 (102 mg, 0.32 mmol) portionwise. The mixture was stirred at 75° C. under argon for 5 h. The mixture was concentrated and purified with flash chromatography (30% EtOAc in hexane) to give 26 as a colorless oil. (56 mg, 67%). 1H NMR (400 MHz, CDCl3) δ 7.71 (dd, J=8.6, 2.3 Hz, 2H), 7.43 (t, J=8.3 Hz, 2H), 4.16 (dq, J=17.6, 7.1 Hz, 2H), 3.74 (tt, J=7.6, 3.8 Hz, 0.4H), 3.29-3.22 (m, 4H), 3.22-3.16 (s, 0.6H), 2.81 (td, J=7.1, 3.7 Hz, 0.4H), 2.46-2.22 (m, 1.6H), 2.12-1.99 (m, 1H), 1.98-1.90 (m, 1H), 1.80 (dddd, J=16.4, 13.5, 7.4, 4.3 Hz, 2H), 1.71-1.52 (m, 2H), 1.47-1.36 (m, 1H), 1.28 (dt, J=13.2, 7.1 Hz, 3H), 1.15 (td, J=7.1, 1.5 Hz, 6H). LC-MS (ESI) m/z 400.1 [M+H]+.


(cis)-Ethyl 3-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)cyclohexane-1-carboxylate (DX3-101-P1) and (trans)-ethyl 3-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)cyclohexane-1-carboxylate (DX3-101-P2)

To a solution of 26 (53 mg, 0.14 mmol) in DCM (3 mL) was added mCPBA (91 mg, 0.54 mmol) and stirred at room temperature overnight. Sat. NaHCO3 was added and the mixture was extracted with EtOAc. The organic layer was washed with brine, dried with Na2SO4, filtered and concentrated. The residue was purified with preparative HPLC to give DX3-101-P1 (10 mg, 21%) and DX3-101-P2 (7 mg, 15%) as white solids. DX3-101-P1 1H NMR (300 MHz, CDCl3) (8.02 (s, 4H), 4.14 (q, J=7.1 Hz, 2H), 3.31 (q, J=7.1 Hz, 4H), 3.07-2.93 (m, 1H), 2.33 (d, J=12.8 Hz, 2H), 2.11 (d, J=11.6 Hz, 1H), 2.01 (d, J=8.0 Hz, 2H), 1.67-1.54 (m, 1H), 1.48-1.31 (m, 3H), 1.26 (t, J=7.1 Hz, 3H), 1.17 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 432.0 [M+H]+.


DX3-101-P2 1H NMR (300 MHz, CDCl3) δ 8.03 (d, J=1.2 Hz, 4H), 4.12 (q, J=7.1 Hz, 2H), 3.46 (tt, J=11.4, 3.8 Hz, 1H), 3.30 (q, J=7.1 Hz, 4H), 2.95-2.84 (m, 1H), 2.26 (d, J=13.4 Hz, 1H), 2.12-1.91 (m, 2H), 1.83 (dt, J=13.3, 3.9 Hz, 1H), 1.72 (ddd, J=13.3, 11.5, 4.8 Hz, 1H), 1.65-1.49 (m, 2H), 1.48-1.34 (m, 1H), 1.23 (t, J=7.1 Hz, 3H), 1.16 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 432.0 [M+H]+.




embedded image


tert-Butyl (S)-3-((4-(N,N-diethylsulfamoyl)phenyl)thio)piperidine-1-carboxylate (28a)

A solution of tert-butyl (R)-3-((methylsulfonyl)oxy)piperidine-1-carboxylate (27a, 137 mg, 0.49 mmol), 23 (120 mg, 0.49 mmol) and Cs2CO3 (239 mg, 0.74 mmol) in DMF (5 mL) was heated at 110° C. under microwave for 6 h. The mixture was diluted with EtOAc, washed with H2O, brine and dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified with flash chromatography (5% MeOH in DCM) to give 28a as a white solid (160 mg, 76%). 1H NMR (300 MHz, CDCl3) δ 7.72 (d, J=8.5 Hz, 2H), 7.43 (d, J=8.2 Hz, 2H), 3.90 (d, J=13.2 Hz, 1H), 3.39-3.28 (m, 1H), 3.24 (q, J=7.1 Hz, 4H), 3.07-2.83 (m, 2H), 2.21-2.09 (m, 1H), 1.90-1.72 (m, 1H), 1.69-1.53 (m, 3H), 1.45 (s, 9H), 1.15 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 451.1 [M+Na]+.


tert-Butyl (S)-3-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-1-carboxylate (29a)

Using a similar procedure as described for DX3-101-P1 with 28a (80 mg, 0.19 mmol) and mCPBA (129 mg, 0.75 mmol), white solid (80 mg, 93%). 1H NMR (300 MHz, CDCl3) δ 8.02 (s, 4H), 4.26 (d, J=12.2 Hz, 1H), 4.06-3.84 (m, 1H), 3.27 (q, J=7.1 Hz, 4H), 2.98 (dt, J=23.3, 7.7 Hz, 2H), 2.77-2.55 (m, 1H), 2.23-2.10 (m, 1H), 1.92-1.66 (m, 2H), 1.52-1.44 (m, 1H), 1.38 (s, 9H), 1.14 (t, J=7.1 Hz, 6H).


Ethyl (S)-3-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-1-carboxylate (DX3-112B)

To a solution of 29a (40 mg, 0.087 mmol) in DCM (1 mL) was added TFA (0.2 mL) and the resulting mixture was stirred at room temperature for 2 h. It was concentrated and dissolved by DCM (1 mL). Et3N (18 mg, 0.17 mmol) and ethyl chloroformate (11 mg, 0.10 mmol) were added subsequently. The mixture was stirred for 2 h at room temperature and concentrated. The residue was purified with flash chromatography (50% EtOAc in hexane) to give DX3-112B (16 mg, 43%) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.05 (s, 4H), 4.39 (d, J=11.4 Hz, 1H), 4.18-3.98 (m, 3H), 3.31 (q, J=7.2 Hz, 4H), 3.17-2.97 (m, 2H), 2.86-2.69 (m, 1H), 2.24-2.10 (m, 1H), 1.93-1.70 (m, 2H), 1.56-1.40 (m, 1H), 1.24 (t, J=7.1 Hz, 3H), 1.17 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 433.0 [M+H]+.


tert-Butyl (R)-3-((4-(N,N-diethylsulfamoyl)phenyl)thio)piperidine-1-carboxylate (28b)

Using a similar procedure as described for 28a with tert-butyl (S)-3-((methylsulfonyl)oxy)piperidine-1-carboxylate (27b, 80 mg, 0.19 mmol), 23 (120 mg, 0.49 mmol) and Cs2CO3 (239 mg, 0.74 mmol), white solid (79 mg, 38%). 1H NMR (300 MHz, CDCl3) δ 7.72 (d, J=8.5 Hz, 2H), 7.43 (d, J=8.1 Hz, 2H), 3.90 (d, J=13.1 Hz, 1H), 3.39-3.31 (m, 1H), 3.24 (q, J=7.2 Hz, 4H), 3.04-2.84 (m, 2H), 2.21-2.08 (m, 1H), 1.91-1.75 (m, 1H), 1.69-1.50 (m, 3H), 1.45 (s, 9H), 1.16 (t, J=7.1 Hz, 6H).


tert-Butyl (R)-3-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-1-carboxylate (29b)

Using a similar procedure as described for 29a with 28b (70 mg, 0.16 mmol) and mCPBA (113 mg, 0.65 mmol), white solid (60 mg, 80%). 1H NMR (300 MHz, CDCl3) δ 8.03 (s, 4H), 4.28 (d, J=12.3 Hz, 1H), 4.01 (s, 1H), 3.29 (q, J=7.2 Hz, 4H), 3.14-2.88 (m, 1H), 2.81-2.58 (m, 1H), 2.18 (d, J=13.4 Hz, 1H), 1.95-1.64 (m, 2H), 1.54-1.46 (m, 1H), 1.40 (s, 9H), 1.16 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 483.1 [M+Na]+.


Ethyl (R)-3-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-1-carboxylate (DX3-114)

Using a similar procedure as described for DX3-112B with 29b (40 mg, 0.087 mmol), Et3N (18 mg, 0.17 mmol) and ethyl chloroformate (11 mg, 0.10 mmol), white solid (23 mg, 61%). 1H NMR (300 MHz, CDCl3) δ 8.04 (s, 4H), 4.39 (d, J=11.4 Hz, 1H), 4.12 (q, J=7.1 Hz, 3H), 3.31 (q, J=7.1 Hz, 4H), 3.14-2.96 (m, 2H), 2.88-2.67 (m, 1H), 2.24-2.10 (m, 1H), 1.83 (s, 2H), 1.56-1.39 (m, 1H), 1.24 (t, J=7.1 Hz, 3H), 1.17 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 433.1 [M+H]+.




embedded image


(R)—N-(Azetidin-3-yl)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxamide (DX3-110)

To a solution of DX3-107 (27 mg, 0.048 mmol) in DCM (1 mL) was added TFA (0.2 mL). The mixture was stirred at room temperature for 2 h and concentrated. The residue was purified with preparative HPLC to give DX3-110 as a white solid (21 mg, 95%). 1H NMR (300 MHz, CDCl3) (9.20 (s, 1H), 8.27 (d, J=7.4 Hz, 1H), 7.98 (d, J=8.3 Hz, 2H), 7.89 (d, J=8.3 Hz, 2H), 4.95-4.77 (m, 1H), 4.47-4.30 (m, 2H), 4.26-4.08 (m, 2H), 3.69 (d, J=10.8 Hz, 1H), 3.54 (d, J=5.9 Hz, 1H), 3.29 (q, J=7.1 Hz, 4H), 2.69 (t, J=10.5 Hz, 1H), 2.63-2.44 (m, 2H), 1.92-1.75 (m, 2H), 1.68-1.45 (m, 2H), 1.16 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 459.1 [M+H]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N-(1-ethylazetidin-3-yl)piperidine-3-carboxamide (DX3-115B)

To a solution of DX3-110 (15 mg, 0.033 mmol) and acetaldehyde (1.7 mg, 0.039 mmol) in MeOH (1 mL) was added 1 drop of AcOH followed by NaBH3CN (3.2 mg, 0.05 mmol). The mixture was stirred at room temperature for 5 h and partitioned between EtOAc and sat. NaHCO3 solution. The organic layer was washed with brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified with flash chromatography (10% MeOH in DCM) to give DX3-115B as a white solid (8.3 mg, 52%). 1H NMR (300 MHz, CDCl3) δ 9.04 (d, J=8.3 Hz, 1H), 7.95 (q, J=8.6, 8.2 Hz, 4H), 5.10-4.82 (m, 1H), 4.53-4.03 (m, 4H), 3.76 (d, J=12.1 Hz, 1H), 3.68-3.49 (m, 1H), 3.30 (q, J=7.1 Hz, 4H), 3.17 (dt, J=13.2, 6.9 Hz, 2H), 2.78 (t, J=10.8 Hz, 1H), 2.65-2.48 (m, 2H), 1.99-1.77 (m, 2H), 1.76-1.46 (m, 2H), 1.30 (t, J=7.2 Hz, 3H), 1.17 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 487.1 [M+H]+.


Ethyl (R)-3-(1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxamido)azetidine-1-carboxylate (DX3-116)

To a solution of DX3-110 (20 mg, 0.044 mmol) and Et3N (9 mg, 0.088 mmol) in DCM (1 mL) was added ethyl chloroformate (5.7 mg, 0.052 mmol). The mixture was stirred at room temperature for 5 h and concentrated. The residue was purified with flash chromatography (10% MeOH in DCM) to give DX3-116 as a white solid (9 mg, 39%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.5 Hz, 2H), 7.89 (d, J=8.5 Hz, 2H), 6.61 (d, J=7.1 Hz, 1H), 4.73-4.58 (m, 1H), 4.32 (ddd, J=9.3, 7.8, 4.5 Hz, 2H), 4.13 (q, J=7.1 Hz, 2H), 3.83 (dt, J=9.4, 5.9 Hz, 2H), 3.68 (d, J=11.6 Hz, 1H), 3.59 (d, J=11.5 Hz, 1H), 3.31 (q, J=7.1 Hz, 4H), 2.80-2.67 (m, 1H), 2.63-2.43 (m, 2H), 1.91-1.55 (m, 4H), 1.26 (td, J=7.2, 1.2 Hz, 3H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 531.1 [M+H]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N-(1-pivaloylazetidin-3-yl)piperidine-3-carboxamide (DX3-116B)

To a solution of DX3-110 (20 mg, 0.044 mmol) and Et3N (9 mg, 0.088 mmol) in DCM (1 mL) was added pivaloyl chloride (6.3 mg, 0.052 mmol). The mixture was stirred at room temperature for 5 h and concentrated. The residue was purified with flash chromatography (10% MeOH in DCM) to give DX3-116B as a white solid (19 mg, 7 9%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.5 Hz, 2H), 7.88 (d, J=8.5 Hz, 2H), 6.93 (s, 1H), 4.63 (d, J=6.4 Hz, 2H), 4.49-3.85 (m, 3H)z, 3.66 (dd, J=30.7, 11.5 Hz, 2H), 3.31 (q, J=7.1 Hz, 4H), 2.71 (t, J=10.7 Hz, 1H), 2.54 (d, J=10.4 Hz, 2H), 1.85 (q, J=9.9 Hz, 2H), 1.78-1.56 (m, 2H), 1.25-1.14 (m, 15H). LC-MS (ESI) m/z 543.3 [M+H]+.


(R)—N-(1-(Cyclopropanecarbonyl)azetidin-3-yl)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxamide (DX3-117B)

Using a similar procedure as described for DX3-116B with DX3-110 (20 mg, 0.044 mmol), Et3N (9 mg, 0.088 mmol) and cyclopropanecarbonyl chloride (5.5 mg, 0.052 mmol), white solid (20 mg, 87%). 1H NMR (300 MHz, CDCl3) δ 8.00 (d, J=8.3 Hz, 2H), 7.89 (d, J=8.4 Hz, 2H), 6.69 (dd, J=22.8, 7.0 Hz, 1H), 4.79-4.54 (m, 2H), 4.42-4.30 (m, 1H), 4.14-4.02 (m, 1H), 3.95-3.80 (m, 1H), 3.66 (d, J=12.0 Hz, 1H), 3.56 (dt, J=11.2, 4.6 Hz, 1H), 3.31 (q, J=7.1 Hz, 4H), 2.86-2.69 (m, 1H), 2.69-2.46 (m, 2H), 1.92-1.64 (m, 4H), 1.47-1.36 (m, 1H), 1.18 (t, J=7.1 Hz, 6H), 0.98 (s, 2H), 0.80 (d, J=6.6 Hz, 2H). LC-MS (ESI) m/z 527.2 [M+H]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N-(1-(methylsulfonyl)azetidin-3-yl)piperidine-3-carboxamide (DX3-117)

To a solution of DX3-110 (20 mg, 0.044 mmol) and Et3N (9 mg, 0.088 mmol) in DCM (1 mL) was added methanesulfonyl chloride (6 mg, 0.052 mmol). The mixture was stirred at room temperature for 2 h and concentrated. The residue was purified with flash chromatography (10% MeOH in DCM) and then preparative HPLC to give DX3-117 as a white solid (2.8 mg, 12%). 1H NMR (300 MHz, CDCl3) δ 8.00 (d, J=8.2 Hz, 2H), 7.89 (d, J=8.2 Hz, 2H), 6.54 (d, J=7.5 Hz, 1H), 4.69 (q, J=7.2 Hz, 1H), 4.17 (q, J=7.6 Hz, 2H), 3.93 (q, J=6.8 Hz, 2H), 3.63 (d, J=11.9 Hz, 1H), 3.53 (d, J=11.6 Hz, 1H), 3.31 (q, J=7.1 Hz, 4H), 2.94 (s, 3H), 2.81 (t, J=10.5 Hz, 1H), 2.72-2.57 (m, 1H), 2.58-2.44 (m, 1H), 1.89-1.67 (m, 4H), 1.18 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 537.0 [M+H]+.


(R)—N-(1-(tert-butylcarbamoyl)azetidin-3-yl)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxamide (DX3-123)

To a solution of DX3-110 (20 mg, 0.044 mmol) and Et3N (9 mg, 0.088 mmol) in DCM (1 mL) was added tert-butyl isocyanate (5.2 mg, 0.053 mmol). The mixture was stirred at room temperature for 3 h and concentrated. The residue was purified with flash chromatography (10% MeOH in DCM) to give DX3-123 as a white solid (23 mg, 95%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.6 Hz, 2H), 7.89 (d, J=8.6 Hz, 2H), 6.65 (d, J=6.9 Hz, 1H), 4.61 (q, J=5.9 Hz, 1H), 4.22 (td, J=8.0, 3.6 Hz, 2H), 3.95 (s, 1H), 3.79-3.65 (m, 3H), 3.59 (d, J=11.6 Hz, 1H), 3.31 (q, J=7.1 Hz, 4H), 2.80-2.68 (m, 1H), 2.62-2.46 (m, 2H), 1.91-1.77 (m, 2H), 1.73-1.59 (m, 2H), 1.35 (s, 9H), 1.18 (t, J=7.2 Hz, 6H). LC-MS (ESI) m/z 558.1 [M+H]+.




embedded image


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N—((R)-pyrrolidin-3-yl)piperidine-3-carboxamide (30)

To a solution of DX3-118 (197 mg, 0.34 mmol) in DCM (4 mL) was added TFA (0.8 mL). The mixture was stirred at room temperature for 2 h and concentrated to give 30 as a colorless gel, which was directly used in the next step without further purification.


(R)—N—((R)-1-Benzoylpyrrolidin-3-yl)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxamide (DX3-125)

To a solution of 30 (20 mg, 0.042 mmol) and Et3N (13 mg, 0.13 mmol) in DCM (1 mL) was added benzoyl chloride (6 mg, 0.042 mmol). The mixture was stirred at room temperature for 3 h and concentrated. The residue was purified with flash chromatography (10% MeOH in DCM) to give DX3-125 as a white solid (18 mg, 75%). 1H NMR (300 MHz, CDCl3) δ 7.99 (t, J=7.6 Hz, 2H), 7.95-7.81 (m, 2H), 7.53 (t, J=6.2 Hz, 2H), 7.45 (s, 3H), 6.62 (dd, J=82.0, 6.8 Hz, 1H), 4.66-4.37 (m, 1H), 4.01-3.72 (m, 2H), 3.70-3.46 (m, 3H), 3.39-3.22 (m, 4H), 2.85-2.66 (m, 1H), 2.66-2.38 (m, 2H), 2.34-2.12 (m, 1H), 2.02-1.88 (m, 1H), 1.87-1.53 (m, 5H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 577.2 [M+H]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N—((R)-1-(3,3-dimethylbutanoyl)pyrrolidin-3-yl)piperidine-3-carboxamide (DX3-128)

To a solution of 30 (20 mg, 0.042 mmol) and Et3N (13 mg, 0.13 mmol) in DCM (1 mL) was added 3,3-dimethylbutanoyl chloride (5.2 mg, 0.042 mmol). The mixture was stirred at room temperature for 3 h and concentrated. The residue was purified with flash chromatography (10% MeOH in DCM) to give DX3-128 as a white solid (18 mg, 75%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.3 Hz, 2H), 7.88 (d, J=8.5 Hz, 2H), 6.31 (dd, J=26.1, 6.9 Hz, 1H), 4.56-4.39 (m, 1H), 3.86-3.69 (m, 1H), 3.69-3.37 (m, 5H), 3.31 (q, J=7.2 Hz, 4H), 2.87-2.75 (m, 1H), 2.72-2.57 (m, 1H), 2.57-2.43 (m, 1H), 2.33-2.12 (m, 3H), 2.03-1.63 (m, 5H), 1.18 (t, J=7.1 Hz, 6H), 1.09 (s, 9H). LC-MS (ESI) m/z 571.2 [M+H]+.


Ethyl (R)-3-((R)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxamido)pyrrolidine-1-carboxylate (DX3-125B)

To a solution of 30 (20 mg, 0.042 mmol) and Et3N (13 mg, 0.13 mmol) in DCM (1 mL) was added ethyl chloroformate (4.6 mg, 0.042 mmol). The mixture was stirred at room temperature for 3 h and concentrated. The residue was purified with flash chromatography (10% MeOH in DCM) to give DX3-125B as a white solid (15 mg, 63%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.6 Hz, 2H), 7.89 (d, J=8.5 Hz, 2H), 6.02 (d, J=7.2 Hz, 1H), 4.46 (dd, J=12.0, 6.0 Hz, 1H), 4.17 (q, J=7.1 Hz, 2H), 3.70 (dd, J=11.4, 6.2 Hz, 1H), 3.62 (d, J=11.6 Hz, 1H), 3.60-3.43 (m, 3H), 3.31 (q, J=7.2 Hz, 5H), 2.86-2.72 (m, 1H), 2.61 (dd, J=13.6, 8.3 Hz, 1H), 2.54-2.38 (m, 1H), 2.28-2.11 (m, 1H), 1.95-1.63 (m, 6H), 1.30 (t, J=7.1 Hz, 3H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 545.2 [M+H]+.


(R)—N—((R)-1-Cyanopyrrolidin-3-yl)-1-((4-(N,N-diethylsulfamoyl)phenyl)sulfonyl)piperidine-3-carboxamide (31)

A solution of 30 (51 mg, 0.11 mmol) in DCM (3 mL) was stirred at 0° C. A solution of NaHCO3 (46 mg, 0.55 mmol) in H2O (1 mL) was added, followed by a solution of cyanogen bromide (14 mg, 0.13 mmol) in DCM (3 mL). The mixture was stirred at 0° C. for 30 min and room temperature for 3 h. The organic layer was separated and washed with sat. NaHCO3, dried over Na2SO4, filtered and concentrated. The residue was purified with flash chromatography (50% EtOAc in DCM) to give 31 as a white solid (41 mg, 75%). 1H NMR (300 MHz, CDCl3) δ 8.00 (d, J=8.1 Hz, 2H), 7.90 (d, J=8.1 Hz, 2H), 6.75 (d, J=7.0 Hz, 1H), 4.47 (s, 1H), 3.75-3.48 (m, 5H), 3.40-3.24 (m, 5H), 2.74 (t, J=10.7 Hz, 1H), 2.63-2.45 (m, 2H), 2.27-2.08 (m, 1H), 2.00-1.76 (m, 3H), 1.73-1.54 (m, 2H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 498.1 [M+H]+.


(R)-1-((4-(N,N-Diethylsulfamoyl)phenyl)sulfonyl)-N—((R)-1-(3-isopropyl-1,2,4-oxadiazol-5-yl)pyrrolidin-3-yl)piperidine-3-carboxamide (DX3-130)

To a solution of 31 (16 mg, 0.032 mmol) and N-hydroxyisobutyrimidamide (4.2 mg, 0.042 mmol) in EtOAc (0.5 mL) and THF (0.5 mL) was added a suspension of ZnCl2 (4.4 mg, 0.032 mmol) in Et2O (0.032 mL) dropwise under argon. The mixture was then stirred at room temperature for 3 h and concentrated. The residue was taken up by EtOH (0.5 mL) and 2M HCl (0.035 mL) was added. The mixture was stirred at 80° C. for 3 h and concentrated. The residue was purified with flash chromatography (10% DCM in MeOH) to give DX3-130 as a white solid (11 mg, 59%). 1H NMR (300 MHz, CDCl3) δ 8.00 (d, J=8.4 Hz, 2H), 7.88 (d, J=8.5 Hz, 2H), 4.68-4.60 (m, 1H), 3.91 (dd, J=11.2, 5.9 Hz, 1H), 3.85-3.70 (m, 2H), 3.59 (dd, J=11.2, 3.9 Hz, 1H), 3.44 (d, J=11.9 Hz, 1H), 3.31 (q, J=7.1 Hz, 5H), 3.07-2.92 (m, 2H), 2.83 (dd, J=11.8, 8.7 Hz, 1H), 2.59-2.49 (m, 1H), 2.35 (dt, J=13.8, 6.5 Hz, 1H), 2.16-2.05 (m, 1H), 1.83-1.63 (m, 4H), 1.33 (d, J=7.0 Hz, 6H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 583.2 [M+H]+.




embedded image


(R)—N,N-Diethyl-4-((3-(3-methyl-1,2,4-oxadiazol-5-yl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-122B)

To a solution of DX2-235 (60 mg, 0.15 mmol) and HATU (87 mg, 0.23 mmol) in DMF (2 mL) was added N-hydroxyacetimidamide (13 mg, 0.18 mmol) and DIEA (58 mg, 0.45 mmol). The mixture was stirred at room temperature for 3 h and then 60° C. overnight. The mixture was diluted with EtOAc, washed with H2O, brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified with preparative HPLC to give DX3-122B as a white solid (11 mg, 17%). 1H NMR (300 MHz, CDCl3) δ 8.00 (d, J=8.2 Hz, 2H), 7.92 (d, J=8.3 Hz, 2H), 4.14-4.02 (m, 1H), 3.78 (d, J=11.9 Hz, 1H), 3.38-3.26 (m, 4H), 3.30-3.18 (m, 1H), 2.80-2.66 (m, 1H), 2.50 (td, J=11.4, 3.1 Hz, 1H), 2.40 (s, 3H), 2.29-2.16 (m, 1H), 2.00-1.71 (m, 2H), 1.71-1.52 (m, 1H), 1.25-1.12 (m, 6H). LC-MS (ESI) m/z 443.0 [M+H]+.




embedded image


(R)—N,N-Diethyl-4-((3-(3-methylisoxazol-5-yl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-123B)

Acetone oxime (11 mg, 0.30 mmol) was dissolved under argon in THF (2 mL), and the solution was cooled to 0° C. Butyllithium (0.12 mL, 0.60 mmol, 2.5 M in hexane) was added dropwise, and the solution was stirred at 0° C. for 2 h. a solution of DX2-235 (50 mg, 0.12 mmol) in THF (2 mL) was added dropwise. The mixture was allowed to warm to room temperature and stirred overnight and concentrated. The residue was re-dissolved in THF (3 mL) and H2O (1 mL) and concentrated H2SO4 (0.04 mL) was added. The resulting mixture was heated at 100° C. for 30 min and diluted with EtOAc, washed with sat. NaHCO3, brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified with flash chromatography (10% MeOH in DCM) to give DX3-123B as a white solid (11 mg, 22%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.2 Hz, 2H), 7.89 (d, J=8.1 Hz, 2H), 5.97 (s, 1H), 3.79 (d, J=11.8 Hz, 1H), 3.62-3.50 (m, 1H), 3.31 (q, J=7.1 Hz, 4H), 3.20-3.08 (m, 1H), 2.69 (dt, J=23.1, 10.8 Hz, 2H), 2.30 (s, 3H), 2.10-1.95 (m, 1H), 1.92-1.60 (m, 3H), 1.17 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 442.0 [M+H]+.




embedded image


(R)—N,N-Diethyl-4-((3-(piperazine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (32)

To a solution of DX3-121 (200 mg, 0.35 mmol) in DCM (4 mL) was added TFA (0.8 mL). The mixture was stirred at room temperature for 2 h and concentrated to give 32 as a colorless gel, which was directly used in the next step without further purification. LC-MS (ESI) m/z 473.1 [M+H]+.


(R)-4-((3-(4-Cyanopiperazine-1-carbonyl)piperidin-1-yl)sulfonyl)-N,N-diethylbenzenesulfonamide (DX3-132B)

Using a similar procedure as described for 31 with 32 (120 mg, 0.25 mmol) and cyanogen bromide (29 mg, 0.28 mmol), white solid (110 mg, 88%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.6 Hz, 1H), 7.88 (d, J=8.7 Hz, 1H), 3.95-3.82 (m, 3H), 3.68 (d, J=19.2 Hz, 5H), 3.31 (q, J=7.2 Hz, 4H), 2.89-2.75 (m, 1H), 2.65-2.50 (m, 1H), 2.38-2.24 (m, 1H), 1.91-1.81 (m, 2H), 1.79-1.63 (m, 1H), 1.56-1.38 (m, 1H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 498.1 [M+H]+.


(R)—N,N-Diethyl-4-((3-(4-(3-methyl-1,2,4-oxadiazol-5-yl)piperazine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-134)

Using a similar procedure as described for DX3-130 with DX3-132B (30 mg, 0.06 mmol) and N-hydroxyacetimidamide (5.8 mg, 0.078 mmol), white solid (23 mg, 69%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.7 Hz, 2H), 7.89 (d, J=8.7 Hz, 2H), 3.89 (d, J=11.6 Hz, 2H), 3.77-3.56 (m, 8H), 3.30 (q, J=7.2 Hz, 4H), 2.88 (t, J=11.3 Hz, 1H), 2.66-2.54 (m, 1H), 2.36-2.28 (m, 1H), 2.26 (s, 3H), 1.94-1.83 (m, 2H), 1.82-1.69 (m, 1H), 1.55-1.41 (m, 1H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 555.1 [M+H]+.


(R)—N,N-Diethyl-4-((3-(4-(3-isopropyl-1,2,4-oxadiazol-5-yl)piperazine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-134B)

Using a similar procedure as described for DX3-130 with DX3-132B (30 mg, 0.06 mmol) and N-hydroxyisobutyrimidamide (8 mg, 0.078 mmol), white solid (23 mg, 66%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.1 Hz, 2H), 7.89 (d, J=8.1 Hz, 2H), 3.89 (d, J=11.9 Hz, 2H), 3.78-3.55 (m, 8H), 3.31 (q, J=7.1 Hz, 4H), 3.01-2.81 (m, 2H), 2.60 (t, J=11.2 Hz, 1H), 2.31 (t, J=11.7 Hz, 1H), 1.89 (d, J=13.4 Hz, 2H), 1.82-1.71 (m, 1H), 1.56-1.41 (m, 1H), 1.31 (d, J=6.9 Hz, 6H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 583.2 [M+H]+.


(R)—N,N-Diethyl-4-((3-(4-(3-(trifluoromethyl)-1,2,4-oxadiazol-5-yl)piperazine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-140)

Using a similar procedure as described for DX3-130 with DX3-132B (30 mg, 0.06 mmol) and 2,2,2-trifluoro-N-hydroxyacetimidamide (10 mg, 0.078 mmol), and purified with prep-HPLC (10-95% CH3CN in H2O with 0.05% TFA, 25 min), white solid (3.4 mg, 9%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=8.3 Hz, 2H), 7.89 (d, J=8.3 Hz, 2H), 3.89 (d, J=11.5 Hz, 2H), 3.73 (d, J=14.7 Hz, 8H), 3.31 (q, J=7.2 Hz, 4H), 2.95-2.82 (m, 1H), 2.61 (t, J=11.3 Hz, 1H), 2.32 (t, J=11.9 Hz, 1H), 1.96-1.85 (m, 2H), 1.78 (d, J=7.0 Hz, 1H), 1.51-1.42 (m, 1H), 1.18 (t, J=7.1 Hz, 6H). LC-MS (ESI) m/z 609.1 [M+H]+.


(R)-4-((3-(4-(1H-Tetrazol-5-yl)piperazine-1-carbonyl)piperidin-1-yl)sulfonyl)-N,N-diethylbenzenesulfonamide (33)

To a suspension of DX3-132B (30 mg, 0.06 mmol) and NaN3 (7.8 mg, 0.12 mmol) in toluene (1 mL) under argon was added Et3N·HCl (16.5 mg, 0.12 mmol) and heated at 80° C. for 5 h. The mixture was then concentrated and EtOAc and H2O was added. The organic layer was separated, washed with brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was directly used in the next step without further purification. LC-MS (ESI) m/z 541.1 [M+H]+.


(R)—N,N-Diethyl-4-((3-(4-(5-isopropyl-1,3,4-oxadiazol-2-yl)piperazine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-146)

To an ice cooled solution of 33 (10.8 mg, 0.02 mmol) and DIEA (5.2 mg, 0.04 mmol) in chlorobenzene (1 mL) was added isobutyric anhydride (4.7 mg, 0.03 mmol) under argon. The mixture was heated at 130° C. for 20 h and concentrated. The residue was purified with prep-HPLC (10-95% CH3CN in H2O with 0.05% TFA, 25 min) to give a white solid (3.2 mg, 27%). 1H NMR (300 MHz, CDCl3) δ 7.99 (d, J=6.1 Hz, 2H), 7.89 (d, J=8.0 Hz, 2H), 3.89 (d, J=11.8 Hz, 2H), 3.81-3.45 (m, 8H), 3.31 (q, J=6.6, 5.5 Hz, 4H), 3.19-2.97 (m, 1H), 2.97-2.82 (m, 1H), 2.60 (t, J=11.6 Hz, 1H), 2.31 (t, J=12.0 Hz, 1H), 1.89 (d, J=13.6 Hz, 2H), 1.81-1.68 (m, 1H), 1.57-1.46 (m, 1H), 1.37 (d, J=6.7 Hz, 6H), 1.19 (t, J=6.8 Hz, 6H). LC-MS (ESI) m/z 583.2 [M+H]+.




embedded image


(R)-(1-((4-(Benzylthio)phenyl)sulfonyl)piperidin-3-yl)(4-(3-isopropyl-1,2,4-oxadiazol-5-yl)piperazin-1-yl)methanone (34)

To a solution of 69b (583 mg, 1.39 mmol) in THF (5 mL) and H2O (5 mL) was added LiOH·H2O (290 mg, 6.94 mmol) at 0° C. and stirred at room temperature for 5 h. The mixture was then diluted with H2O, and the pH was adjusted to 3 by 1N HCl solution. It was extracted with EtOAc, and the organic layer was washed with brine, dried over anhydrous Na2SO4, filtered and concentrated to give (R)-1-((4-(benzylthio)phenyl)sulfonyl)piperidine-3-carboxylic acid as a white solid (480 mg, 88%), which was directly used in the next step. To a solution of (R)-1-((4-(benzylthio)phenyl)sulfonyl)piperidine-3-carboxylic acid as a white solid (300 mg, 0.77 mmol) and HATU (439 mg, 1.16 mmol) in DMF (8 mL) was added 3-isopropyl-5-(piperazin-1-yl)-1,2,4-oxadiazole (150 mg, 0.77 mmol) and DIEA (298 mg, 2.31 mmol). The mixture was stirred at room temperature overnight. The mixture was then diluted with EtOAc, washed with brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified with flash chromatography (10% MeOH in DCM) to give 34 as a white solid (343 mg, 78%). 1H NMR (300 MHz, CDCl3) δ 7.62 (d, J=8.2 Hz, 2H), 7.44-7.28 (m, 7H), 4.24 (s, 2H), 3.83 (d, J=11.7 Hz, 2H), 3.75-3.56 (m, 8H), 2.99-2.81 (m, 2H), 2.49 (t, J=11.3 Hz, 1H), 2.23 (t, J=11.5 Hz, 1H), 1.93-1.64 (m, 3H), 1.56-1.41 (m, 1H), 1.31 (d, J=6.9 Hz, 6H). LC-MS (ESI) m/z 570.3 [M+H]+.


(R)-4-((3-(4-(3-Isopropyl-1,2,4-oxadiazol-5-yl)piperazine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonyl chloride (35)

Using a similar procedure as described for 4a with 34 (100 mg, 0.18 mmol), white solid (98 mg, 100% crude). LC-MS (ESI) m/z 546.3, 548.1 [M+H]+.


(R)—N-ethyl-4-((3-(4-(3-isopropyl-1,2,4-oxadiazol-5-yl)piperazine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-167)

Using a similar procedure as described for DX2-201 with 35 (20 mg, 0.037 mmol) and ethylamine (5.0 mg, 0.11 mmol), white solid (6 mg, 29%). 1H NMR (300 MHz, CDCl3) δ 8.04 (d, J=8.1 Hz, 2H), 7.91 (d, J=8.1 Hz, 2H), 4.68-4.56 (m, 1H), 3.94-3.83 (m, 2H), 3.78-3.61 (m, 7H), 3.17-3.03 (m, 2H), 3.02-2.83 (m, 2H), 2.60 (t, J=11.2 Hz, 1H), 2.32 (t, J=11.8 Hz, 1H), 1.95-1.68 (m, 3H), 1.55-1.43 (m, 1H), 1.36-1.26 (m, 6H), 1.18 (t, J=7.2 Hz, 3H). LC-MS (ESI) m/z 555.2 [M+H]+. Purity: 96.3%.


(R)—N-Isopropyl-4-((3-(4-(3-isopropyl-1,2,4-oxadiazol-5-yl)piperazine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-166B)

Using a similar procedure as described for DX2-201 with 35 (20 mg, 0.037 mmol) and isopropylamine (6.5 mg, 0.11 mmol), white solid (4 mg, 19%). 1H NMR (300 MHz, CDCl3) δ 8.06 (d, J=8.5 Hz, 2H), 7.91 (d, J=8.5 Hz, 2H), 4.52 (d, J=7.7 Hz, 1H), 3.89 (d, J=11.7 Hz, 2H), 3.82-3.49 (m, 9H), 3.03-2.81 (m, 2H), 2.60 (t, J=11.3 Hz, 1H), 2.39-2.28 (m, 1H), 1.95-1.82 (m, 2H), 1.81-1.69 (m, 1H), 1.57-1.43 (m, 1H), 1.32 (d, J=7.0 Hz, 6H), 1.14 (d, J=6.5 Hz, 6H). LC-MS (ESI) m/z 569.2 [M+H]+. Purity: 95.6%.


(R)—N-Cyclopropyl-4-((3-(4-(3-isopropyl-1,2,4-oxadiazol-5-yl)piperazine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-179B)

Using a similar procedure as described for DX2-201 with 35 (20 mg, 0.037 mmol) and cyclopropanamine (6.3 mg, 0.11 mmol), white solid (5 mg, 24%). 1H NMR (300 MHz, CDCl3) δ 8.09 (d, J=8.6 Hz, 2H), 7.93 (d, J=8.7 Hz, 2H), 5.11 (s, 1H), 3.90 (d, J=11.6 Hz, 2H), 3.78-3.57 (m, 8H), 3.01-2.82 (m, 2H), 2.68-2.57 (m, 1H), 2.39-2.24 (m, 2H), 1.89 (d, J=13.3 Hz, 2H), 1.82-1.66 (m, 1H), 1.58-1.43 (m, 1H), 1.31 (d, J=6.9 Hz, 6H), 0.74-0.62 (m, 4H). LC-MS (ESI) m/z 567.2 [M+H]+. Purity: 95.2%.


(R)—N-Cyclobutyl-4-((3-(4-(3-isopropyl-1,2,4-oxadiazol-5-yl)piperazine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-167B)

Using a similar procedure as described for DX2-201 with 35 (20 mg, 0.037 mmol) and cyclobutanamine (7.8 mg, 0.11 mmol), white solid (3.4 mg, 16%). 1H NMR (300 MHz, CDCl3) (8.03 (d, J=8.3 Hz, 2H), 7.90 (d, J=8.3 Hz, 2H), 4.84 (d, J=8.6 Hz, 1H), 3.99-3.80 (m, 3H), 3.79-3.59 (m, 8H), 3.02-2.83 (m, 2H), 2.59 (t, J=11.3 Hz, 1H), 2.34-2.15 (m, 3H), 1.97-1.74 (m, 5H), 1.75-1.59 (m, 2H), 1.57-1.42 (m, 1H), 1.31 (d, J=6.9 Hz, 6H). LC-MS (ESI) m/z 581.2 [M+H]+. Purity: 96.5%.


(R)—N-(tert-Butyl)-4-((3-(4-(3-isopropyl-1,2,4-oxadiazol-5-yl)piperazine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonamide (DX3-178)

Using a similar procedure as described for DX2-201 with 35 (20 mg, 0.037 mmol) and 2-methylpropan-2-amine (8.0 mg, 0.11 mmol), white solid (8 mg, 37%). 1H NMR (300 MHz, CDCl3) δ 8.07 (d, J=8.3 Hz, 2H), 7.89 (d, J=8.3 Hz, 2H), 4.64 (s, 1H), 3.90 (d, J=11.6 Hz, 2H), 3.78-3.59 (m, 8H), 3.03-2.82 (m, 2H), 2.61-2.56 (m, 1H), 2.31 (t, J=11.7 Hz, 1H), 1.94-1.70 (m, 3H), 1.57-1.45 (m, 1H), 1.36-1.24 (m, 15H). LC-MS (ESI) m/z 583.2 [M+H]+. Purity: 95.3%.




embedded image


(R)-(1-((4-(Benzylthio)phenyl)sulfonyl)piperidin-3-yl)(4,4-difluoropiperidin-1-yl)methanone (36)

To a solution of 3b (139 mg, 0.33 mmol) in THF (2 mL) and H2O (2 mL) was added LiOH·H2O (69 mg, 1.65 mmol) at 0° C. and stirred at room temperature for 5 h. The mixture was then diluted with H2O, and the pH was adjusted to 3 by TN HCl solution. It was extracted with EtOAc, and the organic layer was washed with brine, dried over anhydrous Na2SO4, filtered and concentrated to give the corresponding carboxylic acid. It was directly dissolved in DMF (5 mL). Then HATU (190 mg, 0.50 mmol), 4,4-difluoropiperidine (40 mg, 0.33 mmol) and DIEA (129 mg, 1.0 mmol) was added subsequently. The mixture was stirred at room temperature overnight. The mixture was then diluted with EtOAc, washed with brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified with flash chromatography (20% EtOAc in hexane) to give 36 as a light yellow gel (145 mg, 90%).


(R)-4-((3-(4,4-Difluoropiperidine-1-carbonyl)piperidin-1-yl)sulfonyl)benzenesulfonyl chloride (37)

Using a similar procedure as described for 4a with 36 (50 mg, 0.10 mmol), colorless gel (47 mg, 100% crude). 1H NMR (300 MHz, CDCl3) δ 8.21 (d, J=8.6 Hz, 2H), 8.00 (d, J=8.5 Hz, 2H), 3.88 (d, J=11.2 Hz, 2H), 3.77-3.55 (m, 4H), 2.95-2.83 (m, 1H), 2.59 (t, J=11.3 Hz, 1H), 2.33 (td, J=11.9, 2.8 Hz, 1H), 2.05-1.68 (m, 8H).


(R)-4-((3-(4,4-Difluoropiperidine-1-carbonyl)piperidin-1-yl)sulfonyl)-N-isopropylbenzenesulfonamide (DX3-195B)

To a solution of isopropylamine (3 mg, 0.05 mmol) and triethylamine (15 mg, 0.15 mmol) was added 37 (24 mg, 0.05 mmol) portionwise. The mixture was stirred at room temperature overnight. The mixture was concentrated and purified with flash chromatography (10% MeOH in DCM) to give DX3-195B as a white solid (10 mg, 37%). 1H NMR (300 MHz, CDCl3) δ 8.05 (d, J=8.4 Hz, 2H), 7.90 (d, J=8.3 Hz, 2H), 4.67 (d, J=7.7 Hz, 1H), 3.88 (d, J=11.6 Hz, 2H), 3.77-3.49 (m, 5H), 2.96-2.82 (m, 1H), 2.58 (t, J=11.3 Hz, 1H), 2.37-2.22 (m, 1H), 2.09-1.82 (m, 6H), 1.80-1.65 (m, 1H), 1.56-1.42 (m, 1H), 1.13 (d, J=6.5 Hz, 6H). LC-MS (ESI) m/z 494.2 [M+H]+.


(R)-4-((3-(4,4-Difluoropiperidine-1-carbonyl)piperidin-1-yl)sulfonyl)-N,N-dimethylbenzenesulfonamide (DX3-201B)

Using a similar procedure as described for DX3-195B with 37 (50 mg, 0.10 mmol) and dimethyl amine (2.3 mg, 0.05 mmol), white solid (14 mg, 58%). 1H NMR (300 MHz, CDCl3) δ 7.95 (s, 4H), 3.90 (d, J=11.8 Hz, 2H), 3.78-3.58 (m, 4H), 2.89 (t, J=11.3 Hz, 1H), 2.79 (s, 6H), 2.60 (t, J=11.3 Hz, 1H), 2.38-2.27 (m, 1H), 2.10-1.83 (m, 6H), 1.80-1.69 (m, 1H), 1.55-1.43 (m, 1H). LC-MS (ESI) m/z 502.1 [M+Na]+.




embedded image


Ethyl (R)-1-((4-(pentan-3-ylthio)phenyl)sulfonyl)piperidine-3-carboxylate (38)

Using a similar procedure as described for 3a with 2b (80 mg, 0.21 mmol) and pentane-3-thiol (22 mg, 0.21 mmol), light yellow solid (78 mg, 92%). LC-MS (ESI) m/z 400.0 [M+H]+.


Ethyl (R)-1-((4-(pentan-3-ylsulfonyl)phenyl)sulfonyl)piperidine-3-carboxylate (39)

To a solution of 38 (44 mg, 0.11 mmol) in DCM (2 mL) was added mCPBA (76 mg, 0.44 mmol) and stirred at room temperature overnight. Sat. NaHCO3 was added and the mixture was extracted with EtOAc. The organic layer was washed with brine, dried with Na2SO4, filtered and concentrated. The residue was purified with silica chromatography (50% EtOAc in hexane) to give 39 as a white solid (20 mg, 42%). 1H NMR (300 MHz, CDCl3) δ 8.08 (d, J=8.3 Hz, 2H), 7.97 (d, J=8.4 Hz, 2H), 4.17 (q, J=7.1 Hz, 2H), 3.95-3.80 (m, 1H), 3.66 (d, J=11.7 Hz, 1H), 2.89 (ddd, J=11.9, 7.2, 4.9 Hz, 1H), 2.71-2.58 (m, 2H), 2.46 (td, J=11.3, 3.2 Hz, 1H), 2.04 (d, J=13.4 Hz, 1H), 1.87 (ddd, J=15.6, 7.7, 4.9 Hz, 3H), 1.72 (dq, J=20.9, 6.9, 6.4 Hz, 4H), 1.53-1.39 (m, 1H), 1.28 (t, J=7.1 Hz, 3H), 1.05 (t, J=7.5 Hz, 6H). LC-MS (ESI) m/z 432.1 [M+H]+.


(R)-(4-(3-Isopropyl-1,2,4-oxadiazol-5-yl)piperazin-1-yl)(1-((4-(pentan-3-ylsulfonyl)phenyl)sulfonyl)piperidin-3-yl)methanone (DX3-210)

Using a similar procedure as described for 34 with 39 (15 mg, 0.037 mmol) and 3-isopropyl-5-(piperazin-1-yl)-1,2,4-oxadiazole (7.3 mg, 0.037 mmol), white solid (12 mg, 55%). 1H NMR (300 MHz, CDCl3) δ 8.07 (d, J=8.0 Hz, 2H), 7.95 (d, J=8.1 Hz, 2H), 3.90 (d, J=11.8 Hz, 2H), 3.78-3.55 (m, 8H), 2.99-2.81 (m, 3H), 2.62 (t, J=11.3 Hz, 1H), 2.32 (t, J=11.6 Hz, 1H), 1.98-1.68 (m, 7H), 1.58-1.42 (m, 1H), 1.31 (d, J=6.9 Hz, 6H), 1.05 (t, J=7.5 Hz, 6H). LC-MS (ESI) m/z 582.3 [M+H]+.


(R)-Morpholino(1-((4-(pentan-3-ylsulfonyl)phenyl)sulfonyl)piperidin-3-yl)methanone (DX3-209B)

Using a similar procedure as described for 34 with 39 (15 mg, 0.037 mmol) and morpholine (3.2 mg, 0.037 mmol), white solid (10 mg, 57%). 1H NMR (300 MHz, CDCl3) δ 8.07 (d, J=8.5 Hz, 2H), 7.95 (d, J=8.3 Hz, 2H), 3.89 (d, J=11.5 Hz, 2H), 3.80-3.45 (m, 8H), 2.94-2.77 (m, 2H), 2.60 (t, J=11.4 Hz, 1H), 2.31 (td, J=12.0, 2.8 Hz, 1H), 1.96-1.82 (m, 4H), 1.82-1.65 (m, 3H), 1.49 (td, J=12.2, 3.4 Hz, 1H), 1.05 (t, J=7.5 Hz, 6H).




embedded image


(R)-(1-((4-Bromophenyl)sulfonyl)piperidin-3-yl)(4,4-difluoropiperidin-1-yl)methanone (40)

Using a similar procedure as described for 3a from 40 (188 mg, 0.50 mmol) and (4,4-difluoropiperidine (67 mg, 0.55 mmol), white solid (205 mg, 91%). 1H NMR (300 MHz, CDCl3) (7.70 (d, J=8.3 Hz, 2H), 7.63 (d, J=8.4 Hz, 2H), 3.84 (d, J=11.6 Hz, 2H), 3.77-3.57 (m, 4H), 2.88 (t, J=11.6 Hz, 1H), 2.50 (t, J=11.3 Hz, 1H), 2.25 (t, J=11.8 Hz, 1H), 2.15-1.79 (m, 6H), 1.79-1.66 (m, 1H), 1.54-1.39 (m, 1H).


(R)-(4,4-Difluoropiperidin-1-yl)(1-((4-(isobutylthio)phenyl)sulfonyl)piperidin-3-yl)methanone (41a)

Using a similar procedure as described for 3a from 40 (50 mg, 0.11 mmol) and 2-methylpropane-1-thiol (10 mg, 0.11 mmol), white solid (43 mg, 85%). 1H NMR (300 MHz, CDCl3) δ 7.63 (d, J=8.2 Hz, 2H), 7.36 (d, J=8.2 Hz, 2H), 3.83 (d, J=11.7 Hz, 2H), 3.78-3.58 (m, 4H), 2.89 (d, J=6.9 Hz, 3H), 2.48 (t, J=11.4 Hz, 1H), 2.24 (t, J=11.7 Hz, 1H), 2.12-1.89 (m, 5H), 1.90-1.66 (m, 3H), 1.53-1.38 (m, 1H), 1.10 (d, J=6.7 Hz, 6H). LC-MS (ESI) m/z 461.0 [M+H]+.


(R)-(1-((4-(Cyclopropylthio)phenyl)sulfonyl)piperidin-3-yl)(4,4-difluoropiperidin-1-yl)methanone (41b)

Using a similar procedure as described for 3a from 40 (40 mg, 0.089 mmol) and 0cyclopropanethiol (6.6 mg, 0.089 mmol), white solid (26 mg, 65%). 1H NMR (300 MHz, CDCl3) δ 7.63 (d, J=7.0 Hz, 2H), 7.47 (d, J=8.3 Hz, 2H), 3.89-3.76 (m, 2H), 3.77-3.56 (m, 4H), 2.88 (t, J=11.6 Hz, 1H), 2.47 (t, J=11.1 Hz, 1H), 2.28-2.17 (m, 2H), 2.10-1.88 (m, 4H), 1.89-1.67 (m, 3H), 1.52-1.34 (m, 1H), 1.18 (d, J=6.9 Hz, 2H), 0.80-0.70 (m, 2H).


(R)-(1-((4-(Cyclobutylthio)phenyl)sulfonyl)piperidin-3-yl)(4,4-difluoropiperidin-1-yl)methanone (41c)

Using a similar procedure as described for 3a from 40 (40 mg, 0.089 mmol) and cyclobutanethiol (7.8 mg, 0.089 mmol), white solid (36 mg, 88%). 1H NMR (300 MHz, CDCl3) δ 7.59 (d, J=8.5 Hz, 2H), 7.24 (d, J=8.5 Hz, 2H), 4.05-3.94 (m, 1H), 3.86-3.75 (m, 2H), 3.74-3.58 (m, 4H), 2.93-2.78 (m, 1H), 2.64-2.51 (m, 1H), 2.45 (t, J=11.3 Hz, 1H), 2.26-1.92 (m, 10H), 1.87-1.64 (m, 3H), 1.48-1.37 (m, 1H).


(R)-(4,4-Difluoropiperidin-1-yl)(1-((4-(oxetan-3-ylthio)phenyl)sulfonyl)piperidin-3-yl)methanone (41d)

Using a similar procedure as described for 3a from 40 (40 mg, 0.089 mmol) and oxetane-3-thiol (8.0 mg, 0.089 mmol), white solid (16 mg, 39%). 1H NMR (300 MHz, CDCl3) δ 7.64 (d, J=8.4 Hz, 2H), 7.20 (d, J=8.4 Hz, 2H), 5.15 (t, J=6.6 Hz, 2H), 4.69 (t, J=6.3 Hz, 2H), 4.65-4.58 (m, 1H), 3.90-3.58 (m, 6H), 2.94-2.83 (m, 1H), 2.47 (t, J=11.3 Hz, 1H), 2.22 (td, J=11.8, 2.7 Hz, 1H), 2.12-1.89 (m, 4H), 1.88-1.66 (m, 3H), 1.50-1.35 (m, 1H).


(R)-(4,4-Difluoropiperidin-1-yl)(1-((4-(isobutylsulfonyl)phenyl)sulfonyl)piperidin-3-yl)methanone (DX3-218)

To a solution of 41a (21 mg, 0.046 mmol) in DCM (3 mL) was added mCPBA (32 mg, 0.182 mmol) and stirred at room temperature overnight. Sat. NaHCO3 was added and the mixture was extracted with EtOAc. The organic layer was washed with brine, dried with Na2SO4, filtered and concentrated. The residue was purified with silica chromatography (50% EtOAc in hexane) to give DX3-218 (8 mg, 35%) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.10 (d, J=8.2 Hz, 2H), 7.96 (d, J=8.4 Hz, 2H), 3.89 (d, J=11.8 Hz, 2H), 3.79-3.59 (m, 4H), 3.05 (d, J=6.5 Hz, 2H), 2.89 (t, J=11.4 Hz, 1H), 2.58 (t, J=11.4 Hz, 1H), 2.40-2.24 (m, 2H), 2.16-1.82 (m, 6H), 1.81-1.66 (m, 1H), 1.56-1.41 (m, 1H), 1.12 (d, J=6.7 Hz, 6H). LC-MS (ESI) m/z 491.3 [M−H]. Purity: 96.4%.


(R)-(1-((4-(Cyclopropylsulfonyl)phenyl)sulfonyl)piperidin-3-yl)(4,4-difluoropiperidin-1-yl)methanone (DX3-221)

Using a similar procedure as described for DX3-218 from 41b (26 mg, 0.058 mmol), white solid (18 mg, 64%). 1H NMR (300 MHz, CDCl3) δ 8.09 (d, J=8.5 Hz, 2H), 7.95 (d, J=8.3 Hz, 2H), 3.99-3.83 (m, 2H), 3.80-3.57 (m, 4H), 2.90 (t, J=11.5 Hz, 1H), 2.66-2.46 (m, 2H), 2.39-2.24 (m, 1H), 2.14-1.82 (m, 6H), 1.73 (t, J=13.0 Hz, 1H), 1.56-1.37 (m, 3H), 1.20-1.08 (m, 2H).


(R)-(1-((4-(Cyclobutylsulfonyl)phenyl)sulfonyl)piperidin-3-yl)(4,4-difluoropiperidin-1-yl)methanone (DX3-220B)

Using a similar procedure as described for DX3-218 from 41c (18 mg, 0.039 mmol), white solid (13 mg, 68%). 1H NMR (300 MHz, CDCl3) δ 8.06 (d, J=8.3 Hz, 2H), 7.94 (d, J=8.4 Hz, 2H), 3.96-3.80 (m, 3H), 3.77-3.55 (m, 4H), 2.89 (t, J=11.6 Hz, 1H), 2.74-2.51 (m, 3H), 2.35-2.19 (m, 3H), 2.10-1.86 (m, 8H), 1.80-1.69 (m, 1H), 1.52-1.40 (m, 1H).


(R)-(4,4-Difluoropiperidin-1-yl)(1-((4-(oxetan-3-ylsulfonyl)phenyl)sulfonyl)piperidin-3-yl)methanone (DX3-219B)

Using a similar procedure as described for DX3-218 from 41d (16 mg, 0.035 mmol), white solid (10 mg, 59%). 1H NMR (300 MHz, CDCl3) δ 8.09 (d, J=8.2 Hz, 2H), 7.98 (d, J=8.4 Hz, 2H), 5.07-4.97 (m, 2H), 4.87 (td, J=7.7, 3.5 Hz, 2H), 4.52 (tt, J=7.9, 6.1 Hz, 1H), 3.95-3.82 (m, 2H), 3.79-3.58 (m, 4H), 2.89 (t, J=11.5 Hz, 1H), 2.59 (t, J=11.3 Hz, 1H), 2.32 (td, J=11.9, 2.7 Hz, 1H), 2.14-1.81 (m, 6H), 1.81-1.62 (m, 1H), 1.53-1.38 (m, 1H). LC-MS (ESI) m/z 493.1 [M+H]+. Purity: 98.4%




embedded image


(R)-1-((4-(Diethylcarbamoyl)phenyl)sulfonyl)piperidine-3-carboxylic acid (43)

To a solution of 42 (100 mg, 0.36 mmol) and Na2CO3 (114 mg, 1.08 mmol) in H2O (1.5 mL) was added a solution of (R)-piperidine-3-carboxylic acid (47 mg, 0.36 mmol) in THF (1.5 mL) dropwise at 0° C. and stirred at room temperature for 3 h. THF was removed by evaporation and the pH was adjusted to 3 by 1N HCl solution. It was extracted with EtOAc, and the organic layer was washed with brine, dried over anhydrous Na2SO4, filtered and concentrated to give 43 as a white solid (130 mg, 98%). 1H NMR (300 MHz, CDCl3) δ 7.83 (d, J=8.0 Hz, 2H), 7.55 (d, J=7.9 Hz, 2H), 3.85 (d, J=11.1 Hz, 1H), 3.67-3.51 (m, 3H), 3.24 (q, J=6.7 Hz, 2H), 2.73-2.49 (m, 2H), 2.49-2.36 (m, 1H), 2.01 (d, J=13.5 Hz, 1H), 1.88-1.76 (m, 1H), 1.68 (q, J=13.9, 12.4 Hz, 1H), 1.47-1.35 (m, 1H), 1.29 (t, J=7.1 Hz, 3H), 1.14 (t, J=6.9 Hz, 3H).


(R)-4-((3-(4,4-difluoropiperidine-1-carbonyl)piperidin-1-yl)sulfonyl)-N,N-diethylbenzamide (DX3-203)

To a solution of 43 (25 mg, 0.068 mmol) and HATU (39 mg, 0.102 mmol) in DMF (1 mL) was added 4,4-difluoropiperidine (8.2 mg, 0.068 mmol) and DIEA (26 mg, 0.204 mmol). The mixture was stirred at room temperature overnight. The mixture was then diluted with EtOAc, washed with brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified with flash chromatography (10% MeOH in DCM) to give DX3-203 as a white solid (18 mg, 32%). 1H NMR (300 MHz, CDCl3) δ 7.81 (d, J=8.1 Hz, 2H), 7.54 (d, J=8.2 Hz, 2H), 3.93-3.81 (m, 2H), 3.78-3.52 (m, 6H), 3.30-3.16 (m, 2H), 2.86 (tt, J=11.2, 3.4 Hz, 1H), 2.56 (t, J=11.4 Hz, 1H), 2.29 (td, J=11.9, 2.8 Hz, 1H), 2.13-1.79 (m, 6H), 1.78-1.64 (m, 1H), 1.55-1.37 (m, 1H), 1.28 (t, J=7.1 Hz, 3H), 1.14 (t, J=7.1 Hz, 3H).




embedded image


Ethyl (R)-1-((4-(dimethylphosphoryl)phenyl)sulfonyl)piperidine-3-carboxylate (44a)

To a suspension of 2b (60 mg, 0.16 mmol), dimethylphosphine oxide (14 mg, 0.18 mmol) and K3PO4 (0.19 mmol) in DMF (4 mL) was added Pd(OAc)2 (1.8 mg, 0.008 mmol) and XantPhos (5.6 mg, 0.0096 mmol) under argon. The mixture was heated at 120° C. under argon for 5 h. The mixture was then partitioned between EtOAc and water, and the organic layer was separated, washed with brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified with silica chromatography (DCM:MeOH=10:1) to give 44a as a white solid (51 mg, 88%). 1H NMR (300 MHz, CDCl3) δ 7.86-7.66 (m, 4H), 3.97 (q, J=7.1 Hz, 2H), 3.64 (d, J=7.4 Hz, 1H), 3.43 (dt, J=11.8, 4.1 Hz, 1H), 2.53-2.36 (m, 2H), 2.27 (td, J=11.2, 3.0 Hz, 1H), 1.64 (d, J=13.1 Hz, 7H), 1.58-1.37 (m, 1H), 1.35-1.16 (m, 1H), 1.09 (t, J=7.1 Hz, 3H). LC-MS (ESI) m/z 396.0 [M+Na]+.


Ethyl (R)-1-((4-(diethylphosphoryl)phenyl)sulfonyl)piperidine-3-carboxylate (44b)

Using a similar procedure as described for 44a from 2b (80 mg, 0.21 mmol) and diethylphosphine oxide (25 mg, 0.23 mmol), white solid (80 mg, 95%). 1H NMR (300 MHz, CDCl3) δ 7.88 (d, J=6.2 Hz, 4H), 4.13 (q, J=7.1 Hz, 2H), 3.85 (d, J=8.5 Hz, 1H), 3.70-3.58 (m, 1H), 2.67-2.53 (m, 2H), 2.41 (td, J=11.4, 3.1 Hz, 1H), 2.12-1.85 (m, 5H), 1.89-1.75 (m, 1H), 1.75-1.56 (m, 1H), 1.50-1.34 (m, 1H), 1.25 (t, J=7.1 Hz, 3H), 1.21-1.05 (m, 6H). LC-MS (ESI) m/z 424.1 [M+Na]+.


(R)-(4,4-Difluoropiperidin-1-yl)(1-((4-(dimethylphosphoryl)phenyl)sulfonyl)piperidin-3-yl)methanone (DX3-214)

Using a similar procedure as described for 34 from 44a (19 mg, 0.052 mmol) and 4,4-difluoropiperidine (7.6 mg, 0.063 mmol), white solid (19 mg, 83%). 1H NMR (300 MHz, CDCl3) δ 7.99-7.86 (m, 4H), 3.87 (t, J=11.3 Hz, 2H), 3.80-3.59 (m, 4H), 2.90 (t, J=11.7 Hz, 1H), 2.53 (t, J=11.3 Hz, 1H), 2.29 (td, J=11.8, 2.7 Hz, 1H), 2.14-1.90 (m, 4H), 1.89-1.72 (m, 9H), 1.51-1.40 (m, 1H). LC-MS (ESI) m/z 470.9 [M+Na]+. Purity: 97.3%.


(R)-(1-((4-(diethylphosphoryl)phenyl)sulfonyl)piperidin-3-yl)(4,4-difluoropiperidin-1-yl)methanone (DX3-216)

Using a similar procedure as described for 34 from 44b (16 mg, 0.040 mmol) and 4,4-difluoropiperidine (5.8 mg, 0.048 mmol), white solid (18 mg, 95%). 1H NMR (300 MHz, CDCl3) δ 7.89 (d, J=6.2 Hz, 4H), 3.96-3.83 (m, 2H), 3.78-3.60 (m, 4H), 3.51-3.42 (m, 1H), 2.96-2.83 (m, 1H), 2.56 (t, J=11.3 Hz, 1H), 2.30 (t, J=11.8 Hz, 1H), 2.18-1.87 (m, 9H), 1.79-1.70 (m, 1H), 1.55-1.41 (m, 1H), 1.26-1.06 (m, 6H). LC-MS (ESI) m/z 499.0 [M+Na]+. Purity: 99.1%.


Example II
MTT Assay for the Evaluation of Cancer Cell Growth Inhibition

Measurement of cancer cell growth inhibition of the compounds of invention Cytotoxicity of the compounds was assessed by MTT assay. In brief, MIA PaCa-2 cells or BxPc3 cells were seeded in 96-well microtitre plates at density of 200 cells/well. After overnight attachment, cells were treated with compounds at indicated concentration. After 7 days of treatment, MTT solution (3 mg/ml, 20 μl) was added to each well and cells were incubated for 3 h at 37° C. After incubation, media from each well was removed and the dark blue formazan crystals formed by live cells were dissolved in DMSO (100 ml per well). The absorbance intensity was measured at 570 nm on a microplate reader (Molecular Devices, Sunnyvale, Calif., USA). The half maximal inhibitory concentration (IC50) value were calculated using 4 or 6 dots curve plotted with Cumulative Gaussian equation or Log(inhibitor) vs. response equation in GraphPad Prism. The results are listed in Table 2.













TABLE 2








MiaPaCa
BxPC-3



Compound
IC50 (μM)
IC50 (μM)









DX2-201
0.37 ± 0.06
0.63 ± 0.24



DX2-202
6.08 ± 0.76
6.8 ± 2.1



DX2-225
12.09 ± 3.85 
13.35 ± 4.58 



DX2-230
3.92 ± 0.89
3.88 ± 0.57



DX2-242
2.34 ± 1.01
13.82 ± 3.58 



DX2-263
 0.3 ± 0.13
0.35 ± 0.06



DX2-265
2.87 ± 0.49
3.29 ± 0.74



DX2-275
0.93 ± 0.36
1.61 ± 0.37



DX2-293
 1.3 ± 0.45
1.37 ± 0.52



DX3-3
4.12 ± 2.44
2.29 ± 0.7 



DX3-14B-P1
1.35 ± 0.59
3.29 ± 0.06



DX3-39
 5.1 ± 1.29
 6.3 ± 1.44



DX3-44
 4.7 ± 1.92
6.77 ± 0.52



DX3-47
2.94 ± 0.32
4.92 ± 1.94



DX3-49B
3.65 ± 1.68
8.27 ± 2.52



DX3-78B
0.28
0.62



DX3-90
2.55 ± 0.15
3.59 ± 1.39



DX3-107B
0.75 ± 0.08
2.27 ± 0.61



DX3-115
1.04 ± 0.45
1.23 ± 0.79



DX3-119
0.13 ± 0.04
0.33 ± 0.12



DX3-120
0.43 ± 0.18
 1.1 ± 0.16



DX3-122B
4.12 ± 1.16
2.86 ± 3.14



DX3-121B
0.35 ± 0.02
0.79 ± 40.1



DX3-125
0.94
3.72










Example III

Nascent RNA sequencing (Bru-seq) results showed genes in metabolism pathways clustered after 4 hours of DX2-201 treatment. The glycolysis pathway was dramatically upregulated and the oxidative phosphorylation was significantly downregulated. Consistent with this observation, there are also massive metabolite pathways influenced including starch and sucrose metabolism, glycolysis and gluconeogenesis, purine metabolism, pyrimidine metabolism, N-glycan biosynthesis, etc. The overall metabolism change suggested the suppression on oxidative phosphorylation and upregulation in glycolysis upon DX2-201 treatment, subsequently causing the imbalanced ATP and biomass production. As a result of that, DNA repair and mismatch repair gene sets were downregulated, which might be related to the shortage of biomass. In addition, myc target gene sets and E2F target gene set were downregulated, reflecting DX2-201's inhibition on cell growth.


Several other disease related gene sets were also significantly changed, including Parkinson's disease, Huntington's disease and Alzheimer's diseases, which might be also related to the metabolism change, suggesting these compounds' potential to treat certain neurodegenerative diseases.


Proteomic results also showed TCA cycle suppressed while hypoxia pathways, bile acid metabolism, cholesterol homoeostasis and glycolysis upregulated after DX2-201 treatment. Several proteins participating in energy metabolism were dramatically changed. Glycerol kinase is the top upregulated gene with 3-fold change in 24 hours indicating the upregulated usage of glycerol as alternative energy source. Serine protease 23, nuclear receptor family 2 group F member 6, protein NDRG1 were all upregulated. Interestingly, an important enzyme involved in TCA cycle, dihydrolilpoyl dehudrogenase, was downregulated with a fold change of 0.684, suggesting the TCA cycle function was impaired by DX2-201 treatment.









TABLE 3







Top 25 protein-coding genes downregulated in UM16 cell line


treated with DX2-201 (5 μM, 4 hours, Bru-seq)










Fold



Genes
change
Comments












ARRDC4
0.431505
arrestin domain containing 4


SCARNA5
0.43758
small Cajal body-specific RNA 5; small




Cajal body-specific RNA 6


INTS5
0.45152
integrator complex subunit 5


G0S2
0.466649
G0/G1switch 2


HLF
0.472697
Hepatic Leukemia Factor


POLR2J4
0.477451
polymerase (RNA) II (DNA directed)




polypeptide J4, pseudogene


TXNIP
0.499674
Thioredoxin Interacting Protein


DGAT2
0.512885
diacylglycerol O-acyltransferase homolog 2


SNHG21
0.549732
Small Nucleolar RNA Host Gene 21


FAM200A
0.55948
chromosome 7 open reading frame 38


RRS1
0.568556
Ribosome biogenesis regulatory




protein homolog


PCED1A
0.576043
PC-esterase domain containing 1A


A2ML1
0.582029
alpha-2-macroglobulin like 1


PLLP
0.584318
plasma membrane proteolipid (plasmolipin)


FAM86HP
0.594645
family with sequence similarity 86 member H


SNORA62
0.596836
small nucleolar RNA, H/ACA box 62; small




nucleolar RNA, H/ACA box 6


GGCT
0.601165
gamma-glutamyl cyclotransferase


SNORA71C
0.602279
small nucleolar RNA, H/ACA box 71C


DPAGT1
0.603372
dolichyl-phosphate (UDP-N-




acetylglucosamine) N-




acetylglucosaminephosphotransferase




1 (GlcNAc-1-P transferase)


BDKRB2
0.604389
Bradykinin Receptor B2


FAM86C1
0.605286
Family With Sequence Similarity 86




Member C1


FOXA2
0.607488
Forkhead Box A2


TMED1
0.623178
Transmembrane P24 Trafficking Protein 1


SLC5A6
0.623446
Solute Carrier Family 5 Member 6


L3HYPDH
0.630135
Trans-L-3-Hydroxyproline Dehydratase
















TABLE 4







Top 25 protein-coding genes upregulated in UM16 cell line


treated with DX2-201 (5 μM, 4 hours, Bru-seq)










Fold



Genes
change
Comments












NDRG1
2.874353
N-myc downstream regulated 1


ARG2
2.569327
arginase, type 11


KLHL24
2.336295
kelch-like 24 (Drosophila)


ANKRD37
2.182391
ankyrin repeat domain 37


FOXO6
1.954496
forkhead box protein O6


RNF122
1.914821
ring finger protein 122


SOX12
1.90308
SRY (sex determining region Y)-box 12


MXD4
1.81177
MAX dimerization protein 4


HDAC5
1.811478
histone deacetylase 5


MXI1
1.78479
MAX interactor 1


PDK1
1.778718
pyruvate dehydrogenase kinase,




isozyme 1


ERRFI1
1.770078
ERBB receptor feedback inhibitor 1


ZNF546
1.756829
zinc finger protein 546


PRDM1
1.748233
PR domain containing 1, with




ZNF domain


VLDLR
1.741318
very low density lipoprotein receptor


UPRT
1.740434
uracil phosphoribosyltransferase




(FUR1) homolog (S.cerevisiae)


LOXL4
1.730349
lysyl oxidase-like 4


ARHGAP30
1.72799
Rho GTPase activating protein 30


BNIP3L
1.72291
BCL2/adenovirus E1B 19 kDa




interacting protein 3-like


ARRDC3
1.707402
arrestin domain containing 3


YPEL1
1.70622779
Yippee Like 1


PPP1R3C
1.69920866
Protein Phosphatase 1 Regulatory




Subunit 3C


CREBRF
1.67605395
CREB3 Regulatory Factor


SEMA3B
1.66808745
Semaphorin 3B


CCNG2
1.66756367
Cyclin G2
















TABLE 5







Top 25 non-coding RNA downregulated in UM16 cell line


treated with DX2-201 (5 μM, 4 hours, Bru-seq)










Fold



Genes
change
Comments





AL096870.2
0.024652
processed_transcript


Y_RNA
0.028761
misc_RNA


RNU6-398P
0.034513
snRNA


RNU6-122P
0.043142
snRNA


TERC
0.043142
lincRNA


RNU6-548P
0.057523
snRNA


MIR5000
0.057523
miRNA


RNU1-73P
0.093812
snRNA


RNU1-19P
0.156354
snRNA


RNU6-519P
0.156354
snRNA


AC092916.2
0.170566
processed_pseudogene


C1orf50
0.187622
proteincoding


MIR545
0.187622
miRNA


Z99127.3
0.187622
processed_pseudogene


BX679664.2
0.187622
processed_pseudogene


RN7SL128P
0.187622
misc_RNA


AC008521.2
0.187622
processed_pseudogene


RN7SL388P
0.208469
misc_RNA


RN7SL589P
0.216487
misc_RNA


AC091133.4
0.234527
processed_pseudogene


AC019257.2
0.234527
lincRNA


SNORA73
0.234528
snoRNA


ZBTB40-IT1
0.234528
sense_intronic


RN7SL833P
0.234528
misc_RNA


AC106037.1
0.234528
sense_intronic
















TABLE 6







Top 25 non-coding RNA upregulated in UM16 cell line


treated with DX2-201(5 μM, 4 hours, Bru-seq)









Genes
Fold change
Comments












RNU6-1272P
25.49882
snRNA


RNU6-182P
20.39897
snRNA


Y_RNA
15.29913
misc_RNA


DPRXP4
3.189539
processed_pseudogene


RNA5SP379
2.81426
rRNA


RNU6-564P
2.81426
snRNA


RNU6-531P
2.81426
snRNA


AC034198.2
2.345256
antisense_RNA


AC007952.4
2.345252
lincRNA


AL136982.3
2.217333
sense_intronic


AP001527.2
2.217333
lincRNA


HMGN1P15
2.110723
processed_pseudogene


AC139887.1
2.06382
antisense_RNA


AC008687.3
2.032552
lincRNA


AC007686.1
2.010217
processed_pseudogene


AL161729.3
1.993466
lincRNA


EIF5AP3
1.876207
processed_pseudogene


AL645939.5
1.876196
lincRNA


SNORD124
1.876191
snoRNA


RNY3P2
1.876182
misc_RNA


RNU6-396P
1.876182
snRNA


AL627171.1
1.841464
lincRNA


AC105020.4
1.782395
antisense_RNA


AP003168.2
1.688585
sense_intronic


AP001619.1
1.620359
sense_intronic
















TABLE 7







GSEA results of the upregulated hallmark pathways in UM16


cell line treated with DX2-201 (5 μM, 4 hours, Bru-seq)

















NOM
FDR
FWER


NAME
SIZE
ES
NES
p-val
q-val
p-val
















HALLMARK_HYPOXIA
131
0.6294
2.8139
0
0.0000
0


HALLMARK_CHOLESTEROL_HOMEOSTASIS
58
0.4969
1.9354
0
0.0043
0.005


HALLMARK_GLYCOLYSIS
152
0.4109
1.8782
0
0.0054
0.009


HALLMARK_APOPTOSIS
119
0.3812
1.6931
0
0.0232
0.056


HALLMARK_UV_RESPONSE_DN
110
0.3784
1.6434
0
0.0304
0.091


HALLMARK_HEME_METABOLISM
137
0.3641
1.6288
0
0.0296
0.104


HALLMARK_P53_PATHWAY
156
0.3403
1.5682
0
0.0408
0.16


HALLMARK_ESTROGEN_RESPONSE_LATE
133
0.3437
1.5630
0
0.0370
0.166


HALLMARK_IL2_STAT5_SIGNALING
122
0.3402
1.5174
0.0027
0.0488
0.24


HALLMARK_MYOGENESIS
81
0.3657
1.5149
0.0160
0.0449
0.246


HALLMARK_TNFA_SIGNALING_VIA_NFKB
146
0.3199
1.4745
0.0026
0.0556
0.32


HALLMARK_MITOTIC_SPINDLE
190
0.3104
1.4709
0.0026
0.0530
0.33


HALLMARK_KRAS_SIGNALING_DN
45
0.3965
1.4318
0.0539
0.0675
0.415


HALLMARK_HEDGEHOG_SIGNALING
17
0.4848
1.3854
0.0981
0.0884
0.538


HALLMARK_ANDROGEN_RESPONSE
84
0.3155
1.3002
0.0550
0.1543
0.77


HALLMARK_FATTY_ACID_METABOLISM
114
0.2942
1.2729
0.0622
0.1755
0.831


HALLMARK_INFLAMMATORY_RESPONSE
91
0.2995
1.2667
0.0855
0.1740
0.847


HALLMARK_XENOBIOTIC_METABOLISM
105
0.2890
1.2573
0.0829
0.1775
0.87


HALLMARK_BILE_ACID_METABOLISM
57
0.3249
1.2479
0.1200
0.1785
0.883


HALLMARK_MTORC1_SIGNALING
177
0.2680
1.2451
0.0554
0.1726
0.887


HALLMARK_ALLOGRAFT_REJECTION
82
0.2962
1.2241
0.1200
0.1877
0.916





NES = Normalized Enrichment Score, FDR = False Discovery Rate, FWER = Family-Wise Error Rate.


Cutoff: FDR < 0.2













TABLE 8







GSEA results of the downregulated hallmark pathways in UM16


cell line treated with DX2-201 (5 μM, 4 hours, Bru-seq)

















NOM
FDR
FWER


NAME
SIZE
ES
NES
p-val
q-val
p-val
















HALLMARK_MYC_TARGETS_V1
189
−0.5426
−2.4192
0.0000
0.0000
0


HALLMARK_MYC_TARGETS_V2
53
−0.6265
−2.2565
0.0000
0.0000
0


HALLMARK_DNA_REPAIR
127
−0.4085
−1.7322
0.0016
0.0104
0.049


HALLMARK_E2F_TARGETS
195
−0.3804
−1.7010
0.0000
0.0103
0.065


HALLMARK_OXIDATIVE_PHOSPHORYLATION
176
−0.3812
−1.6882
0.0000
0.0092
0.073


HALLMARK_ANGIOGENESIS
16
−0.5078
−1.4124
0.0852
0.0973
0.635


HALLMARK_UNFOLDED_PROTEIN_RESPONSE
97
−0.3254
−1.3390
0.0521
0.1481
0.832





NES = Normalized Enrichment Score, FDR = False Discovery Rate, FWER = Family-Wise Error Rate.


Cutoff: FDR < 0.2













TABLE 9







GSEA results of the upregulated KEGG pathways in UM16


cell line treated with DX2-201 (5 μM, 4 hours, Bru-seq)

















NOM
FDR
FWER


NAME
SIZE
ES
NES
p-val
q-val
p-val
















KEGG_STARCH_AND_SUCROSE_METABOLISM
17
0.7356
2.1177
0.0000
0.0026
0.002


KEGG_DORSO_VENTRAL_AXIS_FORMATION
17
0.6614
1.9145
0.0000
0.0296
0.044


KEGG_GLYCOLYSIS_GLUCONEOGENESIS
33
0.5362
1.8482
0.0069
0.0432
0.095


KEGG_REGULATION_OF_AUTOPHAGY
18
0.5972
1.7250
0.0151
0.1086
0.276


KEGG_LEISHMANIA_INFECTION
31
0.5017
1.6760
0.0097
0.1288
0.382


KEGG_TYPE_II_DIABETES_MELLITUS
22
0.5316
1.6335
0.0178
0.1482
0.495


KEGG_RENAL_CELL_CARCINOMA
53
0.4210
1.6054
0.0069
0.1615
0.577





NES = Normalized Enrichment Score, FDR = False Discovery Rate, FWER = Family-Wise Error Rate.


Cutoff: FDR < 0.2













TABLE 10







GSEA results of the downregulated KEGG pathways in UM16


cell line treated with DX2-201 (5 pM, 4 hours, Bru-seq)

















NOM
FDR
FWER


NAME
SIZE
ES
NES
p-val
q-val
p-val
















KEGG_RIBOSOME
83
−0.6266
−2.4526
0.0000
0.0000
0.0000


KEGG_PURINE_METABOLISM
95
−0.4914
−2.0380
0.0000
0.0011
0.0030


KEGG_RNA_POLYMERASE
25
−0.6348
−1.9613
0.0018
0.0027
0.0100


KEGG_DNA_REPLICATION
34
−0.5795
−1.9358
0.0000
0.0029
0.0150


KEGG_PYRIMIDINE_METABOLISM
77
−0.4987
−1.9279
0.0000
0.0028
0.0180


KEGG_PROTEASOME
36
−0.5628
−1.9164
0.0000
0.0030
0.0240


KEGG_MISMATCH_REPAIR
21
−0.6381
−1.8751
0.0018
0.0053
0.0480


KEGG_SPLICEOSOME
113
−0.4510
−1.8728
0.0000
0.0048
0.0500


KEGG_N_GLYCAN_BIOSYNTHESIS
40
−0.5360
−1.8440
0.0000
0.0063
0.0740


KEGG_OXIDATIVE_PHOSPHORYLATION
87
−0.4451
−1.7650
0.0000
0.0144
0.1780


KEGG_PARKINSONS_DISEASE
82
−0.4446
−1.7382
0.0000
0.0173
0.2340


KEGG_HUNTINGTONS_DISEASE
126
−0.4035
−1.7040
0.0000
0.0215
0.3030


KEGG_ALZHEIMERS_DISEASE
108
−0.3872
−1.6021
0.0000
0.0524
0.5950


KEGG_NUCLEOTIDE_EXCISION_REPAIR
41
−0.4603
−1.5835
0.0175
0.0579
0.6550


KEGG_VIBRIO_CHOLERAE_INFECTION
37
−0.4430
−1.4689
0.0290
0.1376
0.9260


KEGG_AMINOACYL_TRNA_BIOSYNTHESIS
39
−0.4260
−1.4417
0.0430
0.1593
0.9680





NES = Normalized Enrichment Score, FDR = False Discovery Rate, FWER = Family-Wise Error Rate.


Cutoff: FDR < 0.2













TABLE 11







GSEA results of the upregulated GO pathways in UM16 cell line treated with DX2-201 (5 μM, 4 hours, Bru-seq)

















NOM
FDR
FWER


NAME
SIZE
ES
NES
p-val
q-val
p-val
















GO_NUCLEOTIDE_PHOSPHORYLATION
33
0.5920
2.0727
0.0000
0.0224
0.131


GO_RIBONUCLEOSIDE_DIPHOSPHATE_METABOLIC_PROCESS
41
0.5746
2.0919
0.0000
0.0225
0.1


GO_NADH_METABOLIC_PROCESS
24
0.6704
2.0814
0.0023
0.0235
0.121


GO_ATP_GENERATION_FROM_ADP
23
0.6848
2.0929
0.0000
0.0270
0.1


GO_ADP_METABOLIC_PROCESS
29
0.6343
2.1062
0.0000
0.0271
0.081


GO_GLUCOSE_CATABOLIC_PROCESS
18
0.7209
2.1191
0.0023
0.0273
0.062


GO_PYRUVATE_METABOLIC_PROCESS
36
0.6035
2.1281
0.0000
0.0324
0.049


GO_HEXOSE_CATABOLIC_PROCESS
26
0.6548
2.1301
0.0000
0.0622
0.047


GO_MONOSACCHARIDE_CATABOLIC_PROCESS
32
0.5791
1.9771
0.0000
0.0716
0.385


GO_RESPONSE_TO_OXYGEN_LEVELS
179
0.4230
1.9612
0.0000
0.0782
0.449


GO_NEGATIVE_REGULATION_OF_FAT_CELL_DIFFERENTIATION
23
0.6098
1.9052
0.0000
0.0995
0.676





NES = Normalized Enrichment Score, FDR = False Discovery Rate, FWER = Family-Wise Error Rate.


Cutoff: FDR < 0.1.













TABLE 12







GSEA results of the top 25 downregulated GO pathways in UM16 cell line treated with DX2-201 (5 μM, 4 hours, Bru-seq)

















NOM
FDR
FWER


NAME
SIZE
ES
NES
p-val
q-val
p-val
















GO_RIBOSOME_BIOGENESIS
270
−0.616
−2.869
0
0
0


GO_NCRNA_PROCESSING
329
−0.594
−2.802
0
0
0


GO_RRNA_METABOLIC_PROCESS
226
−0.604
−2.757
0
0
0


GO_RIBONUCLEOPROTEIN_COMPLEX_BIOGENESIS
383
−0.552
−2.663
0
0
0


GO_RIBOSOME
186
−0.589
−2.626
0
0
0


GO_NCRNA_METABOLIC_PROCESS
439
−0.528
−2.591
0
0
0


GO_CYTOSOLIC_RIBOSOME
99
−0.626
−2.548
0
0
0


GO_TRANSLATIONAL_INITIATION
134
−0.590
−2.521
0
0
0


GO_RIBOSOMAL_SUBUNIT
142
−0.581
−2.491
0
0
0


GO_ESTABLISHMENT_OF_PROTEIN_LOCAL-
97
−0.589
−2.417
0
0
0


IZATION_TO_ENDOPLASMIC_RETICULUM


GO_STRUCTURAL_CONSTITUENT_OF_RIBOSOME
172
−0.539
−2.395
0
0
0


GO_MULTI_ORGANISM_METABOLIC_PROCESS
132
−0.557
−2.388
0
0
0


GO_RIBOSOMAL_SMALL_SUBUNIT_BIOGENESIS
51
−0.653
−2.377
0
0
0


GO_CYTOSOLIC_PART
170
−0.537
−2.371
0
0
0


GO_RRNA_BINDING
50
−0.650
−2.367
0
0
0


GO_PRERIBOSOME
54
−0.648
−2.348
0
0
0


GO_PROTEIN_LOCALIZATION_TO_ENDOPLASMIC_RETICULUM
111
−0.563
−2.329
0
0
0


GO_LARGE_RIBOSOMAL_SUBUNIT
83
−0.585
−2.298
0
0
0


GO_CYTOSOLIC_LARGE_RIBOSOMAL_SUBUNIT
56
−0.629
−2.298
0
0
0


GO_NUCLEOLAR_PART
51
−0.630
−2.294
0
0
0


GO_TRNA_PROCESSING
89
−0.559
−2.240
0
3.79E−05
0.001


GO_NUCLEAR_TRANSCRIBED_MRNA_CATABOLIC_PRO-
113
−0.539
−2.231
0
3.62E−05
0.001


CESS_NONSENSE_MEDIATED_DECAY


GO_PEPTIDE_METABOLIC_PROCESS
413
−0.461
−2.222
0
3.46E−05
0.001


GO_MATURATION_OF_SSU_RRNA
38
−0.642
−2.206
0
6.60E−05
0.002


GO_AMIDE_BIOSYNTHETIC_PROCESS
389
−0.454
−2.193
0
1.55E−04
0.005





NES = Normalized Enrichment Score, FDR = False Discovery Rate, FWER = Family-Wise Error Rate.


Cutoff: FDR < 0.1.













TABLE 13







GSEA results of the top 25 upregulated transcription factors in


UM16 cell line treated with DX2-201 (5 μM, 4 hours, Bru-seq)

















NOM
FDR
FWER


NAME
SIZE
ES
NES
p-val
q-val
p-val
















FOXO4_01
134
0.464
2.069
0.000
0.014
0.007


FOXO1_02
146
0.459
2.057
0.000
0.007
0.007


FOXO4_02
128
0.454
2.000
0.000
0.007
0.011


FREAC2_01
134
0.440
1.964
0.000
0.008
0.016


AAAYWAACM_HFH4_01
122
0.431
1.896
0.000
0.013
0.033


FOXO3——01
126
0.417
1.860
0.000
0.015
0.046


RTAAACA_FREAC2_01
499
0.344
1.816
0.000
0.025
0.083


FAC1_01
124
0.394
1.751
0.000
0.043
0.157


AP4_Q5
124
0.393
1.743
0.000
0.043
0.173


HFH3_01
99
0.401
1.733
0.000
0.042
0.190


FOXO1_01
126
0.384
1.732
0.000
0.039
0.191


ATF1_Q6
122
0.383
1.701
0.000
0.053
0.264


OCT_Q6
121
0.381
1.697
0.000
0.051
0.274


TTANWNANTGGM_UNKNOWN
27
0.511
1.686
0.007
0.051
0.295


PAX2_02
132
0.374
1.664
0.000
0.061
0.366


NKX62_Q2
102
0.387
1.663
0.003
0.058
0.368


FREAC3_01
121
0.375
1.657
0.000
0.058
0.389


E47_01
131
0.363
1.646
0.003
0.060
0.418


YWATTWNNRGCT_UNKNOWN
34
0.482
1.641
0.007
0.059
0.430


AP4_Q6
92
0.377
1.607
0.000
0.078
0.537


RYCACNNRNNRNCAG_UNKNOWN
38
0.462
1.606
0.012
0.075
0.542


HNF3_Q6
79
0.392
1.604
0.005
0.073
0.547


FOXJ2_02
106
0.364
1.582
0.010
0.090
0.648


OCT1_Q5_01
118
0.357
1.574
0.003
0.094
0.674


AREB6_04
134
0.346
1.566
0.003
0.098
0.708





NES = Normalized Enrichment Score, FDR = False Discovery Rate, FWER = Family-Wise Error Rate.


Cutoff: FDR < 0.1













TABLE 14







GSEA results of the downregulated transcription factors in


UM16 cell line treated with DX2-201 (5 μM, 4 hours, Bru-seq)

















NOM
FDR
FWER


NAME
SIZE
ES
NES
p-val
q-val
p-val
















MYCMAX_01
164
−0.425
−1.853
0.000
0.018
0.044


MYC_Q2
115
−0.429
−1.792
0.000
0.022
0.107


YY1_Q6
184
−0.372
−1.643
0.000
0.072
0.518


E2F_Q6
173
−0.352
−1.565
0.002
0.078
0.833


GGAANCGGAANY_UNKNOWN
92
−0.409
−1.661
0.000
0.080
0.453


E2F1_Q6
175
−0.355
−1.568
0.002
0.085
0.822


E2F1_Q6_01
180
−0.352
−1.578
0.000
0.086
0.78


NMYC_01
176
−0.350
−1.545
0.006
0.087
0.892


E2F_03
178
−0.360
−1.597
0.000
0.097
0.704


SGCGSSAAA_E2F1DP2_01
126
−0.373
−1.581
0.002
0.098
0.773





NES = Normalized Enrichment Score, FDR = False Discovery Rate, FWER = Family-Wise Error Rate.


Cutoff: FDR < 0.1













TABLE 15







cMap analysis results of top 25 similar


compounds (5 μM, 4 hours, Bru-seq)









Score
Name
Description












99.68
PAC-1
Caspase activator


99.65
hinokitiol
Tyrosinase inhibitor


99.47
BMS-536924
IGF-1 inhibitor


99.44
PP-30
RAF inhibitor


99.4
WAY-170523
Metalloproteinase inhibitor


99.26
AZ-628
RAF inhibitor


99.26
SA-1478088
−666


99.12
TW-37
BCL inhibitor


99.08
selumetinib
MEK inhibitor


99.05
VU-0418946-1
HIF modulator


98.98
APHA-
HDAC inhibitor



compound-8



98.91
ISOX
HDAC inhibitor


98.87
NCH-51
HDAC inhibitor


98.87
pyroxamide
HDAC inhibitor


98.8
geldanamycin
HSP inhibitor


98.8
PI-103
MTOR inhibitor


98.8
WYE-354
MTOR inhibitor


98.77
latrunculin-b
Actin polymerization inhibitor


98.77
WZ-3146
EGFR inhibitor


98.77
ZSTK-474
PI3K inhibitor


98.73
trichostatin-a
HDAC inhibitor


98.73
WT-171
HDAC inhibitor


98.73
GSK-1059615
PI3K inhibitor


98.7
U-0126
MEK inhibitor


98.7
QL-XI-92
DDR1 inhibitor
















TABLE 16







cMap analysis results of top 25


opposing compounds (5 μM, 4 hours, Bru-seq)











Score
Name
Description







−99.89
rucaparib
PARP inhibitor



−99.86
SB-216763
Glycogen synthase kinase





inhibitor



−99.86
PKCbeta-inhibitor
PKC inhibitor



−99.82
kenpaullone
CDK inhibitor



−99.41
androstenol
GABA receptor modulator



−99.33
ascorbyl-palmitate
antioxidant



−99.3
acetyl-
Inhibitor of methyl esterification




geranygeranyl-
of geranylgeranylated proteins




cysteine




−99.22
enzastaurin
PKC inhibitor



−99.13
nifedipine
Calcium channel blocker



−99.12
prostaglandin-a1
HSP inducer



−99.01
VX-222
HCV inhibitor



−99.01
danoprevir
HCV inhibitor



−98.81
2-aminopurine
Serine/threonine kinase inhibitor



−98.66
maraviroc
CC chemokine receptor





antagonist



−98.66
valproic-acid
HD AC inhibitor



−98.63
AT-9283
JAK inhibitor



−98.29
thiazolopyrimidine
CDC inhibitor



−97.96
orantinib
FGFR inhibitor



−97.74
GSK-3-inhibitor-IX
Glycogen synthase kinase





inhibitor



−97.36
SB-415286
Glycogen synthase kinase





inhibitor



−96.94
olomoucine
CDK inhibitor



−96.83
GW-843682X
PLK inhibitor



−96.79
RHO-kinase-
Rho associated kinase inhibitor




inhibitor-





III[rockout]




−96.75
esmolol
Adrenergic receptor antagonist



−99.89
rucaparib
PARP inhibitor

















TABLE 17







cMap analysis results of top 25 predicted gene


knockdown (similar) (5 μM, 4 hours, Bru-seq)









Score
Name
Description












99.95
ATP6V0C
ATPases/V-type


99.95
MYC
Basic helix-loop-helix proteins


99.92
SMC1A
Structural maintenance of


99.76
MST1R
chromosomes proteins




Type X RTKs: HGF (hepatocyte




growth factor) receptor family


99.68
SFPQ
RNA binding motif (RRM)




containing


99.68
STK3
MST subfamily


99.66
SERINC3



99.63
ME1



99.44
RRM1
Ribonucleoside-diphosphate




reductases


99.37
TESK1
TESK subfamily


99.25
ADAM15
ADAM metallopeptidase domain




containing


99.21
PXN



99.12
MDH2



99.08
OGG1



98.95
AP2M1



98.69
RANBP9



98.5
NCOA3
Histone acetyltransferases


98.46
NCOA4



98.31
ARID4B



98.25
PTP4A1
Protein tyrosine phosphatases/




Class I Cys-based PTPs:PRLs


97.93
CCND3



97.77
TNFAIP1
BTB/POZ domain containing


97.77
COX4I1
Mitochondrial respiratory chain




complex


97.76
TTR



97.55
MYD88

















TABLE 18







cMap analysis results of top 25 predicted knockdown


(opposing) (5 μM, 4 hours, Bru-seq)









Score
Name
Description












−99.92
DHX8
DEAH-boxes


−99.87
IFNGR1
Interferon receptor family


−99.71
LOXL1



−99.68
SIAH2



−99.58
SLC16A6
SLC16 family of




monocarboxylate transporters


−99.48
PAFAH1B2



−99.34
ADAM10
ADAM metallopeptidase domain




containing


−99.32
RPS6KB2
p70 subfamily


−99.13
BMI1
Polycomb group ring fingers


−99.03
ATF1
basic leucine zipper proteins


−98.95
ABCA5
ATP binding cassette




transporters/subfamily A


−98.86
CP



−98.44
INS



−98.24
TNFRSF19
Tumour necrosis factor (TNF)




receptor family


−98.11
RASSF5



−97.98
RCHY1
RING-type (C3HC4) zinc




fingers


−97.84
CREBBP
Chromatin-modifying enzymes/




K-acetyltransferases


−97.77
TCF7L2



−97.76
RPL7
L ribosomal proteins


−97.75
DNAJC15
Heat shock proteins/DNAJ




(HSP40)


−97.62
KBTBD2
BTB/POZ domain containing


−97.55
PAFAH1B1
WD repeat domain containing


−97.54
RASD1



−97.46
NIPSNAP1



−97.4
SCAP
WD repeat domain containing
















TABLE 19







cMap analysis results of top 25 predicted gene overexpression


(similar) (5 μM, 4 hours, Bru-seq)











Score
Name
Description















99.72
EBF1




99.49
LIG1




99.26
PAX8
Paired boxes



98.94
KLF6
Kruppel-like transcription





factors



98.33
IFNB1
Interferons



98.15
TAF13




98.02
MEIS2
Homeoboxes/TALE class



97.93
CDX2
Homeoboxes/ANTP class:





HOXL subclass



97.92
OVOL2
Zinc fingers, C2H2-type



97.87
UGCG
Glycosyltransferase family 2





domain containing



97.69
SOX2
SRY (sex determining region





Y)-boxes



97.22
SATB2
Homeoboxes/CUT class



96.78
ELK1
ETS Transcription Factors



95.94
HOXC9
Homeoboxes/ANTP class:





HOXL subclass



95.9
HOXB13
Homeoboxes/ANTP class:





HOXL subclass



95.83
EIF4E3
Translation Initiation Factor 4E



95.45
MXD3
Basic helix-loop-helix proteins



95.36
VGLL4




95.32
DUSP28
Protein tyrosine phosphatases/





Class I Cys-based PTPs:





Atypical dual specificity





phosphatases



94.75
ESR1
Estrogen receptors



94.15
VPS28




93.66
TRIP10




92.82
SOX10
SRY (sex determining region





Y)-boxes





Protein tyrosine phosphatases/



92.64
DUSP6
Class I Cys-based PTPs:MAP





kinase phosphatases



92.56
FBXW7
F-boxes/WD-40 domains

















TABLE 20







cMap analysis results of top 25 predicted gene


overexpression (opposing) (5 μM, 4 hours, Bru-seq)









Score
Name
Description





−96.49
TSPAN8
Tetraspanins


−96.34
CETN3
EF-hand domain containing


−96.2
PDGFRA
Type III RTKs: PDGFR, CSFR,




Kit, FLT3 receptor family


−92.63
CPD
Carboxypeptidase A


−92.05
PHF13
Zinc fingers, PHD-type


−91.81
STX4



−91.8
UGT1A9
UDP glucuronosyltransferases


−90.66
FCGR2A
CD molecules


−90.09
ENOSF1



−87.85
RUFY1
Zinc fingers, FYVE domain




containing


−86.48
MMP14
Matrix metallopeptidase


−85.99
SSX3



−85.63
ERGIC2



−85.06
TMEM174



−84.94
PSMA3
Proteasome subunits


−84.42
EIF4EBP2



−84.14
PIAS1
Zinc fingers, MIZ-type


−83.9
PSMD3
Proteasome (prosome,




macropain) subunits


−83.39
SRSF4
RNA binding motif (RRM)




containing


−83.36
FOXR1
Forkhead boxes


−83.2
BRF2



−82.18
RNF5
RING-type (C3HC4) zinc




fingers


−82.17
GNA11



−81.78
LAGE3



−81.78
TMEM5

















TABLE 21







cMap analysis results of top 25 sets of compound or


genetic perturbagens (Similar) (5 μM, 4 hours, Bru-seq)









Score
Name
Description












99.6
IGF-1 inhibitor



99.55
HIF activator



99.5
HDAC inhibitor



99.47
PI3K inhibitor



99.44
Bromodomain




Inhibitor



99.43
MTOR inhibitor



99.37
SRC inhibitor




DNA dependent



99.33
protein kinase




inhibitor



98.98
MEK inhibitor



98.63
Mitochondrial
Genetic, loss of function



complex IV LOF



98.63
HSP inhibitor



98.25
V type ATPases
Genetic, loss of function



LOF



98.09
Aurora kinase




inhibitor



97.72
FLT3 inhibitor



97.63
T-type calcium




channel blocker



97.09
EGFR inhibitor



96.77
Lysine
Genetic, loss of function



acetyltransferases




LOF



96.73
PDGFR/KIT




inhibitor



95.93
Dopamine receptor




antagonist



95.14
Estrogen receptor




antagonist



94.74
JAK inhibitor



93.84
RAF inhibitor



92.36
Homeobox Gene
Genetic, gain of function



GOF



91.2
Leucine rich repeat




kinase inhibitor



88.75
VEGFR inhibitor

















TABLE 22







cMap analysis results of top 25 sets of compound or genetic


perturbagens (opposing) (5 μM, 4 hours, Bru-seq)









Score
Name
Description












−99.03
Glycogen synthase kinase




inhibitor



−92.69
MDM inhibitor



−92.06
Bile acid



−77.08
Tubulin inhibitor



−71.93
Retinoid receptor agonist



−67.82
Integrin subunits beta LOF
Genetic, loss of function


−64.42
TGF beta receptor inhibitor



−62.94
Phospholipases LOF
Genetic, loss of function


−62.25
Aromatase inhibitor



−58.57
PI3K Signaling LOF
Genetic, loss of function


−56.89
X linked mental retardation
Genetic, loss of function



group 1 LOF



−56.68
Bacterial 30S ribosomal




subunit inhibitor



−55.23
Serpin peptidase inhibitors LOF
Genetic, loss of function


−52.83
FXR antagonist



−52.14
NADH ubiquinone
Genetic, loss of function



oxidoreductase supernumerary




subunits LOF



−52.09
Lipocalins GOF
Genetic, gain of




function


−50.12
PPAR receptor agonist



−45.51
Tumor necrosis factor
Genetic, loss of function



superfamily LOF



−43.05
Thymidylate synthase inhibitor



−31.62
Estrogen receptor agonist



−31.13
Poly ADP ribose polymerases
Genetic, loss of function



LOF



−29.32
HIV protease inhibitor



−29.22
S100 calcium binding proteins
Genetic, loss of function



LOF



−27.81
Adenosine receptor agonist



−23.72
Non Homologous End Joining
Genetic, loss of function



LOF
















TABLE 23







Top 25 proteins upregulated in UMI 6 cell line


treated with DX2-201 (5 μM, 24 hours)










Fold



Genes
change
Description












gk
3.009
Glycerol kinase GK


PRSS23
1.844
Serine protease 23 PRSS23


NR2F6
1.842
Nuclear receptor subfamily 2 group F




member 6 NR2F6


CMTM4
1.678
CKLF-like MARVEL transmembrane domain-




containing protein 4 CMTM4


TMEM55B
1.664
Type 1 phosphatidylinositol 4,5-bisphosphate




4-phosphatase TMEM55B


BTBD10
1.651
BTB/POZ domain-containing protein 10 BTBD10


CCNT2
1.65
Cyclin-T2 CCNT2


CYP51A1
1.636
Lanosterol 14-alpha demethylase CYP51A1


UQCC3
1.635
Ubiquinol-cytochrome-c reductase complex




assembly factor 3 UQCC3


NDRG1
1.55
Protein NDRG1 NDRG1


CDC42BPA
1.514
Serine/threonine-protein kinase MRCK




alpha CDC42BPA


scaf8
1.508
Protein SCAF8 SCAF8


KIAA0355
1.48
Uncharacterized protein KIAA0355 KIAA0355


APBB1IP
1.458
Amyloid beta A4 precursor protein-binding family




B member 1-interacting protein APBB1IP


TADA3
1.45
Transcriptional adapter 3 TAD A3


RCCD1
1.443
RCC1 domain-containing protein 1 RCCD1


RDH13
1.429
Retinol dehydrogenase 13 RDH13


OSTC
1.418
Oligosaccharyltransferase complex




subunit OSTC OSTC


RRM2B
1.395
Ribonucleoside-diphosphate reductase




subunit M2 B RRM2B


ALAS1
1.393
5-aminolevulinate synthase, nonspecific,




mitochondrial ALAS1


MROH1
1.386
Maestro heat-like repeat-containing




protein family member 1 MROH1 PE = 2


krt17
1.386
Keratin, type 1 cytoskeletal 17 KRT17


DHTKD1
1.384
Probable 2-oxoglutarate dehydrogenase E1




component DHKTD1, mitochondrial DHTKD1


TCEAL1
1.382
Transcription elongation factor A




protein-like 1 TCEAL1


NAA35
1.377
N-alpha-acetyltransferase 35, NatC




auxiliary subunit NAA35
















TABLE 24







Top 25 proteins downregulated in UM 16 cell line treated with


DX2-201 (5 μM, 24 hours)










Fold



Genes
change
Description












DLD
0.684
Dihydrolipoyl dehydrogenase, mitochondrial DLD


Hist1h1d
0.703
Histone H1.3 HIST1H1D


ZBTB21
0.709
Zinc finger and BTB domain-containing




protein 21 ZBTB21


SCAMP2
0.729
Secretory carrier-associated membrane




protein 2 SCAMP2


DOLPP1
0.731
Dolichyldiphosphatase 1 DOLPP1 PE = 2 SV = 1


pop7
0.736
Ribonuclease P protein subunit p20 POP7


FAU
0.742
40S ribosomal protein S30 FAU


PDCL
0.752
Phosducin-like protein PDCL


OGFOD2
0.754
2-oxoglutarate and iron-dependent oxygenase




domain-containing protein 2 OGFOD2


CDCA5
0.761
Sororin CDCA5


PLP2
0.763
Proteolipid protein 2 PLP2


RUSC1
0.771
RUN and SH3 domain-containing protein 1 RUSC1


NOL8
0.771
Nucleolar protein 8 NOL8


ANKH
0.777
Progressive ankylosis protein homolog ANKH


ABTB2
0.778
Ankyrin repeat and BTB/POZ domain-




containing protein 2 ABTB2


AP2S1
0.781
AP-2 complex subunit sigma AP2S1


FAH
0.785
Fumarylacetoacetase FAH


CCDC88C
0.785
Protein Daple CCDC88C


MED27
0.79
Mediator of RNA polymerase II transcription




subunit 27 MED27


AP1S2
0.792
AP-1 complex subunit sigma-2 AP1S2


DTNBP1
0.794
Dysbindin DTNBP1


NRGN
0.795
Neurogranin NRGN


SLC25A15
0.796
Mitochondrial ornithine transporter 1 SLC25A15


DOPEY2
0.797
Protein dopey-2 DOPEY2


ANKRD54
0.799
Ankyrin repeat domain-containing




protein 54 ANKRD54
















TABLE 25







GSEA results of the top 20 upregulated HALLMARK pathways in UM16


cell line treated with DX2-201(5 μM, 24 hours, proteomics)

















NOM
FDR
FWER


NAME
SIZE
ES
NES
p-val
q-val
p-val
















HALLMARK_HYPOXIA
74
0.595
1.851
0.000
0.003
0.004


HALLMARK_COAGULATION
40
0.636
1.813
0.004
0.004
0.009


HALLMARK_BILE_ACID_METABOLISM
35
0.523
1.465
0.058
0.245
0.589


HALLMARK_CHOLESTEROL_HOMEOSTASIS
37
0.493
1.407
0.059
0.314
0.785


HALLMARK_COMPLEMENT
72
0.401
1.235
0.153
0.425
0.994


HALLMARK_DNA_REPAIR
100
0.381
1.239
0.127
0.454
0.994


HALLMARK_EPITHELIAL_MESENCHYMAL_TRANSITION
48
0.449
1.312
0.103
0.456
0.968


HALLMARK_TNFA_SIGNALING_VIA_NFKB
54
0.448
1.335
0.096
0.457
0.943


HALLMARK_XENOBIOTIC_METABOLISM
81
0.393
1.244
0.146
0.488
0.993


HALLMARK_GLYCOLYSIS
105
0.399
1.281
0.076
0.489
0.984


HALLMARK_UV_RESPONSE_DN
50
0.433
1.256
0.146
0.510
0.991


HALLMARK_MITOTIC_SPINDLE
144
0.348
1.170
0.180
0.529
0.999


HALLMARK_IL2_STAT5_SIGNALING
60
0.384
1.176
0.228
0.552
0.999


HALLMARK_HEME_METABOLISM
73
0.376
1.150
0.250
0.552
0.999


KEGG_MELANOGENESIS
21
0.534
1.369
0.105
0.638
1


HALLMARK_ADIPOGENESIS
109
0.317
1.025
0.436
0.644
1


HALLMARK_PEROXISOME
57
0.347
1.037
0.414
0.645
1


KEGG_GLYCEROLIPID_METABOLISM
17
0.572
1.376
0.088
0.663
1


HALLMARK_APOPTOSIS
75
0.335
1.042
0.403
0.666
1


KEGG_ARGININE_AND_PROLINE_METABOLISM
25
0.543
1.389
0.087
0.668
0.999





NES = Normalized Enrichment Score, FDR = False Discovery Rate, FWER = Family-Wise Error Rate.













TABLE 26







GSEA results of the downregulated HALLMARK pathways in UM16


cell line treated with DX2-201(5 μM, 24hours, proteomics)

















NOM
FDR
FWER


NAME
SIZE
ES
NES
p-val
q-val
p-val
















HALLMARK_MYC_TARGETS_V1
186
−0.424
−1.975
0.000
0.006
0.003


HALLMARK_E2F_TARGETS
157
−0.349
−1.559
0.000
0.077
0.076


HALLMARK_UNFOLDED_PROTEIN_RESPONSE
76
−0.372
−1.517
0.008
0.070
0.102


HALLMARK_G2M_CHECKPOINT
139
−0.313
−1.421
0.000
0.107
0.195


HALLMARK_P53_PATHWAY
78
−0.323
−1.338
0.015
0.146
0.315


HALLMARK_MYC_TARGETS_V2
47
−0.329
−1.215
0.135
0.264
0.565


HALLMARK_ALLOGRAFT_REJECTION
54
−0.287
−1.082
0.309
0.521
0.874


HALLMARK_MYOGENESIS
48
−0.285
−1.077
0.323
0.467
0.878


HALLMARK_TGF_BETA_SIGNALING
25
−0.296
−0.944
0.538
0.771
0.978


HALLMARK_OXIDATIVE_PHOSPHORYLATION
159
−0.214
−0.938
0.620
0.711
0.98


HALLMARK_PROTEIN_SECRETION
79
−0.207
−0.842
0.869
0.862
0.995


HALLMARK_SPERMATOGENESIS
33
−0.229
−0.803
0.768
0.851
0.998





NES = Normalized Enrichment Score, FDR = False Discovery Rate, FWER = Family-Wise Error Rate.













TABLE 27







GSEA results of the top 20 upregulated KEGG pathways in UM16


cell line treated with DX2-201 (5 μM, 24 hours, proteomics)

















NOM
FDR
FWER


NAME
SIZE
ES
NES
p-val
q-val
p-val
















KEGG_B_CELL_RECEPTOR_SIGNALING_PATHWAY
37
0.504
1.432
0.058
0.813
0.995


HALLMARK_ESTROGEN_RESPONSE_EARLY
69
0.301
0.929
0.614
0.813
1


KEGG_ERBB_SIGNALING_PATHWAY
44
0.488
1.410
0.051
0.821
0.997


KEGG_FC_EPSILON_RI_SIGNALING_PATHWAY
29
0.593
1.623
0.011
0.825
0.539


KEGG_NUCLEOTIDE_EXCISION_REPAIR
32
0.469
1.271
0.142
0.829
1


KEGG_VIRAL_MYOCARDITIS
23
0.497
1.292
0.159
0.832
1


KEGG_PEROXISOME
38
0.459
1.302
0.134
0.839
1


KEGG_INOSITOL_PHOSPHATE_METABOLISM
19
0.520
1.277
0.167
0.852
1


KEGG_PATHWAYS_IN_CANCER
119
0.324
1.074
0.341
0.858
1


KEGG_INSULIN_SIGNALING_PATHWAY
66
0.352
1.077
0.357
0.872
1


KEGG_PROP_ANOATE_METABOLISM
21
0.424
1.079
0.385
0.889
1


KEGG_GLYCOLYSIS_GLUCONEOGENESIS
35
0.394
1.086
0.354
0.889
1


KEGG_NON_SMALL_CELL_LUNG_CANCER
27
0.415
1.092
0.368
0.894
1


KEGG_GAP_JUNCTION
31
0.386
1.041
0.409
0.898
1


KEGG_CARDIAC_MUSCLE_CONTRACTION
21
0.469
1.177
0.269
0.900
1


KEGG_ADIPOCYTOKINE_SIGNALING_PATHWAY
28
0.390
1.046
0.427
0.903
1


KEGG_CYTOSOLIC_DNA_SENSING_PATHWAY
19
0.469
1.166
0.287
0.907
1


KEGG_PURINE_METABOLISM
72
0.331
1.023
0.448
0.909
1


KEGG_LYSINE_DEGRADATION
23
0.381
1.007
0.449
0.914
1


HALLMARK_ANDROGEN_RESPONSE
52
0.259
0.760
0.846
0.915
1





NES = Normalized Enrichment Score, FDR = False Discovery Rate, FWER = Family-Wise Error Rate.













TABLE 28







GSEA results of the downregulated KEGG pathways in UM16 cell


line treated with DX2-201 (5 μM, 24 hours, proteomics)

















NOM
FDR
FWER


NAME
SIZE
ES
NES
p-val
q-val
p-val
















KEGG_RIBOSOME
79
−0.520
−2.132
0.000
0.003
0.004


KEGG_CITRATE_CYCLE_TCA_CYCLE
25
−0.445
−1.416
0.045
0.422
0.79


KEGG_SPLICEOSOME
110
−0.310
−1.319
0.033
0.541
0.942


KEGG_PROTEASOME
40
−0.401
−1.425
0.025
0.605
0.776


KEGG_PROTEIN_EXPORT
18
−0.376
−1.116
0.326
0.625
0.998


KEGG_N_GLYCAN_BIOSYNTHESIS
26
−0.366
−1.160
0.246
0.658
0.994


KEGG_RNA_DEGRADATION
42
−0.342
−1.228
0.158
0.672
0.988


KEGG_BASAL_TRANSCRIPTION_FACTORS
17
−0.389
−1.121
0.278
0.687
0.998


KEGG_PARKINSONS_DISEASE
74
−0.290
−1.180
0.148
0.702
0.994


KEGG_VIBRIO_CHOLERAE_INFECTION
22
−0.335
−1.044
0.395
0.754
1


KEGG_ANTIGEN_PROCESSING_AND_PRESENTATION
27
−0.285
−0.917
0.576
0.792
1


KEGG_B_CELL_RECEPTOR_SIGNALING_PATHWAY
37
0.504
1.432
0.058
0.813
0.995


KEGG_ERBB_SIGNALING_PATHWAY
44
0.488
1.410
0.051
0.821
0.997


KEGG_FC_EPSILON_RISIGNALING_PATHWAY
29
0.593
1.623
0.011
0.825
0.539


KEGG_CELL_CYCLE
71
−0.246
−0.995
0.490
0.828
1


KEGG_NUCLEOTIDE_EXCISION_REPAIR
32
0.469
1.271
0.142
0.829
1


KEGG_VIRAL_MYOCARDITIS
23
0.497
1.292
0.159
0.832
1


KEGG_PEROXISOME
38
0.459
1.302
0.134
0.839
1


KEGG_DNA_REPLICATION
30
−0.273
−0.919
0.577
0.844
1





NES = Normalized Enrichment Score, FDR = False Discovery Rate, FWER = Family-Wise Error Rate.






REFERENCES



  • (1) Roesch, A.; Vultur, A.; Bogeski, I.; Wang, H.; Zimmermann, K. M.; Speicher, D.; Korbel, C.; Laschke, M. W.; Gimotty, P. A.; Philipp, S. E.; Krause, E.; Patzold, S.; Villanueva, J.; Krepler, C.; Fukunaga-Kalabis, M.; Hoth, M.; Bastian, B. C.; Vogt, T.; Herlyn, M. Overcoming intrinsic multidrug resistance in melanoma by blocking the mitochondrial respiratory chain of slow-cycling JARID1B (high) cells. Cancer Cell 2013, 23, 811-25.

  • (2) Hu, J.; Locasale, J. W.; Bielas, J. H.; O'Sullivan, J.; Sheahan, K.; Cantley, L. C.; Vander Heiden, M. G.; Vitkup, D. Heterogeneity of tumor-induced gene expression changes in the human metabolic network. Nat Biotechnol 2013, 31, 522-9.

  • (3) Sriskanthadevan, S.; Jeyaraju, D. V.; Chung, T. E.; Prabha, S.; Xu, W.; Skrtic, M.; Jhas, B.; Hurren, R.; Gronda, M.; Wang, X.; Jitkova, Y.; Sukhai, M. A.; Lin, F. H.; Maclean, N.; Laister, R.; Goard, C. A.; Mullen, P. J.; Xie, S.; Penn, L. Z.; Rogers, I. M.; Dick, J. E.; Minden, M. D.; Schimmer, A. D. AML cells have low spare reserve capacity in their respiratory chain that renders them susceptible to oxidative metabolic stress. Blood 2015, 125, 2120-30.

  • (4) Birsoy, K.; Wang, T.; Chen, W. W.; Freinkman, E.; Abu-Remaileh, M.; Sabatini, D. M. An Essential Role of the Mitochondrial Electron Transport Chain in Cell Proliferation Is to Enable Aspartate Synthesis. Cell 2015, 162, 540-51.

  • (5) Sullivan, L. B.; Gui, D. Y.; Hosios, A. M.; Bush, L. N.; Freinkman, E.; Vander Heiden, M. G. Supporting Aspartate Biosynthesis Is an Essential Function of Respiration in Proliferating Cells. Cell 2015, 162, 552-63.

  • (6) Birsoy, K.; Possemato, R.; Lorbeer, F. K.; Bayraktar, E. C.; Thiru, P.; Yucel, B.; Wang, T.; Chen, W. W.; Clish, C. B.; Sabatini, D. M. Metabolic determinants of cancer cell sensitivity to glucose limitation and biguanides. Nature 2014, 508, 108-+.

  • (7) Lissanu Deribe, Y.; Sun, Y.; Terranova, C.; Khan, F.; Martinez-Ledesma, J.; Gay, J.; Gao, G.; Mullinax, R. A.; Khor, T.; Feng, N.; Lin, Y. H.; Wu, C. C.; Reyes, C.; Peng, Q.; Robinson, F.; Inoue, A.; Kochat, V.; Liu, C. G.; Asara, J. M.; Moran, C.; Muller, F.; Wang, J.; Fang, B.; Papadimitrakopoulou, V.; Wistuba, II; Rai, K.; Marszalek, J.; Futreal, P. A. Mutations in the SWI/SNF complex induce a targetable dependence on oxidative phosphorylation in lung cancer. Nat Med 2018, 24, 1047-1057.

  • (8) Sancho, P.; Bameda, D.; Heeschen, C. Hallmarks of cancer stem cell metabolism. British Journal of Cancer 2016, 114, 1305-1312.

  • (9) Kuntz, E. M.; Baquero, P.; Michie, A. M.; Dunn, K.; Tardito, S.; Holyoake, T. L.; Helgason, G. V.; Gottlieb, E. Targeting mitochondrial oxidative phosphorylation eradicates therapy-resistant chronic myeloid leukemia stem cells. Nature Medicine 2017, 23, 1234-+.

  • (10) Viale, A.; Pettazzoni, P.; Lyssiotis, C. A.; Ying, H.; Sanchez, N.; Marchesini, M.; Carugo, A.; Green, T.; Seth, S.; Giuliani, V.; Kost-Alimova, M.; Muller, F.; Colla, S.; Nezi, L.; Genovese, G.; Deem, A. K.; Kapoor, A.; Yao, W.; Brunetto, E.; Kang, Y.; Yuan, M.; Asara, J. M.; Wang, Y. A.; Heffernan, T. P.; Kimmelman, A. C.; Wang, H.; Fleming, J. B.; Cantley, L. C.; DePinho, R. A.; Draetta, G. F. Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function. Nature 2014, 514, 628-32.

  • (11) Lee, K. M.; Giltnane, J. M.; Balko, J. M.; Schwarz, L. J.; Guerrero-Zotano, A. L.; Hutchinson, K. E.; Nixon, M. J.; Estrada, M. V.; Sanchez, V.; Sanders, M. E.; Lee, T.; Gomez, H.; Lluch, A.; Perez-Fidalgo, A.; Wolf, M. M.; Andrejeva, G.; Rathmell, J. C.; Fesik, S. W.; Arteaga, C. L. MYC and MCL1 Cooperatively Promote Chemotherapy-Resistant Breast Cancer Stem Cells via Regulation of Mitochondrial Oxidative Phosphorylation. Cell Metabolism 2017, 26, 633-+.

  • (12) Vazquez, F.; Lim, J. H.; Chim, H.; Bhalla, K.; Gimun, G.; Pierce, K.; Clish, C. B.; Granter, S. R.; Widlund, H. R.; Spiegelman, B. M.; Puigserver, P. PGC1 alpha Expression Defines a Subset of Human Melanoma Tumors with Increased Mitochondrial Capacity and Resistance to Oxidative Stress. Cancer Cell 2013, 23, 287-301.

  • (13) Farge, T.; Saland, E.; de Toni, F.; Aroua, N.; Hosseini, M.; Perry, R.; Bosc, C.; Sugita, M.; Stuani, L.; Fraisse, M.; Scotland, S.; Larrue, C.; Boutzen, H.; Feliu, V.; Nicolau-Travers, M. L.; Cassant-Sourdy, S.; Broin, N.; David, M.; Serhan, N.; Sarry, A.; Tavitian, S.; Kaoma, T.; Vallar, L.; Iacovoni, J.; Linares, L. K.; Montersino, C.; Castellano, R.; Griessinger, E.; Collette, Y.; Duchamp, O.; Barreira, Y.; Hirsch, P.; Palama, T.; Gales, L.; Delhommeau, F.; Garmy-Susini, B. H.; Portais, J. C.; Vergez, F.; Selak, M.; Danet-Desnoyers, G.; Carroll, M.; Recher, C.; Sarry, J. E. Chemotherapy-Resistant Human Acute Myeloid Leukemia Cells Are Not Enriched for Leukemic Stem Cells but Require Oxidative Metabolism. Cancer Discovery 2017, 7, 716-735.

  • (14) Zhang, L.; Yao, Y. X.; Zhang, S. J.; Liu, Y.; Guo, H.; Ahmed, M.; Bell, T.; Zhang, H.; Han, G. C.; Lorence, E.; Badillo, M.; Zhou, S. H.; Sun, Y. T.; Di Francesco, M. E.; Feng, N. P.; Haun, R.; Lan, R.; Mackintosh, S. G.; Mao, X. Z.; Song, X. Z.; Zhang, J. H.; Pham, L. V.; Lorenzi, P. L.; Marszalek, J.; Heffernan, T.; Draetta, G.; Jones, P.; Futreal, A.; Nomie, K.; Wang, L. H.; Wang, M. Metabolic reprogramming toward oxidative phosphorylation identifies a therapeutic target for mantle cell lymphoma. Science Translational Medicine 2019, 11.

  • (15) Wheaton, W. W.; Weinberg, S. E.; Hamanaka, R. B.; Soberanes, S.; Sullivan, L. B.; Anso, E.; Glasauer, A.; Dufour, E.; Mutlu, G. M.; Budigner, G. S.; Chandel, N. S. Metformin inhibits mitochondrial complex I of cancer cells to reduce tumorigenesis. Elife 2014, 3, e02242.

  • (16) Dykens, J. A.; Jamieson, J.; Marroquin, L.; Nadanaciva, S.; Billis, P. A.; Will, Y. Biguanide-induced mitochondrial dysfunction yields increased lactate production and cytotoxicity of aerobically-poised HepG2 cells and human hepatocytes in vitro. Toxicol Appl Pharmacol 2008, 233, 203-10.

  • (17) Matsuzaki, S.; Humphries, K. M. Selective inhibition of deactivated mitochondrial complex I by biguanides. Biochemistry 2015, 54, 2011-21.

  • (18) Bridges, H. R.; Jones, A. J.; Pollak, M. N.; Hirst, J. Effects of metformin and other biguanides on oxidative phosphorylation in mitochondria. Biochem J 2014, 462, 475-87.

  • (19) Shi, Y.; Lim, S. K.; Liang, Q.; Iyer, S. V.; Wang, H. Y.; Wang, Z.; Xie, X.; Sun, D.; Chen, Y. J.; Tabar, V.; Gutin, P.; Williams, N.; De Brabander, J. K.; Parada, L. F. Gboxin is an oxidative phosphorylation inhibitor that targets glioblastoma. Nature 2019, 567, 341-346.

  • (20) Molina, J. R.; Sun, Y.; Protopopova, M.; Gera, S.; Bandi, M.; Bristow, C.; McAfoos, T.; Morlacchi, P.; Ackroyd, J.; Agip, A. A.; Al-Atrash, G.; Asara, J.; Bardenhagen, J.; Carrillo, C. C.; Carroll, C.; Chang, E.; Ciurea, S.; Cross, J. B.; Czako, B.; Deem, A.; Daver, N.; de Groot, J. F.; Dong, J. W.; Feng, N.; Gao, G.; Gay, J.; Do, M. G.; Greer, J.; Giuliani, V.; Han, J.; Han, L.; Henry, V. K.; Hirst, J.; Huang, S.; Jiang, Y.; Kang, Z.; Khor, T.; Konoplev, S.; Lin, Y. H.; Liu, G.; Lodi, A.; Lofton, T.; Ma, H.; Mahendra, M.; Matre, P.; Mullinax, R.; Peoples, M.; Petrocchi, A.; Rodriguez-Canale, J.; Serreli, R.; Shi, T.; Smith, M.; Tabe, Y.; Theroff, J.; Tiziani, S.; Xu, Q.; Zhang, Q.; Muller, F.; DePinho, R. A.; Toniatti, C.; Draetta, G. F.; Heffernan, T. P.; Konopleva, M.; Jones, P.; Di Francesco, M. E.; Marszalek, J. R. An inhibitor of oxidative phosphorylation exploits cancer vulnerability. Nat Med 2018, 24, 1036-1046.

  • (21) Salmond, R. J. mTOR Regulation of Glycolytic Metabolism in T Cells. Frontiers in Cell and Developmental Biology 2018, 6.

  • (22) Diskin, C.; Palsson-McDermott, E. M. Metabolic Modulation in Macrophage effector Function. Frontiers in Immunology 2018, 9.

  • (23) Kumar, V.; Patel, S.; Tcyganov, E.; Gabrilovich, D. I. The Nature of Myeloid-Derived Suppressor Cells in the Tumor Microenvironment. Trends in Immunology 2016, 37, 208-220.

  • (24) Vats, D.; Mukundan, L.; Odegaard, J. I.; Zhang, L.; Smith, K. L.; Morel, C. R.; Wagner, R. A.; Greaves, D. R.; Murray, P. J.; Chawla, A. Oxidative metabolism and PGC-lbeta attenuate macrophage-mediated inflammation. Cell Metab 2006, 4, 13-24.

  • (25) Beier, U. H.; Angelin, A.; Akimova, T.; Wang, L.; Liu, Y.; Xiao, H.; Koike, M. A.; Hancock, S. A.; Bhatti, T. R.; Han, R.; Jiao, J.; Veasey, S. C.; Sims, C. A.; Baur, J. A.; Wallace, D. C.; Hancock, W. W. Essential role of mitochondrial energy metabolism in Foxp3(+) T-regulatory cell function and allograft survival. FASEB J 2015, 29, 2315-26.

  • (26) Gatza, E.; Wahl, D. R.; Opipari, A. W.; Sundberg, T. B.; Reddy, P.; Liu, C.; Glick, G. D.; Ferrara, J. L. Manipulating the bioenergetics of alloreactive T cells causes their selective apoptosis and arrests graft-versus-host disease. Sci Transl Med 2011, 3, 67ra8.

  • (27) Glick, G. D.; Rossignol, R.; Lyssiotis, C. A.; Wahl, D.; Lesch, C.; Sanchez, B.; Liu, X.; Hao, L. Y.; Taylor, C.; Hurd, A.; Ferrara, J. L.; Tkachev, V.; Byersdorfer, C. A.; Boros, L.; Opipari, A. W. Anaplerotic metabolism of alloreactive T cells provides a metabolic approach to treat graft-versus-host disease. J Pharmacol Exp Ther 2014, 351, 298-307.

  • (28) Wahl, D. R.; Petersen, B.; Warner, R.; Richardson, B. C.; Glick, G. D.; Opipari, A. W. Characterization of the metabolic phenotype of chronically activated lymphocytes. Lupus 2010, 19, 1492-501.


Claims
  • 1. A compound described by Formula 1, including all stereoisomers, pharmaceutically acceptable salts (e.g., 2,2,2-trifluoroacetate (TFA) salts and other salts) (e.g., physiologically tolerated acid addition salts), polymorphs, solvates, isotopes, and/or prodrugs thereof,
  • 2. The compound of Formula 1-1, including all stereoisomers, pharmaceutically acceptable salts (e.g., 2,2,2-trifluoroacetate (TFA) salts and other salts) (e.g., physiologically tolerated acid addition salts), polymorphs, solvates, isotopes, and/or prodrugs thereof,
  • 3. The compound of Formula 1-1-1, including all stereoisomers, pharmaceutically acceptable salts (e.g., 2,2,2-trifluoroacetate (TFA) salts and other salts) (e.g., physiologically tolerated acid addition salts), polymorphs, solvates, isotopes, and/or prodrugs thereof;
  • 4. The compound of Formula 1-1-2, including all stereoisomers, pharmaceutically acceptable salts (e.g., 2,2,2-trifluoroacetate (TFA) salts and other salts) (e.g., physiologically tolerated acid addition salts), polymorphs, solvates, isotopes, and/or prodrugs thereof,
  • 5. The compound of Formula 1-2, including all stereoisomers, pharmaceutically acceptable salts (e.g., 2,2,2-trifluoroacetate (TFA) salts and other salts) (e.g., physiologically tolerated acid addition salts), polymorphs, solvates, isotopes, and/or prodrugs thereof;
  • 6. The compound of Formula 1-3, including all stereoisomers, pharmaceutically acceptable salts (e.g., 2,2,2-trifluoroacetate (TFA) salts and other salts) (e.g., physiologically tolerated acid addition salts), polymorphs, solvates, isotopes, and/or prodrugs thereof;
  • 7. The compound as shown in Table 1.
  • 8. A compound as shown in Table 1, including all stereoisomers, pharmaceutically acceptable salts (e.g., 2,2,2-trifluoroacetate (TFA) salts and other salts) (e.g., physiologically tolerated acid addition salts), polymorphs, solvates, isotopes, and/or prodrugs thereof.
  • 9. The compound of claim 1, claim 2, claim 3, claim 4, claim 5, claim 6, or claim 8, wherein the compound is comprised within a pharmaceutical composition.
  • 10. A method of treating, ameliorating, or preventing a hyperproliferative condition and/or autoimmune diseases, inflammatory diseases, cardiovascular diseases, neurodegenerative diseases, comprising administering to a patient a therapeutically effective amount of the pharmaceutical composition of claim 9.
  • 11. The method of claim 10, wherein the hyperproliferative condition is diabetes and/or cancer.
  • 12. The method of claim 11, wherein the cancer is one or more of leukemia, lymphoma, colon cancer, CNS cancer, lung cancer, melanoma, pancreatic cancer, ovarian cancer, renal cancer, breast cancer, prostate cancer, esophageal cancer, cervical cancer and colorectal cancer.
  • 13. The method of claim 11, further comprising administering to said patient one or more anticancer agents, wherein said anticancer agent one or more of a chemotherapeutic agent, and radiation therapy.
  • 14. The method of claim 10, wherein the patient is a human patient.
  • 15. The method of claim 10, wherein administration of the compound results in inhibition of mitochondria function within cancer cells and/or immune cells.
  • 16. The method of claim 10, wherein administration of the compound results in oxidative phosphorylation within cancer cells and/or immune cells.
  • 17. The method of claim 10, wherein administration of the compound results in activating gene expression within one or more of the genes listed in Table 3-12 within cancer cells and/or immune cells.
  • 18. The method of claim 10, wherein administration of the compound results in activating gene expression of one or more of ARG2, KLHL24, ANKRD37, FOXO6, and RNF122 within cancer cells and/or immune cells.
  • 19. The method of claim 10, wherein administration of the compound results in de-activating gene expression of one or more of ARRDC4, SCARNA5, INTS5, G0S2, and HLF.
  • 20. The method of claim 10, wherein administration of the compound results in up-regulating glycerol kinase, serine protease 23, nuclear receptor subfamily 2 group F member 6, CKLF-like MARVEL transmembrane domain-containing protein 4 and Type 1 phosphatidylinositol 4,5-bisphosphate 4-phosphatase expression within cancer cells and/or immune cells.
  • 21. The method of claim 10, wherein administration of the compound results in down-regulating dihydrolipoyl dehydrogenase, histone H1.3, zinc finger and BTB domain-containing protein 21, secretory carrier-associated membrane protein 2 and dolichyldiphosphatase 1 expression within cancer cells and/or immune cells.
  • 22. A method of inhibiting mitochondria function within cancer cells and/or immune cells comprising exposing such cells to a compound of claim 1, claim 2, claim 3, claim 4, claim 5, claim 6, or claim 9 or a pharmaceutical composition of claim 10.
  • 23. A method of inhibiting oxidative phosphorylation within cancer cells and/or immune cells comprising exposing such cells to a compound of claim 1, claim 2, claim 3, claim 4, claim 5, claim 6, or claim 9 or a pharmaceutical composition of claim 10.
  • 24. A method of activating gene expression within one or more of the genes listed in Table 3-12 within cancer cells and/or immune cells comprising exposing such cells to a compound of claim 1, claim 2, claim 3, claim 4, claim 5, claim 6 or claim 9 or a pharmaceutical composition of claim 10.
  • 25. A method of activating gene expression of one or more ARG2, KLHL24, ANKRD37, FOXO6, and RNF122 within cancer cells and/or immune cells comprising exposing such cells to a compound of claim 1, claim 2, claim 3, claim 4, claim 5, claim 6 or claim 9 or a pharmaceutical composition of claim 10.
  • 26. A method of de-activating gene expression of one or more of ARRDC4, SCARNA5, INTS5, G0S2, and HLF comprising exposing such cells to a compound of claim 1, claim 2, claim 3, claim 4, claim 5, claim 6 or claim 9 or a pharmaceutical composition of claim 10.
  • 27. The method of claim 10, wherein administration of the compound results in up-regulating glycerol kinase, serine protease 23, nuclear receptor subfamily 2 group F member 6, CKLF-like MARVEL transmembrane domain-containing protein 4 and Type 1 phosphatidylinositol 4,5-bisphosphate 4-phosphatase expression within cancer cells and/or immune cells comprising exposing such cells to a compound of claim 1, claim 2, claim 3, claim 4, claim 5, claim 6 or claim 9, or a pharmaceutical composition of claim 10.
  • 28. The method of claim 10, wherein administration of the compound results in down-regulating dihydrolipoyl dehydrogenase, histone H1.3, zinc finger and BTB domain-containing protein 21, secretory carrier-associated membrane protein 2 and dolichyldiphosphatase 1 expression within cancer cells and/or immune cells comprising exposing such cells to a compound of claim 1, claim 2, claim 3, claim 4, claim 5, claim 6 or claim 9 or a pharmaceutical composition of claim 10.
  • 29. A kit comprising a compound of claim 1, claim 2, claim 3, claim 4, claim 5, claim 6 or claim 9 or a pharmaceutical composition of claim 10 and instructions for administering said compound to a patient having a hyperproliferative condition and/or autoimmune diseases, inflammatory diseases, cardiovascular diseases, neurodegenerative diseases.
CROSS-REFERENCE TO RELATED APPLICATION

The present application claims priority to U.S. Provisional Patent Application No 63/029,979 filed May 26, 2020, which is hereby incorporated by reference in its entirety.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH

This invention was made with government support under Grant No. CA188252 awarded by the National Institutes of Health. The government has certain rights in the invention.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2021/034052 5/25/2021 WO
Provisional Applications (1)
Number Date Country
63029979 May 2020 US