Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
2-DIARYLMETHYL-4-AMINOTETRAHYDROPYRAN SULFONIMIDAMIDES AS ANTICANCER, ANTIINFLAMMATORY, ANTIFIBROTIC AND NEUROPROTECTIVE AGENTS
Document Type and Number:
WIPO Patent Application WO/2024/040045
Kind Code:
A2
Abstract:
A genus of 2-diarylmethyl-4-aminotetrahydropyran derivatives is disclosed, which includes the following genus: The compounds activate cellular PP2A, suppress oncogenic kinase signaling and negatively regulate MYC and MYCN in cancer. The compounds also restrain transcription by activating PP2A-Integrator-RNAPII, and this activity also contributes the tumor suppressor property of PP2A in transcriptionally addicted cancers The compounds also induce FOXO transcription factor translocation to the nucleus by modulating PP2A and, as a consequence, exhibit anti-proliferative effects. They are useful in the treatment of a variety of disorders, including as a monotherapy in cancer treatment, or used in combination with other drugs to restore sensitivity to chemotherapy where resistance has developed.

Inventors:
OHLMEYER MICHAEL (US)
Application Number:
PCT/US2023/072200
Publication Date:
February 22, 2024
Filing Date:
August 15, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ATUX ISKAY LLC (US)
International Classes:
C07D309/08; A61K31/351
Attorney, Agent or Firm:
HEUBERGER, Benjamin D. (US)
Download PDF:
Claims:
CLAIMS What is claimed is: 1. A compound, having a formula: or a pharmaceuticall , wherein: B is absent or is selected from a direct bond, -CH2CH2-, -CH=CH-, O, S, or -NRB- C(O)-; Q is selected from -O- or NRQ; Rim is selected from hydrogen or lower alkyl; U, V and W are independently cabocyclic aromatic or heteroaromatic rings; n = 0 or 1; X1, X2, X3 and X4 are independently selected in each instance from hydrogen, halogen, nitro, cyano, (C1-C6)alkyl optionally substituted with -OH, (C1-C6)haloalkyl, (C1- C6)haloalkoxy, (C1-C6)haloalkylthio, -NR1R2, -OR1, -C(O)R1, -OC(O)R1, -C(O)NR1R2, - C(O)OR1, -SR1, -SO2R1, and -SO2NR1R2; Y is selected from hydrogen or hydroxyl; Z1 and Z2 are independently selected in each instance from hydrogen, halogen, nitro, cyano, azido, (C1-C6)alkyl, (C1-C6)haloalkyl, (C1-C6)haloalkoxy, (C1-C6)haloalkylthio, - NR1R2, -NR1C(O)R2, -NR1C(O)OR3, -OR1, -C(O)R1, -OC(O)R1, -C(O)NR1R2, -C(O)OR1, - SR1, -SO2R1, -SO2NR1R2, and five membered heterocyclyl; RB is selected from H, or lower alkyl RQ is selected from H, optionally substituted lower alkyl, aryl R1, R2 and R3 are lower alkyl.

2. The compound of claim 1, wherein each instance of lower alkyl is, independently, selected from C1-3 alkyl. 3. The compound of claim 1, having a formula: or a pharmaceuticall wherein J1 is H, F, Cl, Br, or I; J2 is H, F, Cl, Br, or I; J3 is H, F, Cl, Br, or I; J4 is H, F, Cl, Br, or I; J5 is H or OH; J6 is H or C1-6 alkyl; J7 is H, F, Cl, Br, I, CN, N3, C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl; J8 is H, F, Cl, Br, I, CN, N3, C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl; and J9 is H and J10 is H, or J9 and J10 together form an ethylene or ethenylene. 4. The compound of claim 3, wherein J1 is H or F; J2 is H or F; J3 is H or F; J4 is H or F; J5 is H; J6 is H or C1-3 alkyl; J7 is H, C1-6 alkoxyl, or C1-6 haloalkoxyl; and J8 is H, C1-6 alkoxyl, or C1-6 haloalkoxyl. 5. The compound of claim 3, having a formula: or a pharmaceuticall wherein J2 is H or F; J3 is H or F; J6 is H or CH3; and J8 is OCH3 or OCF3. 6. The compound of claim 3, having a formula: or a pharmaceuticall wherein J2 is H or F; J3 is H or F; J6 is H or CH3; and J8 is OCH3 or OCF3. 7. The compound of claim 3, selected from , F F F F3 , or a p 8. The compound of claim 3, selected from , or a pha 9. The compound of claim 3, selected from , F F , or a 10. A composition, comprising the compound of one of claims 1–9. 11. The composition of claim 10, which is a pharmaceutical composition including a pharmaceutically acceptable carrier. 12. A method, comprising administering the compound of one of claims 1–9, or the compositon of one of claims 10–11, to a subject. 13. A method of modulating a protein phosphatase 2A, comprising contacting the protein phosphatase 2A with the compound of one of claims 1–9. 14. A method of treating a protein phosphatase 2A related disease, comprising administering an effective amount of the compound of one of claims 1–9, or the composition of one of claims 10–11, to a subject in need thereof. 15. A method of treating a disease, comprising administering an effective amount of the compound of one of claims 1–9, or the composition of one of claims 10–11, to a subject in need thereof, wherein the disease comprises one or more of: (a) cancer; (b) diabetes; (c) autoimmune disease; (d) age onset proteotoxic disease; (e) mood disorder; (f) acne vulgaris; (g) solid organ transplant rejection; (h) pulmonary disease; (i) cardiac hypertrophy; (j) viral infection; (k) an inflammatory condition; (l) heart failure; or (m) parasitic infection. 16. A method for restoring sensitivity to one or more chemotherapeutic agents in the treatment of cancer, comprising administering an effective amount of the compound of one of claims 1–9, or the composition of one of claims 10–11, to a subject in need thereof. 17. A method for treating a disease or disorder in a patient where the disease or disorder involves the dysregulation of the PI3K-AKT-FOXO signaling pathway, comprising administering an effective amount of the compound of one of claims 1–9, or the composition of one of claims 10–11, to a subject in need thereof. 18. A method for treating a disease or disorder in a patient where the disease or disorder involves the dysregulation of a Myc dependent signaling pathway, comprising administering an effective amount of the compound of one of claims 1–9, or the composition of one of claims 10–11, to a subject in need thereof. 19. A method for treating a metabolic or neurological disease or disorder in a patient wherein the disease or disorder involves the dysregulation of the mTOR-PP2A signaling axis, comprising administering an effective amount of the compound of one of claims 1–9, or the composition of one of claims 10–11, to a subject in need thereof. 20. A kit, comprising the compound of one of claims 1–9, or the composition of one of claims 10–11, and instructions for use. 21. A method, comprising one or more synthetic steps for preparing the compound of one of claims 1–9.

Description:
2-DIARYLMETHYL-4-AMINOTETRAHYDROPYRAN SULFONIMIDAMIDES AS ANTICANCER, ANTIINFLAMMATORY, ANTIFIBROTIC AND NEUROPROTECTIVE AGENTS RELATED APPLICATIONS [0001] This application claims priority of U.S. Provisional Patent Application No. 63/398,077, filed August 15, 2022, the entire content of which is incorporated herein by reference. FIELD OF THE INVENTION [0002] The invention relates to small molecule modulators of PP2A, comprising 2- diarylmethyl-4-aminotetrahydropyran sulfonimidamides, and their use to treat diseases such as cancer, inflammatory and autoimmune conditions, fibrotic and neurodegenerative diseases. BACKGROUND [0003] Protein phosphatase 2A is one of the four major serine threonine phosphatases and is implicated in the negative control of cell growth and division. Protein phosphatase 2A holoenzymes are composed of a structural subunit A and a catalytic subunit C which form a catalytically competent PP2A AC heterodimer. PP2A AC heterodimers then associate with a regulatory B subunit, which controls substrate specificity. Regulatory B subunits occur in four families and their expression is cell-type and context dependent. The PP2A heterotrimeric protein phosphatase is a ubiquitous and conserved phosphatase with diverse cellular functions. Among the targets of PP2A of the classical B-subunit containing holoenzymes are proteins of oncogenic signaling cascades, such as Raf, MEK, and AKT, and in this role they function as tumor suppressors. [0004] Recently PP2A AC heterodimers have been shown to interact directly with the INTS6 subunit of the Integrator Complex without any B subunit. Integrator associates with RNA Polymerase II (RNAPII) to control it’s transcriptional activity and PP2A AC associated Integrator acts to maintain RNAPII in a paused (i.e. transcriptionally stalled) state. This non- classical PP2A-Integrator complex thus functions to restrain transcription and this activity also contributes the tumor suppressor property of PP2A in transcriptionally addicted cancers, for example breast, lung, prostate, ovarian cancers glioblastoma, melanoma, leukemia, and others in adults. Also in pediatric cancers such as neuroblastoma and medulloblastoma in children and infants. [0005] Myc proteins (c-myc, Mycn and Mycl) target proliferative and apoptotic pathways vital for progression in cancer and it is overexpressed and deregulated in many human cancers. The control of Myc abundance through protein degradation has attracted considerable interest and Ser-62 phosphorylation by a number of kinases has been shown to stabilize the protein. PP2A is responsible for Ser-62 dephosphorylation which primes the protein for ubiquitylation and degredation, thus PP2A functions as a negative regulator of Myc. [0006] The FOXO (Forkhead transcription factors, Class O) proteins are a group of transcription factors involved in control of a variety of physiological, metabolic and developmental pathways. They are downstream effectors in a number of signaling pathways including insulin and growth factor signaling; they are also regulated by oxidative stress and nutrient deprivation. Cellular processes affected by FOXO activity include cell cycle control, differentiation, proliferation, apoptosis and autophagy. Disregulation of FOXO mediated processes has been implicated in a number of pathologies including tumorigenesis, inflammation, fibrosis, diabetes, neurodegenerative and other age related conditions, amongst others. Activity of FOXO transcription factors are controlled in part by their sub-cellular localization, in particular their localization to the nucleus from the cytosol, and their subsequent transcriptional activation. [0007] FOXO1 regulates expression of a number of genes that play critical roles in cell cycle and apoptosis. A pivotal regulatory mechanism of FOXO is reversible phosphorylation, catalyzed by kinases and phosphatases. Phosphorylation of FOXO1 is associated with 14-3-3 binding and cytosolic localization, whereas dephosphorylated FOXO1 translocates to the nucleus and is transcriptionally active. FOXO3 is regulated in an analogous manner. [0008] PP2A interacts directly with FOXO1 and dephosphorylates FOXO1. Inhibition of PP2A phosphatases rescues FOXO1-mediated cell death by regulating the level of the pro- apoptotic protein BIM. In addition, PP2A directly regulates FOXO3a subcellular localization and transcriptional activation. Without wishing to be held to any particular theory, it may be that the compounds described herein promote apoptosis by acting on FOXO transcription factors via activation of PP2A. [0009] Prostate cancer is the second leading cause of cancer death in men in America, behind lung cancer. According to the American Cancer Society, approximately 1 man in 36 will die of prostate cancer. Male hormones, specifically testosterone, fuel the growth of prostate cancer. By reducing the amount and activity of testosterone, the growth of advanced prostate cancer is slowed. Endocrine therapy, known as androgen ablation, is the first line of treatment for metastatic prostate cancer. Androgen deprivation therapy for metastatic prostate cancer results in tumor regression and symptomatic improvement in the majority of patients. However, metastatic prostate cancer inevitably progresses despite castrate levels of serum testosterone. Several new therapies have been approved for patients with castration-resistant prostate cancer (CRPC); however, none are curative and tumors ultimately develop resistance. To combat CRPC new approaches and novel therapies are required.

[0010] Breast cancer can affect both men and women. Breast cancer is the most prevalent cancer in women, after skin cancers, with about 1 in every 8 women expected to develop invasive breast cancer at some point. One subset of breast cancer expresses the androgen receptor (AR), which has been implicated as a therapeutic target in that subset. About 10- 20% of breast cancers — more than one out of every 10 — are found to be triple-negative. "Triple negative breast cancer" refers to a breast cancer that does not contain estrogen receptors, progesterone receptors, or human epidermal growth factor receptor 2 (HER2). This means that the growth of the cancer is not supported by the hormones estrogen and progesterone, nor by the presence of too many HER2 receptors. Therefore, triple-negative breast cancer does not respond to hormonal therapy (such as tamoxifen or aromatase inhibitors) or therapies that target HER2 receptors, such as Herceptin (chemical name: trastuzumab). While these tumors are often treatable, the chemotherapy is not targeted, and response durations are short. For doctors and researchers, there is intense interest in finding new medications that can treat breast cancer.

[0011] The compounds described herein, which are 2-diarylmethyl-4- aminotetrahydropyran sulfonimidamides, exhibit anti-proliferative effects by increasing PP2A activity in cells with suppressed or deficient phosphatase activity and are therefore useful as monotherapy in cancer treatment. Additionally, they can be used in combination with other drugs to restore sensitivity to chemotherapy where resistance has developed. The compounds may also be used in treatment of other diseases characterized by deficient PP2A activity, for example lung fibrosis and Alzheimer’s Disease. SUMMARY OF THE INVENTION [0012] A genus of 2-diarylmethyl-4-aminotetrahydropyran sulfonimidamide and related compounds has now been found that modulate PP2A activity. The compounds deactivate pro- growth and pro-survival kinases such as phospo-ERK and phospho-AKT by promoting their deposphorylation by PP2A; they destabilize oncogenic MYC by promoting PP2A mediated dephoshorylation of MYC, and they promote RNAPII promoter proximal pausing during transcription. The compounds described herein exhibit anti-proliferative effects, and are useful in the treatment of a variety of disorders, including as a monotherapy in cancer treatment, or used in combination with other drugs to restore sensitivity to chemotherapy where resistance has developed. [0013] In a first aspect the invention relates to compounds of formula (I): wherein: B is absent or is selected from a direct bond, -CH 2 CH 2 -, -CH=CH-, O, S, or -NR B - C(O)-; Q is selected from -O- or NR Q ; R im is selected from H or lower alkyl U, V and W are independently cabocyclic aromatic or heteroaromatic rings; n = 0 or 1; X 1 , X 2 , X 3 and X 4 are independently selected in each instance from hydrogen, halogen, nitro, cyano, (C1-C6)alkyl optionally substituted with -OH, (C1-C6)haloalkyl, (C1- C 6 )haloalkoxy, (C 1 -C 6 )haloalkylthio, -NR 1 R 2 , -OR 1 , -C(O)R 1 , -OC(O)R 1 , -C(O)NR 1 R 2 , - C(O)OR 1 , -SR 1 , -SO2R 1 , and -SO2NR 1 R 2 ; Y is H or hydroxyl Z 1 and Z 2 are independently selected in each instance from hydrogen, halogen, nitro, cyano, azido, (C1-C6)alkyl, (C1-C6)haloalkyl, (C1-C6)haloalkoxy, (C1-C6)haloalkylthio, - NR 1 R 2 , -NR 1 C(O)R 2 , -NR 1 C(O)OR 3 , -OR 1 , -C(O)R 1 , -OC(O)R 1 , -C(O)NR 1 R 2 , -C(O)OR 1 , - SR 1 , -SO2R 1 , -SO2NR 1 R 2 , and five membered heterocyclyl; R B is selected from H, or lower alkyl R Q is selected from H, optionally substituted lower alkyl, aryl R 1 , R 2 and R 3 are lower alkyl. [0014] In a second aspect, the invention relates to pharmaceutical compositions comprising the compounds described herein. [0015] In a third aspect, the invention relates to methods and uses of the above-described compounds in medicine, particularly for the treatment of a disease chosen from (a) cancer; (b) diabetes; (c) autoimmune disease; (d) age onset proteotoxic disease (particularly neurodegenerative disease); (e) mood disorder; (f) acne vulgaris; (g) solid organ transplant rejection (graft vs. host disease); (h) pulmonary disease (such as COPD or IPF); (i) cardiac hypertrophy and heart failure; (j) viral or parasitic infection; (k) inflammatory conditions (such as asthma) and (l) organ fibrosis (such as kidney fibrosis). These methods include administering to a patient a therapeutically effective amount of a compound described herein. [0016] In a fourth aspect, the invention relates to a method for restoring sensitivity to one or more chemotherapeutic agents in the treatment of cancer. The method includes administering an effective amount of a compound described herein. [0017] In a fifth aspect, the invention relates to a method for treating a disease or disorder in a patient where the disease or disorder involves the dysregulation of PP2A influenced signaling cascades such as the PI3K-AKT and MAP kinase pathways. These methods include administering to a patient a therapeutically effective amount of a compound described herein. [0018] In a sixth aspect, the invention relates to a method for treating a disease or disorder in a patient where the disease or disorder involves the dysregulation of a Myc dependent signaling pathway. These methods include administering to a patient a therapeutically effective amount of a compound described herein. [0019] In a seventh aspect, the invention relates to a method for treating a metabolic disease or disorder in a patient where the disease or disorder involves the dysregulation of the mTOR-PP2A signaling axis. The method includes administering an effective amount of a compound described herein. [0020] In an eighth aspect, the invention relates to a method for treating disease or disorder in a patient where the disease or disorder involves cellular hyperproliferation or growth due to dysregulation of the PP2A-Integrator-RNAPII axis. BRIEF DESCRIPTION OF THE DRAWINGS [0021] Fig.1 depicts a general synthetic scheme for preparing compounds described herein, and intermediates useful therefor. [0022] Fig. 2 depicts a synthetic scheme for preparing Example 1, and intermediates useful therefor. [0023] Fig. 3 depicts a synthetic scheme for preparing Example 3 and Example 4, and intermediates useful therefor. [0024] Fig. 4 depicts a synthetic scheme for preparing an amine intermediate useful for preparing Example 7, and intermediates useful therefor. [0025] Fig. 5 depicts another general synthetic scheme for preparing compounds described herein, and intermediates useful therefor. [0026] Fig. 6 depicts a synthetic scheme for preparing Example 7, and intermediates useful therefor. [0027] Fig. 7 depicts a synthetic scheme for preparing Example 8, and intermediates useful therefor. DETAILED DESCRIPTION OF THE INVENTION [0028] Substituents are generally defined when introduced and retain that definition throughout the specification and in all independent claims. [0029] In a composition aspect, the invention relates to compounds of formula (I):

as described ab [0030] In some embodiments, the invention relates to compounds of formula IIa, IIb, IIc or IId:

[0031] In addition ran ring the sulfur atom of the sulfonimidamide is also a stable stereogenic chiral center and all of the stereoisomers above, IIa, IIb, IIc and IId may occur as diastereoisomers with either configuration at the sulfur center: [0032] In the embodiments described herein, the compound may be of formula I, IIa, IIb, IIc, IId, IIe or IIf, unless otherwise indicated [0033] In some embodiments Q is -O- with n=1, a tetrahydropyran [0034] In some embodiments Q is -NR Q - with n=1, a piperidine [0035] In some embodiments U and V are carbocyclic aromatic [0036] In some embodiments Q is -O- with n=1, Y=H and R im =H, where U and V are carbocyclic aromatic . . [0038] In some em bodiments, B is absent. These embodiments are diarylmethyl compounds:

. [0039] In other emb , oheptane compounds: . [0040] In still furt her embodiments where Q is -O-, B is -CH 2 CH 2 -, W is carbocyclic aromatic: . [0041] In some embodiments, X 1 , X 2 , X 3 , and X 4 are independently selected in each instance from hydrogen, halogen, nitro, cyano, (C1-C6)alkyl optionally substituted with -OH, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )haloalkylthio, -NR 1 R 2 , -OR 1 , -C(O)R 1 , - OC(O)R 1 , -C(O)NR 1 R 2 , -C(O)OR 1 , -SR 1 , -SO2R 1 , and -SO2NR 1 R 2 . In other embodiments, X 2 and X 4 are each hydrogen. In still other embodiments, X and X are each hydrogen, and X and X 3 are each chosen independently from -H, -F, -Cl, -CF 3 ,-C(CH 3 ) 2 OH, or -C(O)NMe 2 . In further embodiments, all of X 1 , X 2 , X 3 and X 4 are each hydrogen. In yet other embodiments, at least one of X 1 , X 2 , X 3 and X 4 is located at a carbon two positions away from a bridgehead carbon. [0042] In some embodiments, Z 1 and Z 2 are independently selected in each instance from hydrogen, halogen, nitro, cyano, azido, (C 1 -C 6 )alkyl, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C1-C6)haloalkylthio, -NR 1 R 2 , -NR 1 C(O)R 2 , -NR 1 C(O)OR 6 , -OR 1 , -C(O)R 1 , -OC(O)R 1 , - C(O)NR 1 R 2 , -C(O)OR 1 , -SR 1 , -SO 2 R 1 , -SO 2 NR 1 R 2 , and five membered heterocyclyl. In other embodiments, Z 1 is H. In still other embodiments, Z 2 is chosen from hydrogen, halogen, and (C1-C6)haloalkoxy. In yet other embodiments, Z 2 is chosen from hydrogen, F, Cl, and OCF3. In further embodiments, Z 2 is in the para position. [0043] In some embodiments: B is absent or -CH2CH2-; R im is selected from H or Me; Q is selected from -O- with n=1 or 0; U, V and W are carbocyclic aromatic; X 2 and X 4 are each hydrogen, and X 1 and X 3 are each chosen independently from -H, -F, -Cl, -CF 3 ,-C(CH 3 ) 2 OH, -C(O)NMe 2 ; Y is H Z 1 is hydrogen; and Z 2 is selected in each instance from hydrogen, halogen, trifluoromethyl and (C1- C6)haloalkoxy. [0044] The compounds described herein contain asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms which may be defined in terms of absolute stereochemistry as (R)- or (S)-. The present invention is meant to include all such possible diastereomers as well as their racemic and optically pure forms. Optically active (R)- and (S)- isomers may be prepared using homo-chiral synthons or homo-chiral reagents. Final compounds or intermediates may be resolved in to their enantiomers using conventional techniques such crystallization of amine intermediates with chiral acids or preparative chiral chromatography on final compounds or intermediates. When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended to include both (E)- and (Z) geometric isomers. Likewise, all tautomeric forms are intended to be included. [0045] The graphic representations of racemic, ambiscalemic and scalemic or enantiomerically pure compounds used herein are a modified version of the denotations taken from Maehr J. Chem. Ed. 62, 114-120 (1985): simple lines provide no information about stereochemistry and convey only connectivity; solid and broken wedges are used to denote the absolute configuration of a chiral element; solid and broken bold lines are geometric descriptors indicating the relative configuration shown but not necessarily denoting racemic character; and wedge outlines and dotted or broken lines denote enantiomerically pure compounds of indeterminate absolute configuration. For example, the graphic representations: nd indicate each single absolute stereochemistry of the stereogenic carbon centers, with the sulfur chirality undefined. For the purpose of the present disclosure, a “pure” or “substantially pure” enantiomer is intended to mean that the enantiomer is at least 95% of the configuration shown and 5% or less of other enantiomers. The graphic representation:

indicates a unknown bon centers, i.e., it could be either of the two structures shown above, as a substantially pure single stereochemistry. The graphic representations with bold solid and broken lines represent relative stereochemistry in racemic compounds. Thus racemic cis diastereoisomer with respect to the central ring: And racemic trans diastereoisomer with respect to the central ring: And, finally, the structure: conveys no inform ure could be a single enantiomer or a mixture of enantiomers, including a racemic micture, or mixture of diastereoisomers. [0046] In any of these possiblities, compounds can be single cis enantiomers of formula IIa or formula IIb or single trans enantiomers formula IIc or formula IId, or a mixture of the two. If a mixture, the mixture will most commonly be racemic, but it need not be. Substantially pure single enantiomers of biologically active compounds such as those described herein often exhibit advantages over their racemic mixture. [0047] Various non-limiting embodiments of the present disclosure are described below. [0048] Thus, in some embodiments, provided herein are compounds of formula (I): wherein: B is absent or is selected from a direct bond, -CH2CH2-, -CH=CH-, O, S, or - NR B -C(O)-; Q is selected from -O- or NR Q ; R im is selected from H or lower alkyl; U, V and W are independently cabocyclic aromatic or heteroaromatic rings; n = 0 or 1; X1, X2, X3 and X4 are independently selected in each instance from hydrogen, halogen, nitro, cyano, (C 1 -C 6 )alkyl optionally substituted with -OH, (C 1 -C 6 )haloalkyl, (C1-C6)haloalkoxy, (C1-C6)haloalkylthio, -NR 1 R 2 , -OR 1 , -C(O)R 1 , -OC(O)R 1 , - C(O)NR 1 R 2 , -C(O)OR 1 , -SR 1 , -SO 2 R 1 , and -SO 2 NR 1 R 2 ; Y is selected from hydrogen or hydroxyl Z 1 and Z 2 are independently selected in each instance from hydrogen, halogen, nitro, cyano, azido, (C1-C6)alkyl, (C1-C6)haloalkyl, (C1-C6)haloalkoxy, (C1- C6)haloalkylthio, -NR 1 R 2 , -NR 1 C(O)R 2 , -NR 1 C(O)OR 3 , -OR 1 , -C(O)R 1 , -OC(O)R 1 , - C(O)NR 1 R 2 , -C(O)OR 1 , -SR 1 , -SO2R 1 , -SO2NR 1 R 2 , and five membered heterocyclyl; R B is selected from H, or lower alkyl R Q is selected from H, optionally substituted lower alkyl, aryl R 1 , R 2 and R 3 are lower alkyl. [0049] In some embodiments, the compound of formula I is a compound of formula IIa: . [0050] In some embodiments, the compound of formula I is a compound of formula IIb: . [0051] In some embodiments of the formulae herein, B is absent. [0052] In some embodiments of the formulae herein, B is -(CH2)2-. [0053] In some embodiments of the formulae herein, B is a direct bond. [0054] In some embodiments of the formulae herein, B is -S-. [0055] In some embodiments of the formulae herein, Q is -O- with n = 1. [0056] In some embodiments of the formulae herein, Q is -O- with n = 0. [0057] In some embodiments of the formulae herein, Q is -NR Q - with n = 1. [0058] In some embodiments of the formulae herein, one of U or V is carbocyclic. [0059] In some embodiments of the formulae herein, both of U and V are carbocyclic. [0060] In some embodiments of the formulae herein, R im is hydrogen. [0061] In some embodiments of the formulae herein, R im is methyl. [0062] In some embodiments of the formulae herein, W is carbocyclic. [0063] In some embodiments of the formulae herein, W is heteroaromatic. [0064] In some embodiments of the formulae herein, X 1 , X 2 , X 3 , and X 4 are independently selected in each instance from hydrogen, halogen, nitro, cyano, (C 1 -C 6 )alkyl optionally substituted with -OH, (C1-C6)haloalkyl, (C1-C6)haloalkoxy, (C1-C6)haloalkylthio, -NR 1 R 2 , - OR 1 , -C(O)R 1 , -OC(O)R 1 , -C(O)NR 1 R 2 , -C(O)OR 1 , -SR 1 , -SO 2 R 1 , and -SO 2 NR 1 R 2 . [0065] In some embodiments of the formulae herein, X 2 and X 4 are each hydrogen. [0066] In some embodiments of the formulae herein, X 1 and X 3 are each chosen independently from hydrogen, halogen, nitro, cyano, (C 1 -C 6 )alkyl optionally substituted with -OH, (C1-C6)haloalkyl, (C1-C6)haloalkoxy, (C1-C6)haloalkylthio, -NR 1 R 2 , -OR 1 , -C(O)R 1 , - OC(O)R 1 , -C(O)NR 1 R 2 , -C(O)OR 1 , -SR 1 , -SO 2 R 1 , and -SO 2 NR 1 R 2 . [0067] In some embodiments of the formulae herein, X 1 and X 3 are each chosen independently from -H, -F, -Cl, -CF 3 , -OMe, or -OCF3. [0068] In some embodiments of the formulae herein, all of X , X , X and X are each hydrogen. [0069] In some embodiments of the formulae herein, Z 1 and Z 2 are independently selected in each instance from hydrogen, halogen, nitro, cyano, azido, (C 1 -C 6 )alkyl, (C 1 -C 6 )haloalkyl, (C1-C6)haloalkoxy, (C1-C6)haloalkylthio, -NR 1 R 2 , -NR 1 C(O)R 2 , -NR 1 C(O)OR 6 , -OR 1 , - C(O)R 1 , -OC(O)R 1 , -C(O)NR 1 R 2 , -C(O)OR 1 , -SR 1 , -SO 2 R 1 , -SO 2 NR 1 R 2 and five membered hetercyclyl. [0070] In some embodiments of the formulae herein, Z 1 and Z 2 are independently selected in each instance from hydrogen, halogen, halo(C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, and halo(C 1 - C6)alkoxy. [0071] In some embodiments of the formulae herein, Z 1 is hydrogen. [0072] In some embodiments of the formulae herein, Z 2 is chosen from hydrogen, halogen, and (C1-C6)haloalkoxy. [0073] In some embodiments of the formulae herein, Z 2 is chosen from hydrogen, F, Cl, CF3, and trifluoromethoxy. [0074] In some embodiments of the formulae herein, Z 2 is trifluoromethoxy. [0075] In some embodiments of the formulae herein, one of Z 1 and Z 2 is para to the sulfonyl amide. [0076] In some embodiments of the formulae herein, Z 1 is hydrogen Z 2 is para to the sulfonyl amide. [0077] In some embodiments, the compounds are provided as a composition. In some embodiments, the composition is a pharmaceutical composition further comprising a pharmaceutically acceptable carrier. [0078] In some embodiments, provided herein are methods for treating a disease in a patient, wherein the disease is chosen from: (a) cancer (b) diabetes (c) autoimmune disease, such as rheumatoid arthritis or multiple sclerosis (d) age onset proteotoxic disease, particularly neurodegenerative disease (e) mood disorder (f) acne vulgaris (g) solid organ transplant rejection (graft vs. host disease) (h) pulmonary disease, such as COPD or pulmonary fibrosis (i) cardiac hypertrophy and heart failure (j) viral or parasitic infection and (k) inflammatory conditions, such as asthma; the method comprising administering to the patient a therapeutically effective amount of a compound or composition provided herein. [0079] In some embodiments, said cancer is selected from the group consisting of: ovarian, endometrial, pancreatic, renal cell, breast, prostate, lung, hepatocellular carcinoma, glioma, leukemia, lymphoma, colorectal cancers, and sarcomas. [0080] In some embodiments, said cancer is chemotherapy resistant cancer. [0081] In some embodiments, the methods herein further comprise administering one or more additional cancer chemotherapeutic agents. [0082] In some embodiments, the methods or compounds herein are for treating an age onset proteotoxic disease, particularly neurodegenerative disease, such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and amyotrophic lateral sclerosis. [0083] In some embodiments, the methods or compounds herein are for treating a pulmonary disease. [0084] In some embodiments, the pulmonary disease is COPD, asthma, or pulmonary fibrosis. [0085] In some embodiments, the methods or compounds herein are for treating an inflammatory or autoimmune disease. [0086] In some embodiments, the inflammatory or autoimmune disease is multiple sclerosis. [0087] Also provided herein are methods for restoring sensitivity to one or more chemotherapeutic agents in the treatment of cancer, the method comprising administering an effective amount of a compound or composition provided herein. [0088] Also provided herein are methods for treating a disease or disorder in a patient where the disease or disorder involves the dysregulation of the PI3K-AKT-FOXO signaling pathway, the method comprising administering to the patient a therapeutically effective amount of a compound or composition provided herein. [0089] Also provided herein are methods for treating a disease or disorder in a patient where the disease or disorder involves the dysregulation of a Myc dependent signaling pathway, the method comprising administering to the patient a therapeutically effective amount of a compound or composition provided herein. [0090] Also provided herein are methods for treating a metabolic or neurological disease or disorder in a patient wherein the disease or disorder involves the dysregulation of the mTOR- PP2A signaling axis, the method comprising administering to the patient a therapeutically effective amount of a compound or composition provided herein. [0091] In some embodiments, provided herein are compounds having a formula: or a pharmaceutically acceptable salt thereof, wherein: B is absent or is selected from a direct bond, -CH 2 CH 2 -, -CH=CH-, O, S, or -NR B - C(O)-; Q is selected from -O- or NR Q ; R im is selected from hydrogen or lower alkyl; U, V and W are independently cabocyclic aromatic or heteroaromatic rings; n = 0 or 1; X1, X2, X3 and X4 are independently selected in each instance from hydrogen, halogen, nitro, cyano, (C 1 -C 6 )alkyl optionally substituted with -OH, (C 1 -C 6 )haloalkyl, (C 1 - C6)haloalkoxy, (C1-C6)haloalkylthio, -NR 1 R 2 , -OR 1 , -C(O)R 1 , -OC(O)R 1 , -C(O)NR 1 R 2 , - C(O)OR 1 , -SR 1 , -SO 2 R 1 , and -SO 2 NR 1 R 2 ; Y is selected from hydrogen or hydroxyl; Z 1 and Z 2 are independently selected in each instance from hydrogen, halogen, nitro, cyano, azido, (C1-C6)alkyl, (C1-C6)haloalkyl, (C1-C6)haloalkoxy, (C1-C6)haloalkylthio, - NR 1 R 2 , -NR 1 C(O)R 2 , -NR 1 C(O)OR 3 , -OR 1 , -C(O)R 1 , -OC(O)R 1 , -C(O)NR 1 R 2 , -C(O)OR 1 , - SR 1 , -SO2R 1 , -SO2NR 1 R 2 , and five membered heterocyclyl; R B is selected from H, or lower alkyl R Q is selected from H, optionally substituted lower alkyl, aryl R 1 , R 2 and R 3 are lower alkyl. [0092] In some embodiments of the compounds herein, each instance of lower alkyl is, independently, selected from C1-3 alkyl. [0093] In some embodiments, the compounds herein have a formula: or a pharmaceuticall wherein J 1 is H, F, Cl, Br, or I; J 2 is H, F, Cl, Br, or I; J 3 is H, F, Cl, Br, or I; J 4 is H, F, Cl, Br, or I; J 5 is H or OH; J 6 is H or C 1-6 alkyl; J 7 is H, F, Cl, Br, I, CN, N3 C16 alkyl C16 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl; J 8 is H, F, Cl, Br, I, CN, N3, C1-6 alkyl, C1-6 alkoxyl, C1-6 haloalkyl, C1-6 haloalkoxyl; and J 9 is H and J 10 is H, or J 9 and J 10 together form an ethylene or ethenylene. [0094] In some embodiments of the compounds herein, J 1 is H or F; J 2 is H or F; J 3 is H or F; J 4 is H or F; J 5 is H; J 6 is H or C 1-3 alkyl; J 7 is H, C1-6 alkoxyl, or C1-6 haloalkoxyl; and J 8 is H, C 1-6 alkoxyl, or C 1-6 haloalkoxyl. [0095] In some embodiments, provided herein are compounds having a formula: or a pharmaceutically wherein J 2 is H or F; J 3 is H or F; J 6 is H or CH 3 ; and J 8 is OCH3 or OCF3. [0096] In some embodiments, provided herein are compounds having a formula: or a pharmaceutically acceptable salt thereof, wherein J 2 is H or F; J 3 is H or F; J 6 is H or CH3; and J 8 is OCH 3 or OCF 3 . [0097] In some embodiments, provided herein are compounds having a formula: F 3 , or a [0098] In some embodiments, provided herein are compounds having a formula: , or a pha [0099] In some embodiments, provided herein are compounds having a formula: , , or a [00100] Also provided herein are pharmaceutical compositions comprising a compound disclosed above, or a pharmaceutically acceptable salt form thereof, and a pharmaceutically acceptable carrier or diluent. [00101] While it may be possible for the compounds of formula I to be administered as the raw chemical, it is preferable to present them as a pharmaceutical composition. According to a further aspect, the present invention provides a pharmaceutical composition comprising a compound of formula I or a pharmaceutically acceptable salt thereof, together with one or more pharmaceutically carriers thereof and optionally one or more other therapeutic ingredients. The carrier(s) must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. [00102] Formulations of the compounds and compositions described herein may be administered by a variety of methods: oral (including, but not limited to, capsules, cachets, tablets, powder, granules, solutions, suspensions, emulsions, tablets, or sublingual tablets), buccal, by inhalation (by using, for instance, an inhaler, a nebulizer, an aerosol, a gas, etc.), nasal, topical (including, but not limited to, lotions, creams, ointments, patches (i.e., transdermal), gels, liniments, pastes), ophthalmic, to the ear, rectal (for instance, by using a suppository or an enema), vaginal, or parenteral, depending on the severity and type of the disease being treated. In some embodiments, the compositions are administered orally or intravenously. The formulations include those suitable for oral, parenteral (including subcutaneous, intradermal, intramuscular, intracranial, intravenous and intraarticular), rectal, vaginal, nasal (inhalation), and topical (including dermal, buccal, sublingual and intraocular) administration. The most suitable route may depend upon the condition and disorder of the recipient. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing into association a compound of formula (I) or a pharmaceutically acceptable salt thereof ("active ingredient") with the carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation. [00103] Formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient may also be presented as a bolus, electuary or paste. [00104] A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, lubricating, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide sustained, delayed or controlled release of the active ingredient therein. [00105] Formulations for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient. Formulations for parenteral administration also include aqueous and non-aqueous sterile suspensions, which may include suspending agents and thickening agents. The formulations may be presented in unit-dose of multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of a sterile liquid carrier, for example saline, phosphate-buffered saline (PBS) or the like, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described. [00106] It will be recognized that the compounds of this invention can exist in radiolabeled form, i.e., the compounds may contain one or more atoms containing an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Radioisotopes of hydrogen, carbon, phosphorous, fluorine, and chlorine include 2 H, 3 H, 13 C, 14 C, 15 N, 35 S, 18 F, and 36 Cl, respectively. Compounds that contain those radioisotopes and/or other radioisotopes of other atoms are within the scope of this invention. Tritiated, i.e. 3 H, and carbon-14, i.e., 14 C, radioisotopes are particularly preferred for their ease in preparation and detectability. Compounds that contain isotopes 11 C, 13 N, 15 O and 18 F are well suited for positron emission tomography. Radiolabeled compounds of formula I of this invention and prodrugs thereof can generally be prepared by methods well known to those skilled in the art. Conveniently, such radiolabeled compounds can be prepared by carrying out the procedures disclosed in the Examples and Schemes by substituting a readily available radiolabeled reagent for a non-radiolabeled reagent. [00107] In some embodiments, provided herein are kits, comprising a compound or composition described herein, and instructions for use. [00108] In some embodiments, provided herein are methods comprising administering a compound or composition described herein to a subject. [00109] In some embodiments, provided herein are methods of modulating a protein phosphatase 2A, comprising contacting the protein phosphatase 2A with a compound or composition described herein. [00110] In some embodiments, provided herein are methods of treating a protein phosphatase 2A related disease, comprising administering an effective amount of a compound or composition described herein to a subject in need thereof. [00111] In some embodiments, provided herein are methods of treating a disease, comprising administering an effective amount of a compound or composition described herein to a subject in need thereof, wherein the disease comprises one or more of: (a) cancer; (b) diabetes; (c) autoimmune disease; (d) age onset proteotoxic disease; (e) mood disorder; (f) acne vulgaris; (g) solid organ transplant rejection; (h) pulmonary disease; (i) cardiac hypertrophy; (j) viral infection; (k) an inflammatory condition; (l) heart failure; or (m) parasitic infection. [00112] In some embodiments, provided herein are methods of restoring sensitivity to one or more chemotherapeutic agents in the treatment of cancer, comprising administering an effective amount of a compound or composition described herein to a subject in need thereof. [00113] In some embodiments, provided herein are methods of treating a disease or disorder in a patient where the disease or disorder involves the dysregulation of the PI3K-AKT-FOXO signaling pathway, comprising administering an effective amount of a compound or composition described herein to a subject in need thereof. [00114] In some embodiments, provided herein are methods of treating a disease or disorder in a patient where the disease or disorder involves the dysregulation of a Myc dependent signaling pathway, comprising administering an effective amount of a compound or composition described herein to a subject in need thereof. [00115] In some embodiments, provided herein are methods of treating a metabolic or neurological disease or disorder in a patient wherein the disease or disorder involves the dysregulation of the mTOR-PP2A signaling axis, comprising administering an effective amount of a compound or composition described herein to a subject in need thereof. [00116] The compounds provided herein can be used for treating cancer in a patient, the method comprising administering to the patient a therapeutically effective amount of a compound of formula I. In some embodiments, the cancer is characterized by dysregulation of the PI3K-AKT-FOXO signaling pathway. For example, the cancer can be selected from the group consisting of: ovarian, pancreatic, renal cell, breast, prostate, lung, hepatocellular carcinoma, glioma, leukemia, lymphoma, colorectal cancers, and sarcomas. [00117] In some embodiments, the cancer is chemotherapy resistant cancer. In some embodiments, the method further comprises administering one or more cancer chemotherapeutic agents. In some embodiments, the one or more cancer chemotherapeutic agents are EGFR inhibitors. [00118] In some embodiments, the cancer is chemotherapy resistant cancer. In some embodiments, the method further comprises administering one or more cancer chemotherapeutic agents targeting transcriptional dysregulation. In some embodiments, the one or more cancer chemotherapeutic agents are CDK inhibitors, particularly CDK9 and/or CDK7 inhibitors. [00119] In some embodiments, the cancer is chemotherapy resistant cancer. In some embodiments, the method further comprises administering one or more cancer chemotherapeutic agents. In some embodiments, the one or more cancer chemotherapeutic agents are mTOR inhibitors. [00120] Certain cancers are characterized by dysregulation and overactivation of cellular transcription carried out by RNA polymerase II (RNAPII) as described in Transcriptional Addiction in Cancer. Bradner, J. E., Hnisz, D. and Young, R. A. February 2017, Cell, Vol. 168, pp. 629-643. BRD4, a member of the bromodomain and extra-terminal domain (BET) family of epigenetic readers, occupies super-enhancers co-opted in transcriptionally addicted cancer cells, and recruits pTEFb to RNAPII to enable promoter proximal pause release and maintain productive elongation (see, for example, Control of Embryonic Stem Cell Identity by BRD4-Dependent Transcriptional Elongation of Super-Enhancer-Associated Pluripotency Genes. Di Micco, R., et al. October 2014, Cell Reports, Vol. 9, pp. 234-247). Therefore BRD4 inhibitor (BRD4i) treatment synergizes with PP2A activation in two ways, first by suppressing BRD4 mediated recruitment of pTEFb to paused RNAPII. A second mode of synergy with BRD4i treatment is by reversing the activating phosphorylation of BRD4 itself by PP2A complexes (see Phospho-BRD4: transcription plasticity and drug targeting. Chiang, C. 2016, Drug Discov Today: Technol, http://dx.doi.org/10.1016). The compounds provided herein can be used for treating cancer in a patient where there is transcriptional dysregulation, the method comprising administering to the patient a therapeutically effective amount of a compound of formula I and a BRD4 inhibitor. Cancers characterized by transcriptional addiction include Breast Cancer, Osteosarcoma, Endometrial Cancer, Acute Myeloid Leukemia, Lung Cancer, Prostate Cancer, Melanoma and Ovarian Cancer. [00121] In some embodiments, administration of a compound of formula I can restore sensitivity to one or more chemotherapeutic agents in a patient wherein the patient has developed a resistance to the one or more chemotherapeutic agents. More particularly, cancers that may be treated by the compounds, compositions and methods described herein include, but are not limited to, the following: cardiac cancers, including, for example sarcoma, e.g., angiosarcoma, fibrosarcoma, rhabdomyosarcoma, and liposarcoma; myxoma; rhabdomyoma; fibroma; lipoma and teratoma; lung cancers, including, for example, bronchogenic carcinoma, e.g., squamous cell, undifferentiated small cell, undifferentiated large cell, and adenocarcinoma; alveolar and bronchiolar carcinoma; bronchial adenoma; sarcoma; lymphoma; chondromatous hamartoma; and mesothelioma; gastrointestinal cancer, including, for example, cancers of the esophagus, e.g., squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, and lymphoma; cancers of the stomach, e.g., carcinoma, lymphoma, and leiomyosarcoma; cancers of the pancreas, e.g., ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, and vipoma; cancers of the small bowel, e.g., adenocarcinoma, lymphoma, carcinoid tumors, Kaposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, and fibroma; cancers of the large bowel, e.g., adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, and leiomyoma; genitourinary tract cancers, including, for example, cancers of the kidney, e.g., adenocarcinoma, Wilm's tumor (nephroblastoma), lymphoma, and leukemia; cancers of the bladder and urethra, e.g., squamous cell carcinoma, transitional cell carcinoma, and adenocarcinoma; cancers of the prostate, e.g., adenocarcinoma, and sarcoma; cancer of the testis, e.g., seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, and lipoma; liver cancers, including, for example, hepatoma, e.g., hepatocellular carcinoma; cholangiocarcinoma; hepatoblastoma; angiosarcoma; hepatocellular adenoma; and hemangioma; bone cancers, including, for example, osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochrondroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; nervous system cancers, including, for example, cancers of the skull, e.g., osteoma, hemangioma, granuloma, xanthoma, and osteitis deformans; cancers of the meninges, e.g., meningioma, meningiosarcoma, and gliomatosis; cancers of the brain, e.g., astrocytoma, medulloblastoma, glioma, ependymoma, germinoma (pinealoma), glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, and congenital tumors; and cancers of the spinal cord, e.g., neurofibroma, meningioma, glioma, and sarcoma; gynecological cancers, including, for example, cancers of the uterus, e.g., endometrial carcinoma; cancers of the cervix, e.g., cervical carcinoma, and pre tumor cervical dysplasia; cancers of the ovaries, e.g., ovarian carcinoma, including serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma, granulosa thecal cell tumors, Sertoli Leydig cell tumors, dysgerminoma, and malignant teratoma; cancers of the vulva, e.g., squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, and melanoma; cancers of the vagina, e.g., clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma, and embryonal rhabdomyosarcoma; and cancers of the fallopian tubes, e.g., carcinoma; hematologic cancers, including, for example, cancers of the blood, e.g., acute myeloid leukemia, chronic myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, and myelodysplastic syndrome, Hodgkin's lymphoma, non Hodgkin's lymphoma (malignant lymphoma) and Waldenström's macroglobulinemia; skin cancers, including, for example, malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Kaposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis; and adrenal gland cancers, including, for example, neuroblastoma. [00122] Cancers may be solid tumors that may or may not be metastatic. Cancers may also occur, as in leukemia, as a diffuse tissue [00123] The compounds described herein can also be administered in combination with existing methods of treating cancers, for example by chemotherapy, irradiation, or surgery. Thus, there is further provided a method of treating cancer comprising administering an effective amount of a compound according to formula I to a patient, wherein a therapeutically effective amount of one or more additional cancer chemotherapeutic agents are administered to the patient. [00124] Also provided herein is a method for treating diabetes in a patient, the method comprising administering to the patient a therapeutically effective amount of a compound of formula I. [00125] Further provided herein is a method for treating an autoimmune disease in a patient, the method comprising administering to the patient a therapeutically effective amount of a compound of formula I. The autoimmune disease can be, for example, inflammatory bowel disease (IBD). Immune responses are constantly and tightly regulated and one important cellular component in maintaining self tolerance (i.e., prevention of autoimmunity) and tolerance of benign commensal gut flora are regulatory T cells (Treg). Treg can be subdivided into multiple phenotypes, but the most common are CD4+CD25+ T cells that express the transcription factor Foxp3. Foxp3 is a direct transcriptional target of FOXO proteins, particularly FOXO1 and FOXO3. Thus activation of FOXO proteins in naïve T- cells promotes and directs differentiation to maintain a population of Treg cells. [00126] Acute immune mediated rejection and chronic immune mediated rejection are key obstacles to successful solid organ transplantation. It is believed that these forms of rejection can be prevented/overcome by amplifying Treg number and or function. Similarly, a common and morbid complication of allogeneic hematopoietic cell transplants (Allo-HCT) used to treat various malignant and non-malignant conditions, is graft versus host disease, in which the transplanted immune cells from the donor damage multiple organs in the recipient (most notably skin, gut, and liver). Increasing experimental and clinical data indicate that Tregs can be harnessed to prevent and or treat this disease process. [00127] Thus compounds of the present invention are useful in treatment of autoimmune and related diseases, by activating FOXO proteins and inducing T cell differentiation to Tregs. Compounds may be administered therapeutically to subjects directly, or alternatively, T cells may be collected from a subject and differentiated ex vivo to Tregs as described by Taylor et al. [Blood 99, 3493-3499 (2002)]. [00128] Aspects of the invention include methods for treatment of autoimmune disease characterized by deficiency in Treg function comprising administering a therapeutically useful amount of compound of formula I. The method can also include extraction of naïve T- cells from a patient, differentiation of T-cells to Tregs ex vivo by treatment with a compound of formula I, optionally supplemented with an HDACi, followed by administration of Tregs to patient with optional separation of compound of formula I from Tregs prior to their administration. As stated above, autoimmune diseases that can be so treated include IBD, solid organ transplant rejection, and GvHD in allo-HCT. [00129] In some embodiments, the compounds can be administered to a patient to treat an autoimmune disorder, for example, Addison’s disease, Amyotrophic Lateral Sclerosis, celiac disease, Crohn's disease, diabetes, eosinophilic fasciitis, Guillain-Barré syndrome (GBS), Graves’ disease, Lupus erythematosus, Miller-Fisher syndrome, psoriasis, rheumatoid arthritis, ulcerative colitis, and vasculitis. In some embodiments, the compound provided herein can be used for treating a disease or disorder in a patient wherein the disease or disorder involves excessive or unregulated cellular proliferation, the method comprising administering to the patient a therapeutically effective amount of a compound of formula I. Also provided herein is a method for treating a disease or disorder in a patient where the disease or disorder involves the dysregulation of the PI3K-AKT-FOXO signaling pathway, the method comprising administering to the patient a therapeutically effective amount of a compound of formula I. [00130] Further provided herein is a method for treating a disease in a patient wherein the disease is characterized by proteotoxicity, including age onset proteotoxicity leading to neurodegeneration, the method comprising administering to the patient a therapeutically effective amount of a compound of formula I. Hyperphosphorylated Tau has been implicated as the pathogenic protein in several neurodegenerative diseases and furthermore PP2A has been shown to be an important phosphatase in reversing aberrant phosphorylation of Tau; see for example Ludovic Martin et al., Tau protein phosphatases in Alzheimer’s disease: The leading role of PP2A in Ageing Research Reviews 12 (2013) 39- 49; Miguel Medina and Jesus Avila, Further understanding of tau phosphorylation: implications for therapy in Expert Rev. Neurotherapy, 15(1), 115-112 (2015) and Michael Voronkov et al., Phosphoprotein phosphatase 2A: a novel druggable target for Alzheimer’s disease in Future Med Chem.2011 May, 3(7) 821-833. Hyperphosphorylated alpha-Synuclein is a second exemplar of a toxic protein, and again PP2A has been shown to reverse its aberrantly phosphorylated state; see for example Kang-Woo Lee et al., Enhanced Phosphatase Activity Attenuates alpha Synucleinopathy in a Mouse Model in Neurobiology of Disease, May 11, 2011, 31(19) 6963- 6971. In some embodiments, the disease is selected from the group consisting of: Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, frontotemporal dementia, progressive supranuclear palsy, corticobasal degeneration and Pick’s disease. [00131] A second feature of Alzheimer’s disease is deposition of amyloid plaques and phosphorylation of Amyloid Precursor Protein (APP) at threonine-668 in the cytoplasmic domain of APP is involved in it’s processing to generate toxic amyloid-beta (see T. Zhang et al, Int. J. Mol. Sci. 2020, 21, 209). Activation of PP2A by treatment with compounds of the present invention decreases threonine-668 phosphorylation and suppresses pathological amyloidogenesis contributing to the development of Alzheimer’s disease. [00132] The compounds provided herein may further be used in a method for treating a mood disorder in a patient by administering to the patient a therapeutically effective amount of a compound of formula I. In some embodiments, the mood disorder is stress-induced depression. [00133] Phosphorylation and inactivation of FOXO1 by exclusion from the nucleus has been implicated in acne pathogenesis. These effects may be mediated by direct transcriptional inactivation of FOXO1 target genes or indirectly by loss of repressive interaction with other nuclear transcription factors such as the androgen receptor which contribute to acne pathogenesis, see Melnik, British Journal of Dermatology (2010) 162, pp 1398-1400, DOI: 10.1111/j.1365-2133.2010.09754.x, and Teng et al Cells (2021) 10, 1219, doi.org/10.3390/cells10051219. The compounds provided herein are there fore useful in the treatment of acne vulgaris via PP2A mediated nuclear localization of FOXO1 and deactivation of Akt signaling. The compounds may be administered by systemic administration or by topical(transdermal) delivery to affected skin. Also provided herein is a method for treating acne vulgaris in a patient by administering to the patient a therapeutically effective amount of a compound of formula I. [00134] Modulators of PP2A have also been shown to reduce markers of cellular senescence in human skin models, see Zonari et al, npj Aging (2023) 9:10. Activation of PP2A suppresses senescence associated secretory profile including expression of pro-inflamatory cytokines, for example IL-6, CXCL1 and CCL2, and reduces cellular senescence accumulation. Thus compounds of the present invention are likely to useful as agents to slow or prevent age related skin deterioration. The compounds may be administered by systemic administration or by topical (transdermal) delivery to affected skin. Also provided herein is a method for treating age related skin deterioration in a patient by administering to the patient a therapeutically effective amount of a compound of formula I. [00135] Further provided herein is a method for treating pulmonary disease such as COPD. Protein phosphatase 2A (PP2A) is a primary serine-threonine phosphatase that modulates inflammatory responses in asthma and COPD. PP2A has shown to be dysregulated in mouse models of COPD, and inhibiting PP2A activity exacerbated inflammatory responses in the lung. Conversely, increasing PP2A activity via PP2A protein transfection down regulated cytokine expression and prevented the induction of proteases following cigarette smoke extract (CSE) treatment. Thus, increasing PP2A activity by treatment with compounds of the present invention may ameliorate or reverse the pathology underlying lung diseases such as COPD. [00136] Idiopathic Pulmonary Fibrosis is a fatal lung disease in which there is progressive and irreversible scaring of the lung associated with changes to alveolar epithelial cells and aberrant fibroblast proliferation and activation. The underlying causative agent in IPF is usually unknown (hence idiopathic) and the prognosis after diagnosis is dismal with a median survival time of three years. IPF is characterized by a continuous expansion of the fibroblast population and excessive deposition of collagen in the alveolar wall leading to scarred, non- functional airspaces progressive hypoxia and death by asphyxiation. In normal lung tissue fibroblasts interact with the extracellular matrix (ECM) and signaling via integrins activates PP2A and this suppresses fibroblast growth and proliferation. In IPF fibroblasts this signaling is defective and PP2A activation is muted; in these aberrant cells, uncontrolled fibroblast proliferation and collagen secretion occurs. Diminished PP2A signaling in PP2A fibroblasts has several consequences: 1. Excessive phosphorylation of the transcription factor FOXO3a, which leads to exit of phospho-FOXO3a from the nucleus to the cytoplasm where it is sequestered by 14-3-3 proteins. PP2A is known to be the phosphatase responsible for dephosphorylating cytoplasmic FOXO3a and promoting it’s nuclear translocation. Activated, phospho-Akt is a major kinase responsible for phosphorylation of FOXO3a and PP2A is the phosphatase responsible for dephosphorylating and deactivating Akt. Thus PP2A activation promotes FOXO3a activity in two ways, by suppressing the activity of a major kinase, Akt, that inactivates it, and second by dephosphorylating cytoplasmic phospho-FOXO3a directly to cause nuclear translocation. Deficient nuclear FOXO3 protects IPF fibroblasts from polymerized collagen matrix induced apoptosis, therefore PP2A activation will suppress growth of, and will induce apoptosis of IPF fibroblasts. 2. Low PP2A activity in IPF fibroblasts results in HDAC4 hyperphosphorylation and decreases it’s nuclear localization, thus the histones of it’s target genes remain acetylated and transcriptionally active which drives excessive collagen secretion from IPF fibroblasts. Thus PP2A activation, by promoting HDAC4 nuclear translocation, will suppress excessive the excessive collagen secretion characteristic of IPF and other systemic fibrotic diseases.3. Activated phosph-ERK is a direct target of PP2A, which it dephosphorylates and deactivates. In scleroderma fibroblasts TGFb reduces PP2A activity and promotes ERK signaling and excessive collagen production. Activation of PP2A will suppress this signaling pathway from TGFb, a known and important pro-fibrotic cytokine. It is reasonable to conjecture that a similar pathway is operative in lung fibroblasts in IPF, thus PP2A activation should be useful there also. 4. PP2A negatively regulates Wnt/b-catenin signaling. Wnt3a induces lung epithelial cell proliferation, fibroblast activation and collagen synthesis in IPF. PP2A activation will suppress these processes and thus exert a therapeutic benefit in lung fibrosis and IPF. 5. Promotion of RNAPII pausing in hyperactivated lung fibroblasts in IPF by PP2A activation suppresses the expression of fibrosis related genes such as smooth muscle actin (ACTA2), collagen genes (COL1A1, COL1A2 and COL3A1) and fibronectin (FN1) (see Sattar et al, Chemical Activation of Protein Phosphatase 2A Counters TGFβ-Dependent Induction of Extracellular Matrix Proteins in Fibroblasts, Am J Respir Crit Care Med 2022; 205: A1941, poster presented PULMONARY FIBROSIS: ANIMAL AND CELL CULTURE MODELS / Thematic Poster Session / Sunday, May 15, 2022, San Francisco ATS meeting). Therefore compounds of the present invention exert a useful therapeutic effect in IPF by suppression of these genes and reducing excesive collagen deposition and scarring in IPF. In summary, PP2A is involved in several major signaling pathways implicated in the pathogenesis of lung fibrosis and IPF and in all the cases cited above PP2A activation is likely to exert a beneficial therapeutic effect. This implies that a well tolerated, effective, small molecule PP2A activator would constitute a novel therapeutic for lung fibrosis. [00137] Tumor associated fibrosis in non-small cell lung cancer (NSCLC) impairs immune surveillance and diminishes the effectiveness of immune checkpoint blockade in treatment of NSCLC, see Herzog et al, Science Translational Medicine, 15 eadh8005 (2023). Compounds of the present invention, which activate the tumor suppressor PP2A and, which display antifibrotic effects as described in paragraph [00136] above are therefore likely to be particularly useful in treatment of NSCLC. Compounds of the present invention may also be used in combination with PD-1/PD-L1 checkpoint blockade where resistance to this therapy is manifest because of tumor fibrosis. [00138] Impaired PP2A/AKT signaling has been observed in cellular models of idiopathic pulmonary hypertension, where it causes obstructive hyperproliferation and apoptosis resistance of distal pulmonary artery smooth muscle cells. Increasing PP2A activity may reverse this, thus, treatment with compounds of the present invention may be an effective treatment for pulmonary hypertension. [00139] Further provided herein is a method for treating cardiac hypertrophy in a patient by administering to the patient a therapeutically effective amount of a compound of formula I. In some embodiments, the cardiac hypertrophy is associated with a disease selected from hypertension, myocardial infarction, heart failure, and valvular heart disease. Cardiac physiology and hypertrophy are regulated by the phosphorylation status of many proteins, including receptors and ion channels, which is partly controlled by a PP2A-alpha4 intracellular signalling axis. Studies indicate that the type 2A protein phosphatases are differentially regulated in both the healthy and hypertrophied myocardium. The data suggest that pressure overload-induced hypertrophy is associated with (1) altered expression of type 2A protein phosphatases and their regulatory subunits and (2) an increase in expression of their non-catalytic inhibitor protein alpha4. Thus, treatment with compounds of the present invention may ameliorate cardiac hypertrophy. Also, significant reduction in endosomal PP2A activity has been observed in heart failure samples versus controls, suggesting that inhibited resensitization of beta-adrenergic receptors occurs in human heart failure. These studies suggest that resensitization of beta adrenergic receptors is inhibited in human heart failure and targeting the PP2A inhibitor SET to derepress and activate PP2A may provide preservation of receptor function and beneficial cardiac remodeling. Thus, treatment with compounds of the present invention may have a beneficial effect in heart failure. [00140] Further provided herein is a method for treating a parasitic infection in a patient by administering to the patient a therapeutically effective amount of a compound of formula I. Examples of parasites that may cause parasitic infections to be treated include, but are not limited to, Plasmodium and Theileria. [00141] Further provided herein is a method for treating inflammatory conditions. Reduced PP2A activity occurs in animal models of allergic airway disease and patients with severe asthma. Treatment with small molecule activators of PP2A such as fingolimod (FTY720) or 2-amino-4-(4-(heptyloxy) phenyl)-2-methylbutan-1-ol (AAL(S)) inhibited the development of inflammation, airway hyperreactivity in mouse models of allergic airway disease. Thus, compounds of the present invention may be useful in the treatment of asthma. Dephosphorylation of tristetraprolin (TTP) functions as an “off-switch” in inflammatory responses, and it’s activity can be promoted by compounds that stimulate PP2A activity. Therapeutic efficacy of protein phosphatase 2A (PP2A)-activating drugs, to target tristetraprolin (TTP), in models of rheumatoid arthritis has been demonstrated in vitro and in vivo. Thus, treatment with compounds of the present invention may be useful in chronic inflammatory conditions such as rheumatoid arthritis. [00142] PP2A enzymes are involved in the regulation of cell transcription, cell cycle, and viral transformation. Many viruses, including cytomegalovirus, parainfluenza, DNA tumor viruses, and HIV-1, utilize different approaches to exploit PPA2 in order to modify, control, or inactivate cellular activities of the host [Garcia et al., Microbes and Infection, 2, 2000, 401−407]. Therefore, the compounds provided herein may further be used in a method for treating a viral infection in a patient by administering to the patient a therapeutically effective amount of a compound of formula I. Examples of viruses that may cause viral infections to be treated include, but are not limited to: a polyomavirus, such as John Cunningham Virus (JCV), Simian virus 40 (SV40), or BK Virus (BKV); influenza, Human Immunodeficiency Virus type 1 (HIV-1), Human Papilloma Virus (HPV), adenovirus, Epstein-Barr Virus (EBV), Hepatitis C Virus (HCV), Molluscum contagiosum virus (MCV); Human T-lymphotropic virus type 1 HTLV-1), Herpes Simplex Virus type 1 (HSV-1), cytomegalovirus (CMV), hepatitis B virus, Bovine papillomavirus (BPV-1), human T-cell lymphotropic virus type 1, Japanese encephalitis virus, respiratory syncytial virus (RSV), and West Nile virus. [00143] Serine/Threonine phosphatases, including PP2A, are involved in modulation of synaptic plasticity (D. G. Winder and J. D. Sweatt, Nature Reviews Neuroscience, vol 2, July 2001, pages 461–474). Persistently decreased PP2A activity is associated with maintenance of Long Term Potentiation (LTP) of synapses, thus treatment PP2A activators such as those described here may reverse synaptic LTP. Psychostimulant drugs of abuse such as cocaine and methamphetamine are associated with deleterious synaptic LTP (L. Mao et al, Neuron 67, September 9, 2010 and A. Stipanovich et al, Nature vol 453, 2008, pages 879–884), which may underlie the pathology of addiction and relapse therefore PP2A activators described here may be useful as treatments for psychostimulant abuse. [00144] Abnormalities in synaptic structure and signaling are linked to autistic spectrum disorder, see for example, Y Chen et al., CTTNBP2, but not CTTNBP2NL, regulates dendritic spinogenesis and synaptic distribution of the striatin-PP2A complex, Molecular Biology of the Cell, 23, November 15, 2012, 4383-4392. PP2A has been shown to be important in normal development of dendritic spines, and treatment with compounds of the present invention may ameliorate or reverse autistic spectrum disorder. [00145] Further provided herein is a method for treating a disease or disorder in which the disease or disorder involves the dysregulation of the mTOR-PP2A signaling axis. Mammalian target of rapamycin (mTOR) is a serine/threonineprotein kinase that regulates cell growth, proliferation, and survival: mTOR is frequently activated in human cancers and is a commonly sought anticancer therapeutic target. PP2A is a key element in mTOR-AKT signaling during nutritional deprivation, and it has important implications in cell cycle progression and quiescence. Dysregulation of cellular metabolism is a feature of cancer, with nutrient transport defects, nutrient sensing defects, dysregulated autophagy and constitutive anabolism being common in tumors; aberrant activation of mTOR is implicated in all of these processes and PP2A activation has been demonstrated to modulate them in vivo. PP2A has been shown to be involved in regulatory feedback loops with mTOR, and PP2A activators of the present invention would be expected to affect these processes directly by interacting with mTOR complexes, or indirectly by counterbalancing mTOR’s effects by dephosphorylating its targets. Perturbation of the mTOR signaling cascade appears to be a common pathophysiological feature of human neurological disorders, including mental retardation syndromes and autism spectrum disorders, and neurodegenerative conditions such as Alzhiemer’s disease. Activation of PP2A has been shown to be effective in animal models of neurodegenerative disease by modulating the PP2A mTOR axis; thus, molecules of the present invention will be useful in treatment of these conditions. PP2A activators of the present invention are likely to be useful in the treatment of diseases in which mTOR signaling is dysregulated; these include cancer, diabetes and neurodegenerative conditions. Compounds of the present invention may also promote innate immunity to infection and promote healthy aging. [00146] In some embodiments of the compounds, compositions, or methods provided herein, the compound is selected from a compound of Table 1, or a pharmaceutically acceptable salt thereof. Abbreviations and Definitions [00147] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art to which this disclosure belongs. A comprehensive list of abbreviations utilized by organic chemists (i.e. persons of ordinary skill in the art) appears in the first issue of each volume of the Journal of Organic Chemistry. The list, which is typically presented in a table entitled “Standard List of Abbreviations” is incorporated herein by reference. In the event that there is a plurality of definitions for terms cited herein, those in this section prevail unless otherwise stated. [00148] The following abbreviations and terms have the indicated meanings throughout: Ac = acetyl Aq = aqueous Boc = t-butyloxy carbonyl Bu = butyl c- = cyclo cat = catalyst Cbz = carboxybenzyl DBA = dibenzylideneacetone DCM = dichloromethane = methylene chloride = CH2Cl2 DMF = N,N-dimethylformamide eq. or equiv. = equivalent(s) Et = ethyl GC = gas chromatography h = hour(s) KHMDS = Potassium bis(trimethylsilyl)amide Lg = leaving group Ln = chiral ligands mCPBA = meta-Chloroperoxybenzoic acid Me = methyl mesyl = methanesulfonyl min. = minute(s) Ms = mesylate NMO or NMMO = N-methylmorpholine oxide Pg = protecting group Ph = phenyl RT = room temperature sat’d or sat. = saturated t- or tert = tertiary Tf = triflate TFA = trifluoroacetic acid THF = tetrahydrofuran tosyl = p-toluenesulfonyl [00149] Throughout this specification the terms and substituents retain their definitions. [00150] The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. As used herein, the singular forms “a”, “an” and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise. It will be further understood that the terms “comprise” (and any form of comprise, such as “comprises” and “comprising”), “have” (and any form of have, such as “has” and “having”), “include” (and any form of include, such as “includes” and “including”), and “contain” (and any form contain, such as “contains” and “containing”) are open-ended linking verbs. As a result, a method or composition that “comprises”, “has”, “includes” or “contains” one or more steps or elements possesses those one or more steps or elements, but is not limited to possessing only those one or more steps or elements. Likewise, a step of a method or an element of a composition that “comprises”, “has”, “includes” or “contains” one or more features possesses those one or more features, but is not limited to possessing only those one or more features. The terms “comprising” and “including” or grammatical variants thereof are to be taken as specifying the stated features, integers, steps or components but do not preclude the addition of one or more additional features, integers, steps, components or groups thereof. For example, "X includes a, b and c" means that X includes, but is not limited to, a, b and c. This term encompasses the terms “consisting of” and “consisting essentially of”. [00151] The phrase “consisting essentially of” or grammatical variants thereof when used herein are to be taken as specifying the stated features, integers, steps or components but do not preclude the addition of one or more additional features, integers, steps, components or groups thereof, but only if the additional features, integers, steps, components or groups thereof do not materially alter the basic and novel characteristics of the claimed composition or method. [00152] Unless otherwise specified, the phrase "such as" is intended to be open-ended. For example, "X can be a halogen, such as fluorine or chlorine" means that X can be, but is not limited to, fluorine or chlorine. [00153] As used herein, and as would be understood by the person of skill in the art, the recitation of “a compound”—unless expressly further limited—is intended to include salts of that compound. In a particular embodiment, the term “compound of formula” refers to the compound or a pharmaceutically acceptable salt thereof. [00154] The term "pharmaceutically acceptable salt" refers to salts prepared from pharmaceutically acceptable non-toxic acids or bases including inorganic acids and bases and organic acids and bases. When the compounds of the present invention are basic, salts may be prepared from pharmaceutically acceptable non-toxic acids including inorganic and organic acids. Suitable pharmaceutically acceptable acid addition salts for the compounds of the present invention include acetic, adipic, alginic, ascorbic, aspartic, benzenesulfonic (besylate), benzoic, boric, butyric, camphoric, camphorsulfonic, carbonic, citric, ethanedisulfonic, ethanesulfonic, ethylenediaminetetraacetic, formic, fumaric, glucoheptonic, gluconic, glutamic, hydrobromic, hydrochloric, hydroiodic, hydroxynaphthoic, isethionic, lactic, lactobionic, laurylsulfonic, maleic, malic, mandelic, methanesulfonic, mucic, naphthylenesulfonic, nitric, oleic, pamoic, pantothenic, phosphoric, pivalic, polygalacturonic, salicylic, stearic, succinic, sulfuric, tannic, tartaric acid, teoclatic, p-toluenesulfonic, and the like. When the compounds contain an acidic side chain, suitable pharmaceutically acceptable base addition salts for the compounds of the present invention include, but are not limited to, metallic salts made from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc or organic salts made from lysine, arginine, N,N'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine) and procaine. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium cations and carboxylate, sulfonate and phosphonate anions attached to alkyl having from 1 to 20 carbon atoms. [00155] The terms "subject" or "subject in need thereof" or “patient” are used interchangeably herein. These terms refer to a patient who has been diagnosed with the underlying disorder to be treated. The subject may currently be experiencing symptoms associated with the disorder or may have experienced symptoms in the past. Additionally, a "subject in need thereof" may be a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological systems of a disease, even though a diagnosis of this disease may not have been made. As a non-limiting example, a "subject in need thereof", for purposes of this application, may include a male who is currently diagnosed with prostate cancer or was diagnosed with prostate cancer in the past, regardless of current symptomatology. [00156] A “patient,” as used herein, includes both humans and other animals, particularly mammals. Thus the methods are applicable to both human therapy and veterinary applications. In some embodiments, the patient is a mammal, for example, a primate. In some embodiments, the patient is a human. [00157] As used herein, the terms “treatment” or “treating are used interchangeably. These terms refer to an approach for obtaining beneficial or desired results including, but not limited to, therapeutic benefit. Therapeutic benefit includes eradication or amelioration of the underlying disorder being treated; it also includes the eradication or amelioration of one or more of the symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient may still be afflicted with the underlying disorder. [00158] Treatment can involve administering a compound described herein to a patient diagnosed with a disease, and may involve administering the compound to a patient who does not have active symptoms. Conversely, treatment may involve administering the compositions to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made. [00159] The terms “administer”, “administering” or “administration” in reference to a dosage form of the invention refers to the act of introducing the dosage form into the system of subject in need of treatment. When a dosage form of the invention is given in combination with one or more other active agents (in their respective dosage forms), “administration” and its variants are each understood to include concurrent and/or sequential introduction of the dosage form and the other active agents. Administration of any of the described dosage forms includes parallel administration, co-administration or sequential administration. In some situations, the therapies are administered at approximately the same time, e.g., within about a few seconds to a few hours of one another. [00160] A “therapeutically effective” amount of the compounds described herein is typically one which is sufficient to achieve the desired effect and may vary according to the nature and severity of the disease condition, and the potency of the compound. It will be appreciated that different concentrations may be employed for prophylaxis than for treatment of an active disease. A therapeutic benefit is achieved with the amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient may still be afflicted with the underlying disorder. [00161] The term modulate with respect to PP2A refers to activation or potentiation of phosphatase activity by three general effects 1. direct allosteric activation of catalytic activity in PP2A complexes. 2. By promotion of assembly of heterotrimeric B subunit containing trimers, or recruitment of PP2A AC heterodimers to the Integrator-RNAPII complex. 3. By displacement of endogeneous PP2A inhibitors or chaparones, thereby derepressing PP2A activity. These effects are not mutually exclusive though their relative importance may depend on cell or tissue type and specific disease state or pathology. [00162] The term “modulate” with respect to a FOXO transcription factor protein refers to activation of the FOXO transcription factor protein and its biological activities associated with the FOXO pathway. Modulation of FOXO transcription factor proteins includes up- regulation (i.e., agonizing, activation or stimulation). The mode of action of a FOXO modulator can be direct, e.g., through binding to the FOXO transcription factor protein as a ligand. The modulation can also be indirect, e.g., through binding to and/or modifying another molecule which otherwise binds to and activates the FOXO transcription factor protein. [00163] “Hydrocarbon” (e.g., (C 1 -C 8 )hydrocarbon) includes alkyl, cycloalkyl, polycycloalkyl, alkenyl, alkynyl, aryl and combinations thereof. Examples include benzyl, phenethyl, cyclohexylmethyl, adamantyl, norbornyl, and naphthylethyl. Hydrocarbyl (or hydrocarbon) refers to any substituent comprised of hydrogen and carbon as the only elemental constituents. Aliphatic hydrocarbons are hydrocarbons that are not aromatic; they may be saturated or unsaturated, cyclic, linear or branched. Examples of aliphatic hydrocarbons include isopropyl, 2-butenyl, 2-butynyl, cyclopentyl, norbornyl, etc. Aromatic hydrocarbons include benzene (phenyl), naphthalene (naphthyl), anthracene, etc. [00164] Unless otherwise specified, alkyl (or alkylene) is intended to include linear or branched saturated hydrocarbon structures and combinations thereof. Alkyl refers to alkyl groups from 1 to 20 carbon atoms, preferably 1 to 10 carbon atoms, more preferably 1 to 6 carbon atoms. Examples of alkyl groups include methyl, ethyl, propyl, isopropyl, n-butyl, s- butyl, t-butyl and the like. [00165] Cycloalkyl is a subset of hydrocarbon and includes cyclic hydrocarbon groups of from 3 to 8 carbon atoms. Examples of cycloalkyl groups include cy-propyl, cy-butyl, cy- pentyl, norbornyl and the like. [00166] Alkoxy or alkoxyl refers to groups of from 1 to 20 carbon atoms, preferably 1 to 10 carbon atoms, more preferably 1 to 6 carbon atoms of a straight or branched configuration attached to the parent structure through an oxygen. Examples include methoxy, ethoxy, propoxy, isopropoxy and the like. Lower-alkoxy refers to groups containing one to four carbons. For the purpose of this application, alkoxy and lower alkoxy include methylenedioxy and ethylenedioxy. [00167] The term "halogen" means fluorine, chlorine, bromine or iodine atoms. In one embodiment, halogen may be a fluorine or chlorine atom. [00168] The terms "haloalkyl," "haloalkoxy," or “haloalkylthio” mean alkyl, alkoxy, or alkylthio, respectively, substituted with one or more halogen atoms. [00169] Heterocycle means an aliphatic or aromatic carbocycle residue in which from one to four carbons is replaced by a heteroatom selected from the group consisting of N, O, and S. The nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized. Unless otherwise specified, a heterocycle may be non- aromatic (heteroaliphatic) or aromatic (heteroaryl). Examples of heterocycles include pyrrolidine, pyrazole, pyrrole, indole, quinoline, isoquinoline, tetrahydroisoquinoline, benzofuran, benzodioxan, benzodioxole (commonly referred to as methylenedioxyphenyl, when occurring as a substituent), tetrazole, morpholine, thiazole, pyridine, pyridazine, pyrimidine, thiophene, furan, oxazole, oxazoline, isoxazole, dioxane, tetrahydrofuran and the like. Examples of heterocyclyl residues include piperazinyl, piperidinyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyrazinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolyl, quinuclidinyl, isothiazolidinyl, benzimidazolyl, thiadiazolyl, benzopyranyl, benzothiazolyl, tetrahydrofuryl, tetrahydropyranyl, thienyl (also historically called thiophenyl), benzothienyl, thiamorpholinyl, oxadiazolyl, triazolyl and tetrahydroquinolinyl. Examples of heteroaryls include imidazole, pyridine, indole, thiophene, benzopyranone, thiazole, furan, benzimidazole, quinoline, isoquinoline, quinoxaline, pyrimidine, pyrazine, tetrazole and pyrazole. In some embodiments, examples of heteroaryls include imidazole, pyridine, thiophene, thiazole, furan, pyrimidine, pyrazine, tetrazole and pyrazole. [00170] As used herein, the term “optionally substituted” may be used interchangeably with “unsubstituted or substituted”. The term “substituted” refers to the replacement of one or more hydrogen atoms in a specified group with a specified radical. “Oxo” may also be included among the substituents referred to in “optionally substituted”; it will be appreciated by persons of skill in the art that, because oxo is a divalent radical, there are circumstances in which it will not be appropriate as a substituent (e.g. on phenyl). In one embodiment, 1, 2, or 3 hydrogen atoms are replaced with a specified radical. In the case of alkyl and cycloalkyl, more than three hydrogen atoms can be replaced by fluorine; indeed, all available hydrogen atoms could be replaced by fluorine. Table 1. Examples Example Number Synthesis Scheme Structure r r 5 2 1 8 ur r [00171] Preparation of compounds can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of appropriate protecting groups, can be readily determined by one skilled in the art. Suitable groups for that purpose are discussed in standard textbooks in the field of chemistry, such as Protective Groups in Organic Synthesis by T.W.Greene and P.G.M.Wuts [John Wiley & Sons, New York, 1999], in Protecting Group Chemistry, 1 st Ed., Oxford University Press, 2000; and in March’s Advanced Organic chemistry: Reactions, Mechanisms, and Structure, 5 th Ed., Wiley-Interscience Publication, 2001. [00172] Sulfonimidamides are isosteres of sulfonamides and their application in medicinal chemistry has been reviewed in Chinthakindi et al, Angew. Chem. Int. Ed. 2017, 56, 4100- 4109. Sulfonimidamides may improve physical and medicinal properties, versus the corresponding sulfonamides, with respect to solubility, lipophilicity, plasma protein binding or permeability. They may also have altered, and potentially beneficial, off-target profile versus the sulfonamides with respect to, for example CYP inhibition or PXR induction resulting in reduced potential for drug-drug inteactions or toxicity. Synthesis of sulfonimidamides of the present invention is achieved by reacting an amine intermediate A1, shown in General Synthesis Scheme 1, using a variety of methods disclosed in the scientific literature. See for example Nandi and Arvidsson, Sulfonimidamides: Synthesis and Applications in Preparative Organic Chemistry, Adv. Synth. Catal., 2018, 360, 2976-3001. One method is shown in General Synthesis Scheme 1 using an N-(alkyl)benzenesulfinamide sufinamide reagent which is activated with t-butyl hypochlorite in carbontetrachloride and reacted with amine intermediate A1. In some instances the alkyl group maybe lower alkyl, such as methyl and is thus R im . In other instances the alkyl group maybe removable and it functions as a protecting group to yield R im = H after deprotection. One example is N-(2,4- dimethoxybenzyl)-4-(trifluoromethoxy)benzenesulfinamide where the protecting group is 2,4-dimethoxybenzyl which is conveniently removed by acid treatment, as described in the examples below. [00173] General Synthesis Scheme 1 is shown in Fig.1. [00174] General synthesis of Amine Intermediates A1. The key reaction step is construction of the central tetrahydropyran ring by a modification of the Prins-Ritter conditions reported by Subba Reddy and Ghanty in Synthetic Communications, 2014, 44:17, pages 2545-2554. The main modifications are 1. Use of methyl enol ether as aldehyde equivalent, 2. Microwave heating and 3. Stoichiometric benzenesulfonimide acid catalyst. The reaction is reported to give cis-relative stereochemistry as the major product, as depicted for Example 2. [00175] Synthesis Scheme 2. Example 1: Synthetic route to N-(2-(10,11-dihydro-5H- dibenzo[a,d][7]annulen-5-yl)tetrahydro-2H-pyran-4-yl)-4- (trifluoromethoxy)benzenesulfonimidamide is shown in Scheme 2 in Fig.2. [00176] An oven dried two-neck round bottomed flask equipped with a pressure equalizing addition funnel was cooled to room temperature while flushing with argon and (methoxymethyl)triphenylphosphonium chloride, 34.3 g (100 mmole, 2 equivalents) was added, and the solid suspended in 200 mL dry THF. The suspension was stirred and cooled to 0 o C in ice, then 40 mL of 2.5M n-butyllithium solution (100 mmole, 2 equivalents) in hexanes was added dropwise. The deep red solution was stirred at 0 o C for 10 minutes, then a solution of dibenzosuberone, 10.4 g (50 mmole, 1 equivalent) in 50 mL dry THF was added from the addition funnel and the solution was stirred at 0 o C for 5 hours, then at room temperature overnight. White precipitate forms after about five hours. The reaction was cooled in ice then quenched with aqueous ammonium chloride (2.6g, 50 mmole in 10 mL water), stirred briefly, then filtered through a pad of Celite to remove bulk of precipitate, which was washed with 50 mL ethyl acetate. The filtrate was evaporated to give an orange oil. 100 mL ethyl acetate was added, then 50 mL hexanes (till solution was slightly cloudy) and the mixture was left to stand in a fridge at 4 o C overnight. The mixture was filtered through a 2 cm pad of silica gel, which was washed with ethyl acetate. The filtrate was evaporated to give a pale yellow oil which was purified by flash chromatography eluting with 2% ethyl acetate in hexane, to give 4.87g (41%) of the product, 5-(methoxymethylene)- 10,11-dihydro-5H-dibenzo[a,d][7]annulene, as a colorless oil. 300 MHz 1 H NMR in CDCl 3 7.46–7.11 (overlapping m, 8H), 6.37 (s, 1H), 3.73 (s, 3H), 3.16 (br s, 4H). TLC-MS ESI +ve ion, 278.9 [M+CH3CN+H] + . [ 00177] 5-(Methoxymethylene)-10,11-dihydro-5H-dibenzo[a,d][7]annulen e, 1.26g (5 mmole, 1 equivalent), 3-buten-1-ol, 0.53 mL (0.46 g, 6.4 mmole, 1.2 equivalent), and 1,3,2- Benzodithiazole-1,1,3,3-tetraoxide, 1.1 g (5 mmole, 1 equivalent) were placed in a 30 mL CEM microwave vial. Dry acetonitrile, 15 mL, was added and the mixture was stirred briefly at room temperature to give a clear homogeneous solution. The mixture was stirred and heated at 150°C for 1 hour. Reaction was diluted into 100 mL ethyl acetate and the organic was washed once with 1M aqueous potassium carbonate, once with saturated brine then dried over anhydrous sodium sulfate. Filtration and evaporation gives crude product which was purified by flash chromatography, eluting with 60 to 70% ethyl acetate hexane. The product, N-(2-(10,11-dihydro-5H-dibenzo[a,d][7]annulen-5-yl)tetrahydr o-2H-pyran-4-yl)acetamide, is a colorless oil that crystallizes to give a white solid on pumping in vacuo: 0.73 g (2.3 mmole, 45%). 300 MHz 1 H NMR in CDCl37.21–7.05 (overlapping m, 8H), 5.32 (br d, 1H), 4.11 (m, 1H), 3.98-3.90 (overlapping m, 2H), 3.83 (m, 1H), 3.50-3.33 (overlapping m, 3H), 2.98-2.81 (overlapping m, 2H) 1.89 (s, 3H), 1.85 (m, 1H), 1.63 (m, 1H), 1.35 (m, 1H), 1.02 (m, 1H). TLC-MS ESI +ve ion, 336.2 [M+H] + . [00178] N-(2-(10,11-dihydro-5H-dibenzo[a,d][7]annulen 5 yl)tetrahydro 2H pyran 4 yl)acetamide, 0.86g (2.6 mmole, 1 equivalent) was placed in a 30 mL CEM microwave vial and dissolved in 15 mL dioxane. 5 mL of 6M hydrochloric acid was added and the mixture stirred briefly at room temperature, then heated in microwave at 150°C for 1 hour. The reaction was cooled and made basic by adding 2 g solid potassium hydroxide, then stirred at room temperature for 30 min as two phases form. The reaction was diluted into 100 mL ethyl actate, washed with water once, then saturated brine and dried over anhydrous sodium sulfate. Filtration and evaporation gives the crude Amine Intermediate, 2-(10,11-dihydro-5H- dibenzo[a,d][7]annulen-5-yl)tetrahydro-2H-pyran-4-amine, which was carried into the next step without further purification. The Prins-Ritter reaction give predominantly the cis diastereoisomer. [00179] N-(2,4-dimethoxybenzyl)-4-(trifluoromethoxy)benzenesulfinami de is a known compound and is prepared as follows: a mixture of 4-trifluoromethoxybenzenesulfonyl chloride (100 g, 383.7 mmole) and sodium sulfite (106.4 g, 844 mmole) and sodium bicarbonate (70.8 g, 844 mmole) in 1L of water was stirred at 65°C for 18 hours under argon atmosphere. The mixture was concentrated to remove water keeping the temperature below 60°C under vacuume. Then the residue was stirred with 1 L methanol at 15–25°C and the reaction mixture was filtered and concentrated to yield sodium 4- (trifluoromethoxy)benzenesulfinate. To a mixture of sodium 4- (trifluoromethoxy)benzenesulfinate (20 g, 80.6 mmole) and DMF (0.2 mL, catalytic) in 200 mL methylene chloride was added oxaloyl chloride (15.4 g, 121.3 mmole) with stirring and cooling to keep the temperature below 20°C. The mixture was stirred for an additional one hour at room temperature then evaporated to give crude 4-(trifluoromethoxy)benzenesulfinic chloride which was then redissolved in 200 mL methylene chloride. 2,4- dimethoxybenzylamine (20.2 g, 121.3 mmole) and triethylamine (24.6 g, 248.1 mmole) was added and the mixture stirred at 15-25°C for 18 hours. The reaction was quenched with 100 mL water and the organic layer separated. The aqueous was back extracted with 100 mL methylene chloride. The combined organic was washed with brine and dried over sodium sulfate. Filteration and evaporation gives the crude product, N-(2,4-dimethoxybenzyl)-4- (trifluoromethoxy)benzenesulfinamide which is purified by flash chromatography, eluting with 10-20% ethyl acetate hexane. [00180] Coupling to give the protected intermediate, N-((2R,4S)-2-(10,11-dihydro-5H- dibenzo[a,d][7]annulen-5-yl)tetrahydro-2H-pyran-4-yl)-N'-(2, 4-dimethoxybenzyl)-4- (trifluoromethoxy)benzenesulfonimidamide is carried out as follows: N (2,4 dimethoxybenzyl)-4-(trifluoromethoxy)benzenesulfinamide (1 equiv) is stirred with t-butyl hypochlorite (1.05 equiv) in carbon tetrachloride at 0°C for 1 hour in the dark. The reaction is concentrated to remove carbon tetrachloride keeping the temperature below 5°C, then the residue is dissolved in THF. The amine intermediate, 2-(10,11-dihydro-5H- dibenzo[a,d][7]annulen-5-yl)tetrahydro-2H-pyran-4-amine (1.05 equiv) and diisopropylethylamine (3 equiv) are added with stirring and cooling at 0°C, then the mixture is stirred at room temperature over night. The reaction is cooled and quenched with water and extracted with ethyl acetate. The combined organic is washed with brine, then dried over sodium sulfate. Filtration and evaporation gives the crude 2,4-dimethoxybenzyl protected compound which is purified by flash chromatography, eluting with ethyl acetate hexane. [00181] Deprotection to the final product, Example 1, is carried out by dissolving the 2,4- dimethoxybenzyl protected compound in methylene chloride, cooling to 0°C, then adding adding trifluoroacetic acid to give a 1:1 solvent:acid ratio by volume. Saturated aqueous sodium bicarbonate is added till the mixture is pH 7-8, then the mixture is extracted with dichloromethane. The combined organic extracts are dried over sodium sulfate. Filtration and evaporation gives the crude product, N-(2-(10,11-dihydro-5H-dibenzo[a,d][7]annulen-5- yl)tetrahydro-2H-pyran-4-yl)-4-(trifluoromethoxy)benzenesulf onimidamide, Example 1, as a mixture of diastereoisomers with respect to the sulfur chiral center. The amine intermediate from the Prins-Ritter synthesis is predominantly the cis diastereoisomer with respect to the central tetrahydropyran ring and this is depicted as Example 2 in Table 1. This material is purified by flash chromatography eluting with ethyl actate-hexane. The mixture of diastereoisomers with respect to the sulfur center, may be sparable on a flash column. Example 1 may also be separable into it’s diastereoisomers by techniques such as HPLC or crystallization. The separate diastereoisomers are depicted as Examples 3 and 4 in Table 1. [00182] Synthesis of Example 3 and Example 4 was carried out as shown in the scheme shown in Fig.3. [00183] N-(2-(10,11-dihydro-5H-dibenzo[a,d][7]annulen-5-yl)tetrahydr o-2H-pyran-4-yl)-4- (trifluoromethoxy)benzenesulfonamide is a known compound and was synthesized as described in WO 2023/023594 as a single cis-diastereoisomer as shown above and labelled sulfonamide intermediate. The intermediate sulfonamide, 1.03 g (2 mmole, 1 equiv) and Hunigs base, 0.68 mL (0.52 g, 4 mmole, 2 equiv) were dissolved in 10 mL dry chloroform in a 35 mL microwave vial. Ph3PCl2, 1.0 g (3 mmole, 1.5 equiv) was added as a solid in one portion and the mixture was stirred and microwaved at 85°C for twenty minutes and then cooled to room temperature. 2,4-dimethoxybenzylamine, 1.34 g (8 mmole, 4 equiv) was added and the mixture stirred at room temerature overnight. the reaction was diluted into 100 mL ethyl acetate then washed with 50 mL 0.1 M hydrochloric acid then 50 mL sat. aq. sodium bicarbonate. The organic was dried over anhydrous sodium sulfate, filetered and evaporated to give a brown oil. Careful flash chromatography eluting with 15% to 25% ethyl acetate separates the 2,4-dimethoxybenzyl protected imidamides as distereoisomers at the sulfur center as glassy foam after pumping in vacuo. Less polar Isomer 1: 400 MHz 1 H NMR in CDCl 3 7.80 (m, 2H), 7.22 (m, 1H) 7.21- 7.07 (overlapping m, 10H), 6.80 (br d, 1H [NH proton]), 6.29- 6.27 (overlapping m 2H), 4.07 (m, 1H), 4.01- 3.81 (overlapping m, 4H), 3.76 (s, 3H), 3.65 (s, 3H), 3.50 (m, 2H) 3.39 (m, 1H), 3.30 (m, 1H), 2.93 (m, 2H), 1.78 (m, 1H), 1.64 (overlapping m, 2H), 1.35 (m, 1H). TLC-MS APCI +ve ion, 667.2 [M+H] + and APCI - ve ion, 665.4 [M-H]- . More polar Isomer 2: 400 MHz 1 H NMR in CDCl37.76 (m, 2H), 7.20 (m, 1H) 7.15- 7.02 (overlapping m, 10H), 6.88 (br d, 1H [NH proton]), 6.29- 6.27 (overlapping m 2H), 4.04-3.98 (overlapping m, 2H), 3.96- 3.91 (overlapping m, 2H), 3.80 (m, 1H), 3.74 (s, 3H), 3.68 (s, 3H), 3.50 (m, 2H) 3.32 (m, 2H), 2.89 (m, 2H), 1.77 (m, 1H), 1.72- 1.59 (overlapping m, 2H), 1.29 (m, 1H). TLC-MS APCI +ve ion, 667.2 [M+H] + and APCI - ve ion, 665.4 [M-H]-. [00184] A 1:1 mixture of the 2,4-dimethoxybenzyl protected imidamides, 0.69 g (approximately 1 mmole) was dissolved in 10 mL methylene chloride. The solution was cooled in ice and 10 mL trifluoroacetic was added, then the mixture was stirred at room temerature overnight. The reaction was diluted into 100 mL ethyl acetate and washed with an ice-cold solution of 9 g potassium carbonate in 100 mL water. The organic was dried over anhydrous sodium sulfate, then filtered and evaporated to give crude Example 2 as a 1:1 mixture of diastereoisomers at the sulfur center. The mixture was further purified by flash chromatography eluting with 30%–40% ethyl acetate in hexane. The diastereoisomers separate to give Example 2 and Example 3 as pale yellow solids after pumping in vacuo. Configuration at the sulfur center is undetermined and the less polar, first eluting isomer is assigned as Example 3: 400 MHz 1 H NMR in CDCl37.93 (m, 2H), 7.23 (m, 2H) 7.15-6.94 (overlapping m, 8H), 3.96 (m, 1H), 3.85 (m, 1H), 3.80-3.68 (overlapping m, 3H [includes broad NH, 2H]), 3.49-3.18 (overlapping m, 4H), 2.93-2.85 (overlapping m, 2H) 1.51-1.43 (overlappimg m, 2H), 1.31 (m, 1H), 1.00 (m, 1H). TLC-MS APCI +ve ion, 517.2 [M+H] + and APCI -ve ion, 515.2 [M-H]-. The more polar, second eluting isomer is assigned as Example 4: 400 MHz 1 H NMR in CDCl 3 7.88 (m, 2H), 7.20 (m, 2H) 7.18- 7.01 (overlapping m, 8H), 3.91 (m, 2H), 3.69 (m, 1H), 3.34 (m, 2H), 3.23 (m, 2H), 3.00 (broad NH, 2H ), 2.91 (m 1H), 2.82 (m, 1H), 1.71 (m, 1H), 1.40 (m, 1H), 1.27 (m, 1H), 0.90 (m, 1H). TLC-MS APCI +ve ion, 517.2 [M+H] + and APCI -ve ion, 515.2 [M-H]-. [00185] Example 9 and Example 10 maybe prepared using the method described above from N-(2-(2,8-difluoro-10,11-dihydro-5H-dibenzo[a,d][7]annulen-5 -yl) tetrahydro-2H-pyran-4- yl)-4-(trifluoromethoxy)benzenesulfonamide, as starting material where preparation is given in WO 2023/023594. [00186] Example 7 is prepared from 2-(bis(4-fluorophenyl)methyl)tetrahydro-2H-pyran-4- amine, synthesized as shown in Fig. 4, by coupling with N-(2,4-dimethoxybenzyl)-4- (trifluoromethoxy)benzenesulfinamide using the method described above for Example 1. [00187] The Prins-Ritter reaction used in the synthesis of Examples 1 and 7 gives mainly 2,4-cis relative stereochemistry across the central tetrahydropyran ring, see Subba Reddy and Ghanty in Synthetic Communications, 2014, 44:17, pages 2545-2554 and Yadav et al, Tetrahedron Letters 48 (2007) pages 4903-4906. Compounds with 2,4-trans stereochemistry may be accessed by inversion of the 4-amino stereocenter on the tetrahydropyran using one of the several methods developed by Fiksdahl, see for example Sørbye et al, Tetrahedron: Asymmetry 9 (1998) pages 681-689, or Said and Fiksdahl, Tetrahedron: Asymmetry 12 (2001) 1947-1951. The 2,4-trans amine intermediates may be used in the synthesis of compounds such as Example 5 shown in Table 1. [00188] Compounds wherein R im = lower alkyl, for example methyl, as depicted for Example 6 above are prepared from N-methyl-4-(trifluoromethoxy)benzenesulfinamide and coupling to amine intermediates as described above. [00189] Compounds wherein Y=H maybe prepared via the route shown in General Scheme 2, show in Fig.5. [00190] Conditions in Scheme 2 are adapted from those published in Chen and Gibson, RSC Adv. 2015, 5, 4171–4174 or Chinthakindi et al, Eur. J. Org. Chem., 2018, 10.1002/ejoc.201801541 and use of these conditions in the syntheses of Example 7 and Example 8 are described. [00191] Synthesis of Example 7 (Fig. 6). 2-(bis(4 fluorophenyl)methyl)tetrahydro 2H pyran-4-amine (Amine Intermediate A2 in the Fig. 4) was dissolved in 10 mL methylenechloride and 0.41 mL (2.4 mmole, 1.2 equiv) of Hunigs base was added followed by 0.41 mL (0.63g, 2.4 mmol, 1.2 equiv) of 4-trifluoromethoxybenzenesulfonyl chloride. The mixture was stirred at room temperature overnight then diluted into 100mL ethyl acetate and the organic was washed with 1M HCl(aq), 1x50 mL, followed by water, 1x50 mL then dried over anhydrous magnesium sulfate. Filtration and evaporation give crude Sulfonamide Intermediate, which was purified by flash chromatography eluting with 20% ethylacetate in hexane. [00192] N-(2-(bis(4-fluorophenyl)methyl)tetrahydro-2H-pyran-4-yl)-4- (trifluoromethoxy)benzenesulfonamide, is a white solid: 400 MHz 1 H NMR in CDCl3 7.84 (m, 2H), 7.27 (m, 2H), 7.17 (m, 2H), 7.07 (m, 2H), 6.93 (m, 4H), 4.62 (d, 1H), 3.96 (m, 1H), 3.84 (m, 1H),, 3.78 (m, 1H), 3.42 (m, 1H), 3.36 (m, 1H), 1.72 (m, 1H), 1.58 (m, 1H), 1.37 (m, 1H), 1.04 (m, 1H). TLC-MS APCI -ve ion 526.5 [M-H]-. [00193] To a stirred suspension of Ph 3 PCl 2 (1 equiv) in dry chloroform is added triethylamine (1.6 equiv). The mixture is stirred at room temperature for 20 minutes then cooled to 0°C. The Sulfonamide Intermediate, N-(2-(bis(4-fluorophenyl)methyl)tetrahydro- 2H-pyran-4-yl)-4-(trifluoromethoxy)benzenesulfonamide, is added dropwise with stirring as solution in chloroform. The mixture is stirred at between 0°C and 50°C for 10 to 60 minutes. The reaction is cooled to 0°C then 2.4-dimethoxybenzylamine is added as a solution in chloroform and the mixture is stirred at between 0°C and room temperature for between 1 and 24 hours. The reaction is concentrated and then ether is added and the mixture stirred, then filtered. The filtrate is evaporated to give the crude product which is purified by flash chromatography. Deprotection is carried out by dissolving in dichloromethane, cooling to 0°C, and adding trifluoroacetic acid to give 1:1 CH2Cl2:TFA. The reaction is concentrated, then taken up in ethyl acetate and washed with aqueous sodium bicarbonate, then dried over sodium sulfate. Filtration and evaporation gives crude material with is purified by flash chromatography to give Example 7. [00194] Alternate conditions used for synthesis of Example 7 were as follows: N-(2-(bis(4- fluorophenyl)methyl)tetrahydro-2H-pyran-4-yl)-4-(trifluorome thoxy)benzenesulfonamide, 0.527 g (1 mmole) and Hunigs base, 0.26 mL (0.19 g, 1.5 mmole, 1.5 equiv) were dissolved in 10 mL dry chloroform in a 35 mL microwave vial. Ph 3 PCl 2 , 0.4g (1.2mmole, 1.2 equiv) was added as a solid in one portion then the vial was sealed and the mixture was stirred and microwaved at 85°C for twelve minutes; mixture darkens on adding Ph 3 PCl 2 . The mixture was cooled to room temperature, then 2,4-dimethoxybenzylamine, 0.5 g (3 mmole, 3 equiv) was added and the mixture was stirred overnight at room temperature. The reaction was diluted with 100 mL dichloromethane and washed with 50 mL 1% citric acid then 50 mL sat. aq. sodium bicarbonate. The organic solution was dried over sodium sulfate and evaporated to give a brown oil which was purified by flash chromatography eluting with 20% ethyl acetate/hexane.300 MHz 1 H NMR in CDCl 3 is consistent with 1:1 mixture of diastereoismers at sulfur chiral center, for example each methoxy singlet of 2,4-methoxybenzyl occurs is doubled to give four singlets at 3.76 and 3.74 ppm and 3.68 and 3.67 ppm. TLC-MS APCI +ve ion, 677.2 [M+H] + and APCI -ve ion, 675.3 [M-H]- is consistent with 2,4- dimethoxybenzyl protected intermediate: [00195] The above compoun was eprotecte y ssoving 0.64 g in 10 mL methylene chloride with cooling to 0°C. 10 mL trifluoroacetic was added and the mixture stirred at 0°C for 5 minutes, then room temperature for 7 hours, a deep purple color developed. The reaction was diluted into 100 mL ethyl acetate and washed with a solution of 8.3 g potassium carbonate in 100 mL water. The organic solution was dried over anhydrous sodium sulfate, filtered then evaporated to give a crude residue which was purified by flash chromatography eluting with 30% ethyl acetate in hexane. Example 7 separates on chromatography into two diastereoisomers at the sulfur center. Less polar isomer, 7-isomer1: 400 MHz 1 H NMR in CDCl 3 7.97 (m, 2H), 7.42 (m, 2H) 7.23 (m, 2H), 7.09 (m, 2H), 6.97- 6.89 (overlapping m 4H), 3.92 (m, 1H), 3.90 (m, 1H), 3.84 (overlapping m, 2H), 3.42 (m, 1H), 3.34 (m, 1H) 3.06 (br singlet, 2H [NH protons]), 1.64 (m, 1H), 1.56 (m, 1H), 1.31 (m, 1H), 1.06 (m, 1H). MS ESI +ve ion, 527.0 [M+H] + , MS ESI -ve ion, 525.0 [M-H]-. More polar isomer, 7-isomer2: 400 MHz 1 H NMR in CDCl 3 7.93 (m, 2H), 7.22 (m, 2H), 7.15 (m, 2H), 7.04 (m, 2H), 6.96- 6.80 (overlapping m 4H), 3.96 (m, 1H), 3.80 (m, 1H), 3.75 (m, 1H), 3.40 (overlapping m, 2H), 3.02 (br singlet, 2H [NH protons]), 1.75 (m, 1H), 1.43-1.34 (overlapping m, 2H), 0.95 (m, 1H). MS ESI +ve ion, 527.0 [M+H] + , MS ESI -ve ion, 525.1 [M-H]-. [00196] Example 8 wherein R im is methyl maybe prepared by the route shown in Fig.7. [00197] N-Methyl-4-(trifluoromethoxy)benzenesulfonamide (1 equiv) in chloroform is added dropwise to a stirred suspension of Ph3PCl2 (1 equiv) in dry chloroform is added triethylamine (1.6 equiv). The mixture is stirred at between 0 and 50°C for between 30 minutes and 8 hours. The mixture is cooled to 0°C then a solution of 2-(bis(4- fluorophenyl)methyl)tetrahydro-2H-pyran-4-amine in chloroform is added. The mixture is stirred at between 0 and 50°C for between 30 minutes and 24 hours. The reaction is diluted into ethyl acetate and washed with aqueous sodium bicarbonate. The organic is dried over sodium sulfate, filtered and concentrated to give crude product. The crude material is purified by flash chromatography to give Example 8. [00198] Cancer Cell Viability Inhibition [00199] D425 medulloblastoma cells were purchased from SigmaAldrich. D425 meduloblastoma cells were cultured in improved Dulbecco's modified Eagle's medium (DMEM) Richter's modification supplemented with 20% fetal bovine serum and 1% Penicillin-streptomycin. Incubation was performed at 37°C and 5% CO 2. [00200] D425 cells were seeded at 3000–3500 cells/90 µL growth medium per well on 96- well tissue culture plates. Cells were incubated overnight to allow them to recover. Next day cells were treated with tested compounds and incubated for 48 hours. [00201] Cell Proliferation assays in triplicate were performed at each concentration. Compounds test range was 1- 30 µM (0, 5, 10, 15, 20, 25, and 30). The compounds were dissolved in DMSO. A series of dilutions were made in 1% DMSO in growth medium so that the final concentration of DMSO is 0.1% in all of treatments. [00202] After treatment, cells were allowed to equilibrate at room temperature for one hour. Cell proliferation was measured by Luminescence quantification using Promega CellTiter- Glo Luminescent Cell Viability Assay. To perform the assay, 90 µL of Celltiter-Glo substrate was added to each well, plates were shake for 2 minutes and allowed to equilibrate for 10 minutes at room temperature. Luminescence intensity was measured using the Spectramax i3x plate reader. [00203] The Luminescence intensity data were analyzed using the computer software Graphpad Prism. In the absence of the compound, the luminescence intensity (Lt) in each data set was defined as 100% cell viability. The percent cell in the presence of each compound was calculated according to the following equation: %Cell= L/L t , were L= the luminescence intensity in the presence of the compound. [00204] The values of % cell versus a series of compound concentrations (0, 5, 10, 15, 20, 25, and 30 µM) were then plotted using Nonlinear regression analysis of Sigmoidal dose- response curve. IC50 value was determined by the concentration causing a half-maximal percent activity. [00205] IC50 for example compounds, determined by the method above are given in Table 2. Table 2. Example IC50 for D425 cell growth (μM) Note 7-isomer1 14.3 n = 3 3 3 3 [00206] A172 gl fied Eagle's medium (DMEM) supplemented with 10% fetal bovine serum and 1% Penicillin-streptomycin. Incubation was performed at 37°C and 5% CO2. [00207] A172 cells were seeded at 3000–3500 cells/90 µL growth medium per well on 96- well tissue culture plates. Cells were incubated overnight to allow them to recover and reattach. Next day cells were treated with tested compounds and incubated for 48-72 hours. [00208] Cell Proliferation assays in triplicate were performed at each concentration. Compounds test range was 1- 30 µM (0, 5, 10, 15, 20, 25, and 30). The compounds were dissolved in DMSO. A series of dilutions were made in 1% DMSO in growth medium so that the final concentration of DMSO is 0.1% in all of treatments. [00209] After treatment, cells were allowed to equilibrate at room temperature for 1/2 to one hour. Cell proliferation was measured by Luminescence quantification using Promega CellTiter-Glo Luminescent Cell Viability Assay. To perform the assay, 100 µL of Celltiter- Glo substrate was added to each well, plates were shake for 2 minutes and allowed to equilibrate for 10 minutes at room temperature. Luminescence intensity was measured using the Spectramax i3x plate reader. [00210] The Luminescence intensity data were analyzed using the computer software Graphpad Prism. In the absence of the compound, the luminescence intensity (L t ) in each data set was defined as 100% cell viability. The percent cell in the presence of each compound was calculated according to the following equation: %Cell= L/L t , were L= the luminescence intensity in the presence of the compound. [00211] The values of % cell versus a series of compound concentrations (0, 5, 10, 15, 20, 25, and 30 µM) were then plotted using Nonlinear regression analysis of Sigmoidal dose- response curve. IC50 value was determined by the concentration causing a half-maximal percent activity. [00212] Cellular in vitro phosphatase activation assay was performed using the method of Theendakara et al, Molecular and Cellular Neuroscience, 83 (2017), pages 83-91 which is described below: [00213] A172 glioblastoma cells were cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum and 1% Penicillin-streptomycin. Incubation was performed at 37°C and 5% CO 2. [00214] A172 cells were seeded at 5x10 5 cells/2.5 ml growth medium (without antibiotics) per well on 6-well tissue culture plates. Cells were incubated overnight to allow them to recover and reattach. Next day, when the cells were at about 75% confluence, transfection was performed with 2-2.5 ug of ApoE cDNA following the manufacture's protocol: Lipofectamine 2000 from Invitrogen (Cat # 12566014) or Neuroporter transfection kit form Sigma (Cat# NPT01). After 24 hrs. the cells were treated with 5 μM test compounds and incubated for an additional 24 hours. [00215] After 24hrs, cells were washed twice with imidazole buffer and lysed using NP-40 Lysis buffer. The lysate was cleared up from small molecules including endogenous phosphates using desalting spin columns. Malachite Green Phosphatase assays were performed in triplicate using 5ug of total protein per assay and following the manufacture's protocol (Sigma cat # MAK307). The amount of phosphate release in the assay was determined by measuring absorbance at 620 nm using the Spectramax i3x plate reader. [00216] The absorbance intensity data were analyzed using the computer software Graphpad Prism. The phosphate concentration for each sample was determined from the standard curve using phosphate standard concentrations. In order to compare samples, the first set (column A) were there was no transfection performed and no compound added, was designated a baseline; the absorbance measurement in this column was set as 100% phosphatase activity. [00217] Phosphatase activation, determined by the method above are given in Table 3. Table 3. Example % increase from ApoE transfected cell at Note 5 μM test compound concentration 3 [00218] Whi f illustration, the foregoing descriptions and examples should not be deemed to be a limitation on the scope of the invention. Accordingly, various modifications, adaptations, and alternatives may occur to one skilled in the art without departing from the spirit and scope of the present invention.