Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMBINATION THERAPY FOR PI3K-ASSOCIATED DISEASE OR DISORDER
Document Type and Number:
WIPO Patent Application WO/2019/232403
Kind Code:
A1
Abstract:
Described herein are compositions and methods for treating a disease or disorder associated with PI3K signaling. For example, such compositions can include use of modulators of glucose metabolism, use of at least one kinase in the insulin-receptor/PI3K/AKT/mTOR pathway, and/or use of diet that influences the subject's metabolic state.

Inventors:
CANTLEY LEWIS C (US)
HOPKINS BENJAMIN (US)
MUKHERJEE SIDDHARTHA (US)
GONCALVES MARCUS (US)
Application Number:
PCT/US2019/034949
Publication Date:
December 05, 2019
Filing Date:
May 31, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV CORNELL (US)
CANTLEY LEWIS C (US)
HOPKINS BENJAMIN (US)
MUKHERJEE SIDDHARTHA (US)
GONCALVES MARCUS (US)
International Classes:
A23L33/20; A61K31/155; A61K38/00; A61P3/10; A61P35/00; C07C279/00
Domestic Patent References:
WO2018027084A22018-02-08
Foreign References:
CN105147696A2015-12-16
US20170100354A12017-04-13
US20150290300A12015-10-15
Other References:
BOWMAN, C ET AL.: "Ketoacidosis with Canagliflozin Prescribed for Phosphoinositide 3-Kinase Inhibitor-Induced Hyperglycemia: A Case Report", JOURNAL OF INVESTIGATIVE MEDICINE HIGH IMPACT CASE REPORTS, vol. 5, no. 3, 23 August 2017 (2017-08-23), pages 1 - 4, XP055658220
OLEKSYSZYN, J: "The complete control of glucose level utilizing the composition of ketogenic diet with the gluconeogenesis inhibitor, the anti-diabetic drug metformin, as a potential anti-cancer therapy", MEDICAL HYPOTHESES, vol. 77, no. 2, 6 May 2011 (2011-05-06), pages 171 - 173, XP028245385, DOI: 10.1016/j.mehy.2011.04.001
ALLEN, BG ET AL.: "Ketogenic diets as an adjuvant cancer therapy: History and potential mechanism", REDOX BIOLOGY, vol. 2, 7 August 2014 (2014-08-07), pages 963 - 970, XP055264210, DOI: 10.1016/j.redox.2014.08.002
HOPKINS, BD ET AL.: "Obesity and Cancer Mechanisms: Cancer Metabolism", JOURNAL OF CLINICAL ONCOLOGY, vol. 34, no. 35, 7 November 2016 (2016-11-07), pages 4277 - 4283, XP055658221
See also references of EP 3801069A4
Attorney, Agent or Firm:
PERDOK, Monique M. et al. (US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A method of inhibiting cell proliferation, comprising contacting a cell with an effective amount of a modulator of glucose metabolism; and contacting the cell with an effective amount of at least one inhibitor of at least one kinase in the insulin-receptor/PBK/AKT/mTOR pathway to thereby inhibit cell proliferation.

2. The method of claim 1 , wherein the modulator of glucose metabolism is a glucose-uptake inhibitor selected from a sodium-glucose-linked transport protein 1 (SGLT1) inhibitor, a sodium- glucose-linked transport protein 2 (SGLT2) inhibitor, a dual SGLT1/SGLT2 inhibitor, or a combination thereof.

3. The method of claim 1 , wherein the modulator of glucose metabolism is dapagliflozin, empagliflozin, canagliflozin, ipragliflozin, metformin, tofogliflozin, sergliflozin etabonate, remogliflozin etabonate, ertugliflozin, sotagliflozin, conagliflozin, or a combination thereof.

4. The method of claim 1 , wherein the modulator of glucose metabolism is a sodium-glucose-linked transport protein 2 (SGLT2) inhibitor.

5. The method of claim 1 , wherein the modulator of glucose metabolism is metformin.

6. The method of claim 1 , wherein the inhibitor of at least one kinase in the insulin-receptor/PBK/AKT/mTOR pathway is an inhibitor of a PI3K, protein kinase B (AKT), mechanistic target of rapamycin (mTOR), or a combination thereof.

7. The method of claim 1 , wherein the inhibitor of at least one kinase in the insulin-receptor/PBK/AKT/mTOR pathway is idelalisib, copanlisib, buparlisib (BKM120), alpelisib (BYL719), taselisib (GDC-0032), pictilisib (GDC-0941), apitolisib (GDC-0980), serabelisib (TAK-117), dactolisib, MK2206, linsitinib (OSI-906), or a combination thereof.

8. The method of claim 1 , wherein the inhibitor of at least one kinase in the insulin-receptor/PBK/AKT/mTOR pathway is buparlisib (BKM120), alpelisib (BYL719), taselisib (GDC-0032), pictilisib (GDC-0941), linsitinib (OSI-906), or a combination thereof.

9. The method of claim 1 , wherein the modulator of glucose metabolism is metformin or a sodium-glucose-linked transport protein 2 (SGLT2) inhibitor; and wherein the inhibitor of at least one kinase in the insulin- receptor/PBK/AKT/mTOR pathway is buparlisib (BKM120), alpelisib (BYL719), taselisib (GDC-0032), pictilisib (GDC-0941), linsitinib (OSI-906), or a combination thereof.

10. A pharmaceutical composition comprising a modulator of glucose metabolism and a pathway inhibitor that inhibits at least one kinase in the insulin- receptor/PBK/AKT/mTOR pathway.

11. A method of treating a cell proliferative disease, comprising:

a. administering to a subject in need thereof an effective amount of at least one inhibitor of at least one kinase in the insulin- receptor/PBK/AKT/mTOR pathway; and

b. administering to the subject a ketogenic diet and/or at least one

modulator of glucose metabolism,

to thereby inhibit cell proliferation in the subject.

12. The methods of claim 11, wherein the inhibitor of at least one kinase in the insulin-receptor/PBK/AKT/mTOR pathway is an inhibitor of a PI3K, an inhibitor of protein kinase B (AKT), an inhibitor of mechanistic target of rapamycin (mTOR), or a combination thereof.

13. The method of claim 11 , wherein the inhibitor of at least one kinase in the insulin-receptor/PBK/AKT/mTOR pathway is idelalisib, copanlisib, buparlisib (BKM120), alpelisib (BYL719), taselisib (GDC-0032), pictilisib (GDC-0941), apitolisib (GDC-0980), serabelisib (TAK-117), dactolisib, MK2206, linsitinib (OSI-906), or a combination thereof.

14. The method of claim 11 , wherein the inhibitor of at least one kinase in the insulin-receptor/PBK/AKT/mTOR pathway is buparlisib (BKM120), alpelisib (BYL719), taselisib (GDC-0032), pictilisib (GDC-0941), linsitinib (OSI-906), or a combination thereof.

15. The method of claim 11 , wherein the subject is administered at least one modulator of glucose metabolism.

16. The method of claim 15, wherein the modulator of glucose metabolism is a sodium-glucose-linked transport protein 1 (SGLT1) inhibitor, a sodium- glucose-linked transport protein 2 (SGLT2) inhibitor, a dual SGLT1/SGLT2 inhibitor, or a combination thereof.

17. The method of claim 15, wherein the modulator of glucose metabolism is dapagliflozin, empagliflozin, canagliflozin, ipragliflozin, tofogliflozin, sergliflozin etabonate, remogliflozin etabonate, ertugliflozin, sotagliflozin, conagliflozin, or a combination thereof.

18. The method of claim 15, wherein the modulator of glucose metabolism is a sodium-glucose-linked transport protein 2 (SGLT2) inhibitor.

19. The method of claim 15, wherein the modulator of glucose metabolism is metformin.

20. The method of claim 11 , wherein the subject is administered the ketogenic diet.

21. The method of claim 11 , wherein the subject is on a ketogenic diet before, during, or before and during the administering of the at least one inhibitor of a kinase in the insulin-receptor/PBK/AKT/mTOR pathway.

22. The method of claim 11 , wherein the cell proliferative disease is a leukemia, polycythemia vera, lymphoma, Waldenstrom’s macroglobulinemia, heavy chain disease, a sarcoma, a carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinoma,

cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms tumor, cervical cancer, uterine cancer, testicular cancer, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma,

craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodenroglioma, schwannoma, meningioma, melanoma, neuroblastoma, retinoblastoma, or a combination thereof.

23. The method of claim 11 , comprising administering to the subject metformin and an inhibitor of at least one kinase in the insulin- receptor/PBK/AKT/mTOR pathway to thereby treat cancer in the subject, while the subject is on a ketogenic diet before, during, or before and during the administering.

24. A kit comprising at least one inhibitor of a mammalian insulin- receptor/PBK/AKT/mTOR pathway and one or more modulators of glucose metabolism.

25. A kit comprising at least one inhibitor of a mammalian insulin- receptor/PBK/AKT/mTOR pathway, and one or more components of a ketogenic diet.

Description:
COMBINATION THERAPY FOR PI3K-ASSOCIATED

DISEASE OR DISORDER

This application claims benefit of priority to the filing date of U.S. Provisional Application Ser. No. 62/679,329, filed June 1, 2018, the contents of which are specifically incorporated by reference herein in their entirety.

FIELD OF THE DISCLOSURE

The present disclosure relates generally to methods of treating diseases or disorders associated with insulin-receptor/PI3K/AKT/mTOR pathway signaling, including hematologic malignancies and solid tumors. In particular, the disclosure relates to increasing the efficacy of an insulin-receptor/PI3K/AKT/mTOR pathway inhibitor by modulation of glucose metabolism either pharmacologically or with diet.

BACKGROUND

An association exists between some diseases and disorders and the insulin- receptor phosphatidylinositol kinase (phosphoinositide 3-kinase (PI3K)), protein kinase B (AKT) and mammalian target of rapamycin (mTOR) signaling pathway (called the insulin-receptor/PI3K/AKT/mTOR pathway). In cancer, mutations in PIK3CA are observed at similar frequency to mutations in KRAS (Kandoth, C. et al. Mutational landscape and significance across 12 major cancer types. Nature 502, 333- 339, doi:l0. l038/naturel2634 (2013); Millis, S. Z., Ikeda, S., Reddy, S., Gatalica, Z. & Kurzrock, R. Landscape of Phosphatidylinositol-3-Kinase Pathway Alterations Across 19784 Diverse Solid Tumors. JAMA Oncol l, 1565-1573,

doklO. lOOl/jamaoncol.2016.0891 (2016).)

Even though therapies targeting the insulin-receptor/PI3K/AKT/mTOR pathway are desirable, the medical community has struggled to identify effective compositions and methods for targetting PI3K as well as upstream and downstream regulators of insulin-receptor/PI3K/AKT/mTOR signalling. Thus, there exists a long- felt and unmet need for compositions and method for treating a disease or disorder associated with PI3K signaling. The present disclosure provides such compositions and methods, and more.

SUMMARY OF THE DISCLOSURE

The present disclosure relates generally to compositions and methods for treating a disease or disorder associated with PI3K signaling that can include administration of a modulator of glucose metabolism with or without a diet that influences the subject’s metabolic state. In some cases, the method can include administering an inhibitor of at least one kinase in the insulin- receptor/PI3K/AKT/mTOR pathway and administering a modulator of glucose metabolism. In some cases, the method includes administering the pathway inhibitor and/or a modulator of glucose metabolism to a subject who consumes a ketogenic diet during treatment. The disclosure further relates to pharmaceutical compositions that can include a pathway inhibitor, modulator of glucose metabolism, or a combination thereof.

The disclosure provides a method of treating a disease or disorder associated with PI3K signaling, that includes administering to a subject in need thereof an effective amount of a modulator of glucose metabolism; and administering to the subject an effective amount of a pathway inhibitor of the insulin- receptor/PI3K/AKT/mTOR pathway.

The disclosure also provides a method of treating a disease or disorder associated with PI3K signaling, that includes administering an effective amount of a pathway inhibitor, wherein optionally the pathway inhibitor is capable of inhibiting at least one kinase in the insulin-receptor/PBK/AKT/mTOR pathway.

The subject can consume or be administered a ketogenic diet during any of these treatments.

The disclosure also provides a pharmaceutical composition that includes a modulator of glucose metabolism and a pathway inhibitor, wherein optionally the pathway inhibitor is capable of inhibiting at least one kinase in the insulin- receptor/PBK/AKT/mTOR pathway.

The disclosure also provides a method of inhibiting cell proliferation or cell- proliferative disease, that includes administering to a subject an effective amount of a glucose-uptake inhibitor; and administering to the subject an effective amount of a PI3K inhibitor.

The subject can be in need of treatment or the treatment can be performed to inhibit the onset of a disease or condition.

The disclosure also provides a method of inhibiting cell proliferation or a cell- proliferative disease, that involves administering to a subject an effective amount of a PI3K inhibitor, wherein the subject consumes a ketogenic diet during treatment. In some embodiments, the modulator of glucose metabolism is a glucose- uptake inhibitor. For example, such a glucose-uptake inhibitor can be a sodium- glucose-linked transport protein 1 (SGLT1) inhibitor, a sodium-glucose-linked transport protein 2 (SGLT2) inhibitor, a dual SGLT1/SGLT2 inhibitor, or a combination thereof.

In some embodiments, the glucose-uptake inhibitor is selected from dapagliflozin, empagliflozin, canagliflozin, ipragliflozin, tofogliflozin, sergliflozin etabonate, remogliflozin etabonate, ertugliflozin, sotagliflozin, conagliflozin, or a combination thereof.

In some embodiments, the modulator of glucose metabolism is metformin.

In some embodiments, the modulator of glucose metabolism is an insulin receptor/ insulin like growth factor 1 (IGF1) receptor inhibitor, wherein optionally the insulin receptor/IGF 1 receptor inhibitor is linsitinib (OSI-906).

In some embodiments, the pathway inhibitor is selected from idelalisib, copanlisib, buparlisib (BKM120), alpelisib (BYL719), taselisib (GDC-0032), pictilisib (GDC-0941), apitolisib (GDC-0980), serabelisib (TAK-117), dactolisib, apelisib, MK2206, linsitinib (OSI-906), or a combination thereof.

In some embodiments, the PI3K inhibitor is selected from the group consisting of idelalisib, copanlisib, buparlisib (BKM120), alpelisib (BYL719), taselisib (GDC- 0032), pictilisib (GDC-0941), apitolisib (GDC-0980), serabelisib (TAK-117), dactolisib, and apelisib.

In some embodiments, the disease or disorder associated with PI3K signaling is a cancer or cell-proliferative disorder, a metabolic disorder, a neurodegenerative disease, an inflammatory disease, or a combination thereof.

In some embodiments, disruption of systemic glucose homeostasis improves efficacy of pathway-inhibitor treatment compared to pathway inhibitor alone.

In some embodiments, the inhibition of cell proliferation or cell-proliferative disease is enhanced compared to administration of the PI3K inhibitor without a glucose-uptake inhibitor.

BRIEF DESCRIPTION OF THE FIGURES

FIG. 1A-1D demonstrate that treatment with PI3K inhibitors causes systemic feedback resulting in increases in blood glucose and insulin. FIG. 1A graphically illustrates blood glucose levels measured over time in mice treated with the indicated PI3K inhibitor compounds (N=5/arm, p- value <0.0001 by Two-Way ANOVA for all blood glucose curves of mice in treatment cohorts as compared to vehicle). To mimic clinical treatment these mice were not starved for this assay. FIG. 1B graphically illustrates insulin levels, measured over time in mice treated with the indicated PI3K inhibitor compounds (N=5/arm, p-value <0.0001 by Two-Way ANOVA for all blood glucose curves of mice in treatment cohorts as compared to vehicle). To mimic clinical treatment these mice were not starved for this assay. FIG. 1C graphically illustrates c- Peptide levels accessed from serum samples taken from the same animals as used for FIG. 1A-1B at the end of the time course, 240 min, as a representative of area under the curve insulin release p-values comparing vehicle treatment to buparlisib (BKM120), Alpelisib (BYL719), and taselisib (GDC-0032), by t-test were 0.017, <0.0001 , and 0.007 respectively. FIG. 1D-1 graphically illustrates the quantity of fluorodeoxyglucose tracer detected via fluorodeoxyglucose-Positron Emission Tomography (FDG-PET) scans (shown in FIG. 1D-2) of orthotopically implanted Kras-Tp53-Pdx-Cre (KPC) tumors imaged 90 minutes after a single treatment with buparlisib (BKM120) (N=4/arm). These result demonstrate that PI3K inhibitor- induced spikes in insulin caused an increase in glucose uptake in the tumors of these animals in the acute setting after a single dose of BKM120 (p-value =0.0002, by t- test).

FIG. 2A-2C demonstrate the impact of feedback levels of insulin on cellular proliferation, signaling and survival. FIG. 2A shows a Western blot of proteins from orthotopic Kras-Tp53-Pdx-Cre (KPC) cell lines K8484 and K8082 treated with or without the PI3K inhibitor BKM120 (luM) and/or insulin (lOng/ml), further illustrating the physiologic responses to insulin as observed in FIG. 1 A-1D. FIG. 2B graphically illustrates results of a proliferation assay of the KPC cell line K8484 grown in the presence or absence of insulin (lOng/ml) and BKM120 (luM). p-values determined by ANOVA comparing conditions +/- insulin are shown. FIG. 2C graphically illustrates results of a proliferation assay of KPC cell lines K8082 grown in the presence or absence of insulin (lOng/ml) and BKM120 (luM). p-values determined by ANOVA comparing conditions +/- insulin are shown.

FIG. 3A-3G demonstrate that targeting the PI3K inhibitor induces glucose/insulin feedback in vivo. FIG. 3A graphically illustrates blood glucose levels over time of wildtype c57/bl6 mice baring syngeneic K8484 KPC allografted tumors after treatment with a single dose of BKM120 with metformin pretreatment, SGLT2- inhibitor (SGLT2i) pretreatment, or a ketogenic diet (N = 4/arm). p- values calculated by Two-way repeated measures ANOVA for metformin, SGLT2i, and the ketogenic diet were 0.2136 (not significant), <0.0001 , and 0.007 respectively. FIG. 3B graphically illustrates blood levels of c-peptide of the same mice as for FIG. 3A taken 180 minutes after treatment with BKM120. p-values calculated by unpaired t-test for metformin, SGLT2i, and ketogenic diet were 0.7566 (not significant), 0.0386, and 0.0117 respectively. FIG. 3C shows immunohistochemical images illustrating pS6 (ser-235) expression to observe the level of active PI3K signaling in these tumors. FIG. 3D graphically illustrates quantification of the staining observed in FIG. 3C shown as positive cells per high power field (20 fields/arm taken as 5 high power images averaged for each of the 4 mice) p-values comparing pS6 positive cells in BKM120 alone treated tumors as compared to those treated with BKM120 in combination with metformin, SGLT2i, or the ketogenic diet were 0.6186, <0.0001, and <0.0001 respectively. FIG. 3E-1 to FIG. 3E-4 show IVIS images of luciferase reporter luminescence in mice with KPC K8484 tumors after 12 days of treatment with PI3Ka specific inhibitor BYL-719 alone or in combination with other agents. FIG. 3E-1 shows IVIS images of luciferase reporter luminescence in mice with KPC K8484 tumors after 12 days of treatment with PI3Ka specific inhibitor BYL-719 alone. FIG. 3E-2 shows IVIS images of luciferase reporter luminescence in mice with KPC K8484 tumors after 12 days of treatment with PI3Ka specific inhibitor BYL-719 combined with metformin for ten days prior to BYL-719 treatment (N=l0 tumors/arm). FIG. 3E-3 shows IVIS images of luciferase reporter luminescence in mice with KPC K8484 tumors after 12 days of treatment with PI3Ka specific inhibitor BYL-719 combined with a ketogenic diet for ten days prior to BYL-719 treatment (N=l0 tumors/arm). FIG. 3E-4 shows IVIS images of luciferase reporter luminescence in mice with KPC K8484 tumors after 12 days of treatment with PI3Ka specific inhibitor BYL-719 combined with canagliflozin (SGLT2i) for ten days prior to BYL-719 treatment (N=l0 tumors/arm). Dietary interventions in this study were initiated at time of tumor implantation, BYL-719 treatment was initiated nine days after implantation. FIG. 3F graphically illustrates quantification of luminescence from the images of these tumors. FIG. 3G graphically illustrates survival of these animals showing that the addition of either SGLT2 inhibitor or administration of ketogenic diet to treatment with BYL-719 increases overall survival of these ani als as determined by Log-rank (Mantel-Cox) test, p-value = 0.0019 and <0.0001 respectively.

FIG. 4A-4E demonstrate the impact of circumventing the on-target glucose/insulin feedback of PI3K inhibitors upon tumor growth. FIG. 4A graphically illustrates K8484 KPC tumor volume (where the tumor cells express doxycycline inducible hairpins targeting Renilla (ShRenilla) or Insulin Receptor (ShIR)) in mice treated with doxycycline and with the PI3K inhibitor BYL-719 and/or with a ketogenic diet. Tumors in this experiment were allowed to grow for fourteen days reaching an average size greater than 1 cm 3 at which point diet, doxycycline induction of hairpin expression, and PI3K inhibitor treatments were initiated (N > 5 tumors/arm as indicated). FIG. 4B graphically illustrates ES272 Pik3ca mutant breast cancer allograft tumor volume treated with BYL-719 and/or insulin along with the ketogenic (keto) as indicated. Tumors in this experiment were allowed to grow for 10 days after implantation into mice prior to the initiation of diet and indicated treatments. FIG. 4C graphically illustrates tumor volume of subject-derived endometrial xenografts (PDX) treated with BKM120 and/or while having a ketogenic diet (N=5/arm). ANOVA comparison between the BKM120 alone and BKM120 plus ketogenic diet treated mice indicates that the addition of ketogenic diet significantly enhances treatment efficacy in this model (p-value = 0.0028). FIG. 4D illustrates the histology of phospho-Insulin Receptor (pINSR), phospho-AKT (pAKT), phospho-S6 (pS6), cleaved caspase 3 (Cl. Casp 3), and Ki67 of tumors taken 4 hours after the last treatment with Vehicle, ketogenic diet, BKM120, or the combination of the ketogenic diet with BKM120 (BKMl20/Keto). FIG. 4E graphically illustrates the quantification of phospho-Insulin Receptor (pINSR), phospho-AKT (pAKT), phospho-S6 (pS6), cleaved caspase 3 (Cl. Casp 3), and ki67 of the tumors shown in FIG. 4D taken 4 hours after the last treatment with Vehicle, ketogenic diet, BKM120, or the combination of the ketogenic diet with BKM120 (BKMl20/Keto). Quantification is depicted as score per high powered field, four images were taken for each of the five mice p-values from t-tests comparing the blinded scoring in BKM120 treated tumors as compared to those treated with BKM120 with the ketogenic diet were 0.005, 0.005, 0.017 and 0.028 and for pINSR, pAKT, pS6 and Cl. Casp 3 respectively. FIG. 5A-5D illustrate blood Glucose and C-Peptide levels after treatment with agents that target PI3K pathway. FIG. 5 A graphically illustrates blood glucose levels over time where time 0 is the time of treatment with the indicated inhibitor. FIG. 5B graphically illustrates blood glucose levels over time where time 0 is the time of treatment with additional inhibitors as indicated. FIG. 5C graphically illustrates c- peptide levels from mice in FIG. 5 A taken 240 and 180 minutes after treatment with inhibitors, as a surrogate for total insulin release in these animals, showing that the PI3K inhibitors and IGFR/INSR inhibitors dramatically increase insulin release in these animals. In all cases compounds that caused acute increases in blood glucose levels also increased serum insulin levels. FIG. 5D graphically illustrates c-peptide levels from the mice described in FIG. 5B taken 240 and 180 minutes after treatment with inhibitors, as a surrogate for total insulin release in these animals, showing that the PI3K inhibitors and IGFR/INSR inhibitors dramatically increase insulin release in these animals. In all cases compounds that caused acute increases in blood glucose levels also increased serum insulin levels.

FIG. 6A-6G demonstrate the impact of the feedback levels of insulin observed in FIG. 1A-D upon BKM120 efficacy in vitro. FIG. 6A-1 graphically illustrates cellular proliferation in minimal growth media of MDA-MB-468 breast cancer cells whose growth is partially rescued by the addition of the observed feedback levels of insulin (lOng/ml) induced by BKM120 in mice. FIG. 6A-2 graphically illustrates proliferation in minimal growth media of BT-549 breast cancer cells whose growth is partially rescued by the addition of the observed feedback levels of insulin (lOng/ml) induced by BKM120 in mice. FIG. 6A-3 graphically illustrates proliferation in minimal growth media of PC-3 prostate adenocarcinoma cells whose growth is partially rescued by the addition of the observed feedback levels of insulin (lOng/ml) induced by BKM120 in mice. FIG. 6B graphically illustrates cell viability assay demonstrating the effects that these feedback levels of insulin have upon 2 subject (Pt)-derived organoid cultures (Pt A and Pt B) being treated in a dose response with BKM120 as measured by cell titer-glo at 96 hours. FIG. 6C graphically illustrates proliferation (percent confluence) in minimal growth media partially rescued by the addition of the observed feedback levels of insulin induced by BKM120 in mice as observed in FIG. 1A-D. FIG. 6D shows proliferation (percent confluence) of FICT1 l6-neo cells with and without treatment with the physiologically observed levels of insulin (lOng/ml) and treatment with clinically relevant PI3K inhibitors GDC-0032 and BYL-719. FIG. 6E graphically illustrates proliferation (percent confluence) of HCT116 PTEN knockout (KO) cells with and without treatment with the physiologically observed levels of insulin (lOng/ml) and treatment with clinically relevant PI3K inhibitors GDC-0032 and BYL-719. FIG. 6F shows proliferation (percent confluence) of DLDl-Neo under the indicated treatment conditions, which were the same as in FIG. 6G. FIG. 6G graphically illustrates proliferation (percent confluence) DLD- 1 PTEN Knockout cells under the same treatment conditions as in FIG. 6F. Of note, the loss of PTEN in the isogenic sets of colon cancer lines does not uniformly alter the response to insulin in the setting of PI3K inhibition. In the context of PTEN loss, physiologic levels of insulin can restore normal proliferation in HCT1 l6s despite the presence of PI3K inhibitors.

FIG. 7A-7F illustrates blood glucose, tumor volume, ketone concentration, and triglyceride levels for KPC K8484 allografts treated with PI3K inhibitors with or without supplemental approaches to target systemic insulin feedback. FIG. 7A graphically illustrates blood glucose curves of mice from FIG. 3E-3G treated with control diet, ketogenic diet, metformin (250mg/kg), or canagliflozin (SGLT2i) (6mg/kg), after the first dose of BYL-719 (45mg/kg). FIG. 7B graphically illustrates tumor volumes of mice treated with the metabolic modifying agents as shown in FIG. 3A-3G without PI3K inhibitors. FIG. 7C graphically illustrates average tumor volume (lines) with scatter (points) for each of the indicated treatment cohorts. FIG. 7D graphically illustrates tumor volumes from an independent experiment of mice treated daily with B KM 120 with or without 6mg/kg of Canagliflozin administered 60 minutes prior to the PI3K treatment so that peak SGLT2 inhibition is aligned with peak blood glucose levels post PI3K inhibitor treatment. FIG. 7E graphically illustrates blood ketone of mice shown in FIG. 3A-D after a single treatment with B KM 120 with or without pre treatment with metformin, Canagliflozin, or the ketogenic diet as indicated. FIG. 7F graphically illustrates triglyceride levels as determined by Calorimetric assay of mice shown in FIG. 3A-D after a single treatment with BKM120 with or without pretreatment with metformin, Canagliflozin, or the ketogenic diet as indicated.

FIG. 8A-8H illustrates the role of inhibiting insulin receptor in the observed changes in tumor response. FIG. 8A shows a Western blot of cell lysates from K8484 cells used to generate xenografts in FIG. 4A after treatment with doxycycline to induce the sh-Renilla and sh-INSR hairpins as indicated. FIG. 8B graphically illustrates tumor volumes of the individual mice allografted with KPC-K8484 tumors as measured by caliper over time. FIG. 8C graphically illustrates a survival curve of mice in FIG. 8A. FIG. 8D graphically illustrates blood glucose levels from mice 240 minutes after the indicated treatments. Two of the glucose measurements in the OST 906 and BKM120 were beyond the range of the detector (e.g. >600). FIG. 8E graphically illustrates c-peptide levels from mice 240 minutes after the indicated treatments. FIG. 8F graphically illustrates the mass of mice over the course of the indicated treatments. As has been previously published mice lose 10-20% of their mass upon initiation of the ketogenic diet. FIG. 8G graphically illustrates tumor volume for the tumors in FIG. 8A for treatment with OSI-906, a INSR/IGFR inhibitor, or GDC-0032 with or without a keto diet. Treatment efficacy was significantly improved in PIK3CA + MYC mutant murine breast tumor allografts, ES- 278, grown in wild-type c57/bl6 when combined with ketogenic diet. FIG. 8H graphically illustrates tumor volumes of wildtype c57/bl6 mice baring KPC allografted tumors as measured by calipers over time. Mice were treated as indicated with combinations of BYL-719, the ketogenic diet, or insulin as in FIG. 4B. Mice in the ketogenic-B YL719-insulin cohort lost >20% of their body mass over the 1 week of treatment so the experiment was terminated at day 7.

FIG. 9A-9E illustrates the impact of PI3K inhibitor treatments upon Subject- derived xenograft models of bladder cancer and syngeneic allograft models of PIK3CA mutant breast cancer. FIG. 9A graphically illustrates tumor volume over time of a subject-derived endometrial xenografts (PDX) derived from a subject with bladder cancer (Subject C) and treated with the pan PI3K inhibitor GDC-0941 or the RI3K-b sparing compound GDC-0032 alone or with a ketogenic diet. Lines indicate average tumor volume of each treatment group, points indicate individual tumor volumes over time. Tumors were allowed to grow until their diameters were greater than 0.6cm prior to the initiation of treatment. FIG. 9B graphically illustrates the mass of the tumors (from FIG. 9 A) taken at the time harvest on day 12. FIG. 9C graphically illustrates tumor growth over time of mice with orthotopic allografts of an PIK3CA (F11047R) mutant murine breast cancer, ES272, treated as indicated with BKM120 alone or in combination with a ketogenic diet. FIG. 9D graphically illustrates tumor mass at harvest from mice with orthotopic allografts of an PIK3CA (H1047R) mutant murine breast cancer, ES272, treated as indicated with BKM120 alone or in combination with a ketogenic diet. FIG. 9E graphically illustrates the mass of the mice described in FIG. 9C-9D over time.

FIG. 10A-10E illustrate the impact of Capanilisib with or without ketogenic diet upon growth of orthotopic Kras-Tp53-Pdx-Cre (KPC) K8082 tumor model grown in the flank of wildtype c57/bl6 mice. FIG. 10A graphically illustrates survival of mice with KPC K8082 allografts grown in the flank and treated as indicated with BAY 80-6946 alone and in combination with a ketogenic diet pretreatment as indicated (p-value comparing BAY 80-6946 to the combination of BAY 80-6946 with ketogenic diet in this study is 0.0019 by Mantel-Cox Fog-rank test). FIG. 10B graphically illustrates the volume each tumor in this cohort graphed as individual lines. FIG. 10C graphically illustrates single blood glucose measurements of samples taken from the animals in FIG. 10B and 10C at 240 min after treatment. FIG. 10D graphically illustrates c-peptide measurements of samples taken from the animals in FIG. 10B and 10C at 240 min after treatment. FIG. 10E graphically illustrates the mass of the animals described in FIG. 10A-10D over the time on treatment. Tumors were allowed to grow until their diameters were >0.6cm prior to the initiation of treatment.

FIG. 11A-11F demonstrate the impact of BKMl20/Ketogenic combination on a syngeneic model of AMF. FIG. 11A shows IVIS images of AMF burden (as reported by mCherry) in mice over time of the indicated treatment. FIG. 11B graphically illustrates survival of mice having the syngeneic model of AMF treated with BKM120 alone or in combination with a ketogenic diet. Individual lines are shown for initiation of ketogenic diet before (pre) or at the same time as the initiation of B KM 120 treatment (Co), both demonstrate that B KM 120 efficacy is significantly enhanced by the addition of the ketogenic diet (p = 0.0316 and 0.349 for pre and co respectively). The asterisks (*) denote mice that were sacrificed due to paralysis resulting from AMF infiltrating the CNS rather than deaths typically seen in these mice due to tumor burden. Of note the mice in the BKM + Ketogenic diet group were frequently sacrificed due to paralysis which was not frequently a cause of mortality in the other treatment groups. FIG. 11C graphically illustrates disease burden of AMF model shown in FIG. 4A-4E as measured by percent AMF cells in bone marrow. FIG. 11D graphically illustrates spleen weights across the treatment groups. FIG. 11E graphically illustrates AML burden in mice that were pretreated with B KM 120 and/or a ketogenic diet to demonstrate that the effects observed in the AML studies are not the result of implantation issues related to the pretreatment. FIG. 11F shows images of mice treated as indicated with B KM 120 or the ketogenic diet where the diet and BKM120 therapy were initiated on the same day (co-treatment).

FIG. 12A-12C illustrate the in vivo impact of multiple approaches to targeting glucose/insulin feedback concurrently. FIG. 12A graphically illustrates blood glucose levels in wildtype c57/bl6 mice baring syngeneic K8484 KPC allografted tumors after treatment with a single dose of BKM120 after metformin pretreatment, SGLT2- inhibitor (SGLT2i) pretreatment, a ketogenic diet alone, or combinations thereof (N = 4/arm). FIG. 12B graphically illustrates ketone levels in wildtype c57/bl6 mice baring syngeneic K8484 KPC allografted tumors after treatment with a single dose of BKM120 with metformin pretreatment, SGLT2-inhibitor (SGLT2i) pretreatment, a ketogenic diet alone, or combinations thereof (N = 4/arm). FIG. 12C graphically illustrates c-peptide levels in wildtype c57/bl6 mice baring syngeneic K8484 KPC allografted tumors after treatment with a single dose of BKM120 with metformin pretreatment, SGLT2-inhibitor (SGLT2i) pretreatment, a ketogenic diet alone, or combinations thereof (N = 4/arm).

DETAILED DESCRIPTION

The present disclosure relates generally to compositions and methods for treating a disease or disorder associated with PI3K signaling. The methods can include administration of a modulator of glucose metabolism, use of a diet that influences the subject’s metabolic state, or a combination thereof. In some cases, the method includes co- administering a pathway inhibitor (such as an inhibitor of at least one kinase in the insulin-receptor PI3K/AKT/mTOR pathway) and administering a modulator of glucose metabolism. In some cases, the method of treating includes administering the pathway inhibitor to a subject who consumes a ketogenic diet during treatment. The disclosure further relates to pharmaceutical compositions comprising a pathway inhibitor and modulator of glucose metabolism.

Some studies indicate that administration of various modulators of glucose metabolism alone or administration of various inhibitors of the insulin- receptor/PI3K/AKT/mTOR pathway can be associated with an unfavorable safety profile and suboptimal anti-cancer activity. However, the types of combination therapies described herein can reduce or eliminate such side effects. For example, the inhibitors may be administered in lower doses or for shorter duration when combined with the other therapeutic agents and/or a ketogenic diet. Such combination therapies can provide reduced toxicity and avoid some of the side effects of monotherapies of drug-only therapies.

The insulin-receptor PI3K/AKT/mTOR pathway has not proven to be as druggable a target as those in the field would have hoped given its central role in cellular signaling. More than twenty PI3K inhibitors have entered clinical trials but only two (idelalisib and copanlisib) have been approved for use in cancer therapy. These agents are effective for treating lymphomas by primarily targeting the PIK3CD encoded enzyme pl 10D rather than the more broadly mutated PIK3CA-encoded enzyme, pl lOoc. Several drugs that target pl lOoc have entered approval trials, however the toxicity profile has been a challenge to manage and the responses have not correlated with PIK3CA mutations as would be expected. (Massacesi, C. et al. PI3K inhibitors as new cancer therapeutics: implications for clinical trial design. Onco Targets Ther 9, 203-210 (2016); Mayer, I. A. et al. A Phase lb Study of Alpelisib (BYL719), a PBKalpha-Specific Inhibitor, with Letrozole in ER+/HER2- Metastatic Breast Cancer. Clin Cancer Res 23, 26-34 (2017)).

As disclosed herein, pharmacologic blockade of PI3K elevates serum glucose and raises serum insulin. This hyperinsulinemia can re-activate the PI3K and mTOR signaling pathway in tumors within an hour or two of dosing, thereby compromising the effectiveness of PI3K blocking. The present disclosure provides a variety of interventions to reduce serum insulin. For example, the methods described herein can include administration of metformin, a Na + /Glucose co-transporter inhibitor, use of a ketogenic diet, or a combination thereof. As shown herein, a diverse group of human tumor organoids and cell lines grown as tumors in vivo, as well as genetically engineered tumors exhibit enhanced responses to PI3K inhibitors when the subjects were on a ketogenic diet. The enhanced responses were found in tumors with or without PIK3CA mutations. These results indicate that treating subjects with modulators of glucose metabolism and/or maintaining subjects on ketogenic diets enhances subject responses to PI3K inhibitors across a wide spectrum of cancers. Modulators of glucose metabolism and ketogenic diet improve drug efficacy with pathway inhibitors, including an array of agents that target the PI3K pathway in addition to BKM120 and BYL719, including the pan PI3K inhibitor GDC-0941, the RI3K-b sparing compound GDC-0032, the mTOR/PI3K dual inhibitor GDC-0980, the orally bioavailable inhibitor of class I PI3K alpha isoform serabelisib (TAK-l 17), and the recently approved PBK-oc/d inhibitor Copanlisib.

For example, the addition of the ketogenic diet to BKM120 reduced immunohistochemical markers of insulin signaling compared to tumors treated with BKM120 alone in PTEN/PIK3CA mutant endometrial PDX tumors. In these tumors, the ketogenic diet enhanced the ability of BKM120 to reduce levels of the phosphorylated insulin receptor, the phosphorylated AKT, and the phosphorylated S6. Such reduction in signaling correlated with decreased levels of cell proliferation as shown by Ki67 staining, and increased levels of apoptosis as indicated by cleaved caspase 3 staining.

In one embodiment, the disclosure includes a method of inhibiting cell proliferation, comprising contacting a cell with an effective amount of a modulator of glucose metabolism; and contacting the cell with an effective amount of at least one inhibitor of an insulin-receptor/PI3K/AKT/mTOR pathway, to thereby inhibit cell proliferation. In certain embodiments, at least one inhibitor inhibits at least one kinase in the insulin-receptor/PI3K/AKT/mTOR pathway to thereby inhibit cell proliferation. In particular embodiments, the insulin-receptor/PI3K/AKT/mTOR pathway is a mammalian, e.g., human, insulin-receptor/PI3K/AKT/mTOR pathway. The method may be practiced in vivo, ex vivo, or in vitro, in various embodiments.

In another embodiments, the disclosure includes a method of treating a cell proliferative disease, comprising: administering to a subject in need thereof an effective amount of at least one inhibitor of an insulin-receptor/PI3K/AKT/mTOR pathway; and administering to the subject a ketogenic diet and/or at least one modulator of glucose metabolism, to thereby inhibit cell proliferation in the subject.

In certain embodiments, the subject is administered the ketogenic diet. In certain embodiments, the subject is administered the modulator of glucose metabolism. In certain embodiments, the subject is administered both the ketogenic diet and the modulator of glucose metabolism. In certain embodiments, at least one inhibitor inhibits at least one kinase in the insulin-receptor PI3K/AKT/mTOR pathway to thereby inhibit cell proliferation. In particular embodiments, the insulin- receptor/PI3K/AKT/mTOR pathway is a mammalian, e.g., human, insulin- receptor/PI3K/AKT/mTOR pathway.

In particular embodiments of any of the methods disclosed herein, “administration” includes providing the pathway inhibitor, modulator of glucose metabolism, and/or ketogenic diet to the subject, e.g., to be ingested or administered at a later time, or providing a prescription for the pathway inhibitor, modulator of glucose metabolism, and/or ketogenic diet to the subject. In certain embodiments, “administration” of the ketogenic diet comprises instructing the subject to follow a ketogenic diet.

Methods and compositions disclosed herein can be used to increase the effectiveness of treatment with a pathway inhibitor (e.g., an inhibitor of the insulin- receptor/PI3K/AKT/mTOR signaling pathway). Thus, the disclosure provides a method for increasing the effectiveness or efficacy of treatment with a pathway inhibitor (e.g., an inhibitor of the insulin-receptor/PI3K/AKT/mTOR signaling pathway). Such a method can include treating a subject with an effective amount of the pathway inhibitor and optionally an effective amount of a modulator of glucose metabolism. The disclosure also provides a method for increasing the effectiveness or efficacy of treatment with a pathway inhibitor (e.g., an inhibitor of the insulin- receptor/PI3K/AKT/mTOR signaling pathway). Such a method can include treating a subject with an effective amount of the pathway inhibitor, wherein the subject consumes a ketogenic diet during treatment. The subject can be in need of such treatment. Alternatively, treatment can be used to reduce the incidence or onset of disease in the subject.

Methods and compositions disclosed herein may allow the use of a lower dosage of a pathway inhibitor. Thus, the disclosure provides a method for treating a disease or disorder associated with PI3K signaling, comprising administering to a subject in need thereof an effective amount of the pathway inhibitor (e.g., an inhibitor of the insulin-receptor/PBK/AKT/mTOR signaling pathway) and an effective amount of a modulator of glucose metabolism, wherein the effective amount of the pathway inhibitor is a lower amount than the amount effective in the absence of treatment with the modulator of glucose metabolism. The disclosure also provides a method for treating a disease or disorder associated with PI3K signaling, comprising administering to a subject in need thereof an effective amount of the pathway inhibitor (e.g., an inhibitor of the insulin- receptor/PI3K/AKT/mTOR signaling pathway) wherein the subject consumes a ketogenic diet during treatment, wherein the effective amount of the pathway inhibitor is a lower amount than the amount effective in the absence of the subject consuming the ketogenic diet during treatment. In certain embodiments, the effective amount of the pathway inhibitor is less than 90%, less than 80%, less than 70%, less than 60%, less than 50%, less than 40%, or less than 30%, when used in combination with the modulator of glucose metabolism and/or ketogenic diet, than is the amount of the pathway inhibitor when used alone.

Methods and compositions disclosed herein may allow for less frequent administration of a pathway inhibitor. Thus, the disclosure provides a method for treating a disease or disorder associated with PI3K signaling, comprising

administering to a subject in need thereof an effective amount of the pathway inhibitor (e.g., an inhibitor of the insulin-receptor/PI3K/AKT/mTOR signaling pathway) and an effective amount of a modulator of glucose metabolism, wherein the pathway inhibitor is administered less frequently than the frequency effective in the absence of treatment with the modulator of glucose metabolism. The disclosure also provides a method for treating a disease or disorder associated with PI3K signaling, comprising administering to a subject in need thereof an effective amount of a pathway inhibitor (e.g., an inhibitor of the insulin-receptor PI3K/AKT/mTOR signaling pathway) wherein the subject consumes a ketogenic diet during treatment, wherein the pathway inhibitor is administered less frequently than the frequency effective in the absence of the subject consuming the ketogenic diet during treatment. For example, at least one, or at least two, or at least three, or at least four, or at least five, or at least six, or at least seven, or at least eight, or at least ten, or at least thirteen, or at least fifteen fewer doses of the pathway inhibitor and/or the modulator of glucose metabolism can be administered over a duration of treatment than when the pathway inhibitor or the modulator of glucose metabolism is administered alone. Diseases or disorders associated with PI3K signaling

The term“disease or disorder associated with PI3K signaling” is intended to be construed expansively as referring to a disease or disorder caused by gain of function or loss of function in one or more members of the insulin- receptor/PBK/AKT/mTOR pathway. The“disease or disorder associated with PI3K signaling” is a disease or disorder of a mammal such as a human, a domesticated animal, a zoo animal, or an experimental animal.

The term“disease or disorder associated with PI3K signaling” is not limited to specific forms of PI3K. There are multiple PI3K genes and the phrase encompasses diseases and or disorders associated with any of them. The human PI3K genes include at least the genes listed in Table 1.

Table 1: PI3K genes

The term“disease or disorder associated with PI3K signaling” is not limited to diseases or disorders caused directly by gain of function or loss of function in PI3K but rather encompasses diseases and disorders caused by gain of function or loss of function in other genes of the insulin-receptor/PI3K/AKT/mTOR pathway. Upstream and downstream regulators of PI3K signaling include many drugable targets, such as signaling receptors, protein kinase B (known as AKT), mechanistic target of rapamycin (mTOR), as well as others.

The term“disease or disorder associated with PI3K signaling” is not limited to particular disease pathologies. Pathologically diverse diseases and disorders often have common mechanistic underpinnings. Mechanism-based treatments therefore are conventionally defined in terms of the target mechanism rather than by the tissue origin or pathological characteristics of the disease or disorder.

The diseases and disorders associated with PI3K signaling therefore include various types of disease or disorder. In one embodiment, a disease or disorder associated with PI3K signaling is a cell proliferative disease. In one embodiment, a disease or disorder associated with PI3K signaling is a neurodegenerative disease. In one embodiment, a disease or disorder associated with PI3K signaling is an inflammatory disease or condition. In one embodiment, a disease or disorder associated with PI3K signaling is a metabolic disease.

In some embodiments, the disease or disorder associated with PI3K signaling is a cell proliferative disease including, but not limited to, one or more leukemias (e.g., acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, acute myeloblastic leukemia, acute promyelocytic leukemia, acute myelomonocytic leukemia, acute monocytic leukemia, acute erythroleukemia, chronic leukemia, chronic myelocytic leukemia, chronic lymphocytic leukemia), polycythemia vera, lymphomas (Hodgkin’s disease, non-Hodgkin’s disease), Waldenstrom’s

macroglobulinemia, heavy chain disease, and solid tumors such as sarcomas and carcinomas (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma,

lymphangiosarcoma, lymphangioendothelio sarcoma, synovioma, mesothelioma, Ewing’s tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms tumor, cervical cancer, uterine cancer, testicular cancer, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodenroglioma, schwannoma, meningioma, melanoma, neuroblastoma, and retinoblastoma), or a combination thereof.

In some embodiments, the disease or disorder associated with PI3K signaling is a neurodegenerative disease including, but not limited to, brain trauma, spinal cord trauma, trauma to the peripheral nervous system, Alzheimer’s disease, Pick’s disease, diffuse Lewy body disease, progressive supranuclear palsy (Steel-Richardson syndrome), multisystem degeneration (Shy-Drager syndrome), motor neuron diseases including amyotrophic lateral sclerosis, degenerative ataxias, cortical basal degeneration, ALS-Parkinson’s-Dementia complex of Guam, subacute sclerosing panencephalitis, Huntington’ s disease, Parkinson’ s disease, synucleinopathies, primary progressive aphasia, striatonigral degeneration, Machado-Joseph

disease/spinocerebellar ataxia type 3 and olivopontocerebellar degenerations, Gilles De La Tourette’s disease, bulbar and pseudobulbar palsy, spinal and spinobulbar muscular atrophy (Kennedy’s disease), primary lateral sclerosis, familial spastic paraplegia, Werdnig-Hoffman disease, Kugelberg-Welander disease, Tay-Sach’s disease, Sandhoff disease, familial spastic disease, Wohlfart-Kugelberg-Welander disease, spastic paraparesis, progressive multifocal leukoencephalopathy, and prion diseases (including Creutzfeldt-Jakob, Gerstmann-Straussler-Scheinker disease, Kuru and fatal familial insomnia, age-related dementia, vascular dementia, diffuse white matter disease (Binswanger’s disease), dementia of endocrine or metabolic origin, dementia of head trauma and diffuse brain damage, dementia pugilistica or frontal lobe dementia, neurodegenerative disorders resulting from cerebral ischemia or infraction including embohc occlusion and thrombotic occlusion as well as intracranial hemorrhage of any type, intracranial and intravertebral lesions, hereditary cerebral angiopathy, hereditary amyloid, Down’ s syndrome, macroglobulinemia, secondary familial Mediterranean fever, Muckle-Wells syndrome, multiple myeloma, pancreatic- related amyloidosis, cardiac-related amyloidosis, chronic hemodialysis arthropathy, Finnish amyloidosis, Iowa amyloidosis, or a combination thereof.

In some embodiments, the disease or disorder associated with PI3K signaling is an inflammatory disorder including, but not hmited to, Type II diabetes, insulin resistance cardiovascular disease, arrhythmia, atherosclerosis, coronary artery disease, hypertriglyceridemia, dyslipidemia, retinopathy, nephropathy, neuropathy, obesity, and macular edema, ileitis, ulcerative colitis, Barrett’s syndrome, Crohn’s disease, or a combination thereof.

In some embodiments, the disease or disorder associated with PI3K signaling is a metabolic disease including, but not limited, Type II diabetes, insulin resistance cardiovascular disease, arrhythmia, atherosclerosis, coronary artery disease, hypertriglyceridemia, dyslipidemia, retinopathy, nephropathy, neuropathy, obesity, macular edema, or a combination thereof.

In some embodiments of the methods of the present disclosure, the subject is treatment naive. In some embodiments, the subject is resistant to hyperglycemia. In some embodiments, the subject is not hyperglycemic. In some embodiments, the subject is not hyperglycemic prior to treatment. In some embodiments, the subject is hypoglycemic. In some embodiments, the subject exhibits glycemic control. In some embodiments, the disease or disorder associated with PI3K signaling is associated with impaired glucose hemostasis. In some embodiments, the disease or disorder associated with PI3K signaling is associated with impaired glucose homeostasis. In some embodiments, the disease or disorder associated with PI3K signaling is associated with normal glucose homeostasis. In some cases, the subject is in need of treatment. However, in some cases the subject is treated to reduce the incidence or onset of a disease or disorder.

Modulators of Glucose Metabolism

The compositions and methods of the present disclosure can include, in some embodiments, a modulator of glucose metabolism. Various modulators of glucose metabolism can be used, and include without limitation lipids, amino acids, small- molecule drugs, antibodies, proteins, nucleic acids, and gene editing systems. In some embodiments, the modulator of glucose metabolism inhibits glucose metabolism. For example, the modulator of glucose metabolism can be a glucose-uptake inhibitor. In some embodiments, the glucose-uptake inhibitor can be an inhibitor of a sodium- glucose transport protein. In some embodiments, the modulator of glucose metabolism can be an inhibitor of a glucose transporter. In some embodiments, the glucose-uptake inhibitor can be selected from the group consisting of a sodium- glucose-linked transport protein 1 (SGLT1) inhibitor, a sodium-glucose-linked transport protein 2 (SGLT2) inhibitor, or a dual SGLT1/SGLT2 inhibitor. The modulator of glucose metabolism can, in some cases, be administered while the subject is consuming a ketogenic diet or while the subject is being administered a ketogenic diet.

Glucose is essential for energy production in the living body and the glucose transporter plays a critical role in various organs. Glucose transporters are classified into two families. Facilitative glucose transporters (GLUTs) transport glucose by facilitated diffusion. Sodium- glucose-linked transporters (SGLTs) co- transport sodium ion and glucose (or other substrates) using an electrochemical gradient across the membrane. (Harada et al. Role of sodium-glucose transporters in glucose uptake of the intestine and kidney. J Diabetes Investig. 2012 Aug 20; 3(4): 352-353.; Gallo et al. Probing SGLT2 as a therapeutic target for diabetes: Basic physiology and consequences. Diab Vase Dis Res. 2015 Mar; 12(2): 78-89; Wright et al. Biology of Human Sodium Glucose Transporters. Physiological Reviews. 9l(2):733-794 (2011).) The six SGLT protein and gene families are shown in Table 2. In certain embodiments, the modulator of glucose metabolism inhibits one or more SGLT protein.

Table 2: Sodium-dependent glucose transporter and tissue distribution

In some embodiments, the glucose-uptake inhibitor can be dapagliflozin, empagliflozin, canagliflozin, ipragliflozin, tofogliflozin, sergliflozin etabonate, remogliflozin etabonate, ertugliflozin, sotagliflozin, conagliflozin, or a combination thereof. In some embodiments, the modulator of glucose metabolism can be metformin. In some embodiments, the modulator of glucose metabolism can be an insulin receptor/IGF 1 receptor inhibitor. In some embodiments, the modulator of glucose metabolism can be linsitinib (OSI-906). Dapagliflozin (tradename Farxiga™ in the U.S. and Forxiga™ in the EU and Russia) is a drug of the gliflozin class, used to treat type-2 diabetes. It was developed by Bristol-Myers Squibb in partnership with AstraZeneca. Dapagliflozin has the following chemical structure:

Empagliflozin (tradename Jardiance™) is a drug of the gliflozin class, approved for the treatment of type-2 diabetes in adults in 2014. It was developed by Boehringer Ingelheim and Eli Lilly and Company. Empagliflozin is an inhibitor of SGLT2 and causes sugar in the blood to be excreted by the kidneys and eliminated in urine. Empagliflozin has the following chemical structure:

Canagliflozin (tradename Invokana™ or Sulisent™) is a medication used for the treatment of type-2 diabetes. It is of the gliflozin class or SGLT2 inhibitors class. Canagliflozin has the following chemical structure:

Ipragliflozin (tradenames Suglat™ and Jardiance™) is a pharmaceutical drug for treatment of type-2 diabetes. Ipragliflozin, jointly developed by Astellas Pharma and Kotobuki Pharmaceutical, was approved as an adjunct to diet and exercise to improve glycemic control in adults with type-2 diabetes mellitus. Ipragliflozin has the following chemical structure:

Tofogliflozin (codenamed CSG452) is an experimental drug for the treatment of diabetes mellitus and is being developed by Chugai Pharma in collaboration with Kowa and Sanofi. It is an inhibitor of SGLT2. Tofogliflozin has received its first global approval for this indication in Japan as either monotherapy or in combination with other antihyperglycemic agents. Tofogliflozin has the following chemical structure:

Sergliflozin etabonate (codenamed GW869682X) is an investigational anti diabetic drug being developed by GlaxoSmithKline. It is a SGLT2 inhibitor.

Sergliflozin etabonate has the following chemical structure:

Remogliflozin etabonate is a proposed drug of the gliflozin class for the treatment of non-alcoholic steatohepatitis (“NASH”) and type-2 diabetes.

Remogliflozin is being developed by Avolynt, Inc. Remogliflozin etabonate has the following chemical structure:

Ertugliflozin (tradename Steglatro™) is a drug for the treatment of type-2 diabetes. In the United States, it was approved by the Food and Drug Administration for use as a monotherapy and as a fixed dose combination with either sitagliptin or with metformin. Ertugliflozin has the following chemical structure:

Sotagliflozin, or LX4211 , is an orally-delivered small molecule compound that is currently in development by Lexicon Pharmaceuticals for the treatment of type- 1 and type-2 diabetes mellitus. Sotagliflozin inhibits SGLT1 and SGLT2.

Sotagliflozin has the following chemical structure:

Canagliflozin (trade name Invokana or Sulisent) is a medication used for the treatment of type-2 diabetes. It is of the gliflozin class or SGLT2 inhibitor class. Canagliflozin has the following chemical structure:

Metformin is a first- line medication for the treatment of type-2 diabetes, particularly in people who are overweight. Metformin has the following chemical structure:

Linsitinib (codename OSI-906) is an experimental drug candidate for the treatment of various types of cancer. In some cases, it can act as a modulator of glucose metabolism, a pathway inhibitor, or both. It is an inhibitor of the insulin receptor and of the insulin-like growth factor 1 receptor (IGF-1R). (Fassnacht et al. Linsitinib (OSI-906) versus placebo for subjects with locally advanced or metastatic adrenocortical carcinoma: a double -blind, randomized, phase 3 study. Lancet Oncology. 16(4):426-435 (2015).) Linsitinib has the following chemical structure:

In some cases, a modulator of glucose metabolism can be administered concurrently with a pathway inhibitor of the insulin-receptor/PBK/AKT/mTOR pathway. In some cases, a modulator of glucose metabolism can be administered before a pathway inhibitor of the insulin-receptor/PBK/AKT/mTOR pathway. In some cases, a modulator of glucose metabolism can be administered after a pathway inhibitor of the insulin-receptor/PBK/AKT/mTOR pathway is administered. A ketogenic diet can be consumed or administered before, during or after a modulator of glucose metabolism is administered.

Pathway inhibitor

In some embodiments, the disclosure provides a method of treating a disease or disorder associated with PI3K signaling that includes administering to a subject an effective amount of a pathway inhibitor. In some embodiments, the disclosure provides a method of treating a disease or disorder associated with PI3K signaling that includes administering to a subject an effective amount of a pathway inhibitor, where the subject is on a ketogenic diet during treatment. The subject can be in need of such treatment or the subject can be treated to reduce the incidence or onset of a disease or disorder. In some embodiments, the pathway inhibitor is capable of inhibiting at least one kinase in the insulin-receptor/PBK/AKT/mTOR pathway. In some embodiments, the pathway inhibitor is capable of inhibiting one or more targets such as any of INSR/IGFR, PI3K, AKT, mTOR, or a combination thereof. In some embodiments, the pathway inhibitor is capable of inhibiting a PI3K. In some embodiments, the pathway inhibitor is capable of inhibiting one or more of pl lO-a, r110-b, r110-g, r110-d, p85-a, r85-b, r55-g, r150, plOl, r87, PI3K-C2a, PI3K-C2 , PI3K-C2y, and Vps34.

In some embodiments, the pathway inhibitor can be idelalisib, copanlisib, buparlisib (BKM120), alpelisib (BYL719), taselisib (GDC-0032), pictilisib (GDC- 0941), apitolisib (GDC-0980), serabelisib (TAK-117), dactolisib, apelisib, MK2206, linsitinib (OSI-906), or a combination thereof.

Idelalisib (tradename Zydelig™, codenamed GS-1101 or CAL-101) is a drug used for the treatment of certain hematological malignancies. The substance acts as a phosphoinositide 3-kinase inhibitor. More specifically, it blocks pl lO-d, the delta isoform of the enzyme phosphoinositide 3-kinase (PI3K). It was developed by Gilead Sciences. Idelalisib has the following chemical structure:

Copanlisib (trade name Aliqopa™, codenamed BAY 80-6946), is a kinase inhibitor developed by Bayer Healthcare Pharmaceuticals Inc. and approved for the treatment of adult subjects experiencing relapsed follicular lymphoma who have received at least two prior systemic therapies. Copanlisib has the following chemical structure:

Buparlisib (codenamed BKM120) is an orally bioavailable specific oral inhibitor of the pan-class I phosphatidylinositol 3-kinase (PI3K) family of lipid kinases with potential antineoplastic activity. Buparlisib specifically inhibits class I PIK3 in an ATP-competitive manner, thereby inhibiting the production of the secondary messenger phosphatidylinositol-3,4,5-trisphosphate and activation of the PI3K signaling pathway. Buparlisib has the following chemical structure:

Some studies indicate that administration of BKM120 alone can be associated with an unfavorable safety profile and minimal antitumor activity during advanced or recurrent endometrial carcinoma. However, the types of combination therapies described herein can reduce or eliminate such side effects. For example, BKM120 may be administered in lower doses when combined with the other therapeutic agents and/or a ketogenic diet. Such combination therapies can provide reduced toxicity and avoid side effects.

Alpelisib (codename BYL719) is an orally bioavailable phosphatidylinositol 3 -kinase (PI3K) inhibitor with potential antineoplastic activity. Alpelisib specifically inhibits PIK3, thereby inhibiting the activation of the PI3K signaling pathway.

Alpelisib has the following chemical structure:

Taselisib (codename GDC-0032), developed by Roche, is an orally bioavailable inhibitor of the class I phosphatidylinositol 3-kinase (PI3K) alpha isoform (PIK3CA), with potential antineoplastic activity. Taselisib selectively inhibits PIK3CA and its mutant forms, which may result in tumor cell apoptosis and growth inhibition in PIK3CA-expressing tumor cells. Taselisib has the following chemical structure:

Pictilisib (codename GDC-0941), developed by Roche, is a potent inhibitor of RI3Ka/d with IC50 of 3 nM in cell-free assays, with modest selectivity against pi 10b (11-fold) and pi 10g (25-fold). Pictilisib has the following chemical structure:

Apitolisib (codename GDC-0980, RG7422) is a potent, class I PI3K inhibitor for RI3Ka/b/d/g with IC50 of 5 nM/27 nM/7 hM/14 nM in cell-free assays, respectively. Apitolisib is also an mTOR inhibitor with Ki of 17 nM in a cell-free assay. Apitolisib has the following chemical structure:

Serabelisib (also known as MLN1117, INK1117, and TAK-117) is an orally bioavailable inhibitor of the class I phosphoinositide 3-kinase (PI3K) alpha isoform. Serabelisib selectively inhibits PI3K alpha kinase, including mutations of PIK3CA, in the PI3K/Akt/mTOR pathway. Serabelisib has the following structure:

Dactolisib (codenamed NVP-BEZ235 and BEZ-235) is an imidazoquinoline derivative acting as a PI3K inhibitor. Dactolisib also inhibits mTOR. Dactolisib has the following chemical structure:

MK2206 is an orally bioavailable allosteric inhibitor of the serine/threonine protein kinase Akt (protein kinase B) with potential antineoplastic activity. MK2206 binds to and inhibits the activity of Akt in a non- ATP competitive manner. MK2206 has the following chemical structure:

Linsitinib (codename OSI-906) is an experimental drug candidate for the treatment of various types of cancer. In some cases, it can act as a modulator of glucose metabolism, a pathway inhibitor, or both. It is an inhibitor of the insulin receptor and of the insulin-like growth factor 1 receptor (IGF-1R). (Fassnacht et al. Linsitinib (OSI-906) versus placebo for subjects with locally advanced or metastatic adrenocortical carcinoma: a double -blind, randomized, phase 3 study. Lancet Oncology. 16(4):426-435 (2015).) Linsitinib has the following chemical structure:

In some cases, combinations of inhibitors of at least one kinase in the insulin- receptor/PBK/AKT/mTOR pathway can be used or administered to a subject. For example, the inhibitor of at least one kinase in the insulin-receptor/PBK/AKT/mTOR pathway can be a combination of two or more of buparlisib (BKM120), taselisib (GDC-0032), pictilisib (GDC-0941), or linsitinib (OSI-906).

In particular embodiments of methods that comprise the use of both a pathway inhibitor and a modulator of glucose metabolism, and compositions that comprise both a pathway inhibitor and a modulator of glucose metabolism, a combination comprising any of the combinations of agent shown in any row of Table 6 may be used.

Table 6. Combinations of modulators of glucose metabolism and

pathway inhibitors

In some cases, a pathway inhibitor of the insulin-receptor/PI3K/AKT/mTOR pathway can be administered concurrently with a modulator of glucose metabolism.

In some cases, a pathway inhibitor of the insulin-receptor/PI3K/AKT/mTOR pathway can be administered before a modulator of glucose metabolism. In some cases, a pathway inhibitor of the insulin-receptor/PI3K/AKT/mTOR pathway can be administered after a modulator of glucose metabolism. A ketogenic diet can be consumed or administered before, during or after a pathway inhibitor of the insulin- receptor/PBK/AKT/mTOR pathway is administered. Ketogenic Diet

In some embodiments, the disclosure provides a method of treating a disease or disorder associated with PI3K signaling where the subject consumes a ketogenic diet during treatment. In some embodiments, the method includes administering a ketogenic diet to the subject.

In some cases, the subject can be administered a pathway inhibitor of the insulin-receptor/PI3K/AKT/mTOR pathway while consuming or being administered a ketogenic diet. In some embodiments, the method can include administering an effective amount of a modulator of glucose metabolism to the subject while administering the ketogenic diet and/or while administering a pathway inhibitor of the insulin-receptor/PI3 K/AKT/mT OR pathway.

In some embodiments, the subject consumes a ketogenic diet prior to administration of the pathway inhibitor. In some embodiments, the subject consumes a ketogenic diet after administration of the pathway inhibitor. In some embodiments, the subject consumes a ketogenic diet concurrently with administration of the pathway inhibitor.

Ketogenic diets have been used in epileptic subjects since the l970s and have been shown to reduce blood glucose levels and increase insulin sensitivity as compared to normal western diets. (Hopkins, B. D., Goncalves, M. D. & Cantley, L.

C. Obesity and Cancer Mechanisms: Cancer Metabohsm. J Clin Oncol 34, 4277- 4283, doi:l0.l200/JC0.20l6.67.97l2 (2016); Sampaio, L. P. Ketogenic diet for epilepsy treatment. Arq Neuropsiquiatr 74, 842-848, doi:l 0.1590/0004- 282X20160116 (2016).) In some embodiments, ketogenic diet includes a high-fat, low-carbohydrate diet. In some embodiments, the ketogenic diet is stricter than the modified Atkins diet. In some embodiments, the ketogenic diet includes consuming defined amounts of calories, fluids, and proteins. A ketogenic diet is available at most major hospitals.

A classic ketogenic diet is defined by a set ratio of grams of fat to grams of carbohydrate plus protein. The most common ratio is 3:1 or 4:1 grams of fat to grams of carbohydrate plus protein. In this classic ketogenic diet approximately 90% of the energy comes from fat and 10% from carbohydrate and protein combined. Calories are typically restricted to 80-90% of the daily recommendations for age. In some cases, fluid restriction is imposed on the subject consuming the diet. A ketogenic diet useful for experimental purposes is, in one example, AIN- 76A purified rat and mouse diet, available from ThermoFischer®, or an equivalent thereof.

In some embodiments, the ketogenic diet includes at most 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, or 15% protein, with the remainder of the diet made up of fat, fiber, ash, and carbohydrates. In some embodiments, the ketogenic diet includes at most 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, or 10% carbohydrates, with the remainder of the diet made up of fat, fiber, ash, and protein. In some embodiments, the ketogenic diet includes fat measured in grams and carbohydrates and proteins collectively measured in grams in a ratio of 2.5, 2.6, 2.7, 2.8, 2.9, 3, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4, 4.1, 4.2, 4.3, 4.4, or 4.5 to one (1) of fat to

carbohydrate/protein. A comparison of a ketogenic diet with a normal diet is shown below as Table 3. Although the ketogenic diet shown in Table 3 was used for experimental purposes, the ketogenic diet administered or consumed by a subject pursuant to one or more of the methods described herein can have similar percentages of fat, protein, carbohydrate, ash, and the other components listed in Table 3.

In some cases, such a ketogenic diet can involve ingestion of a 3 :1 ratio of ketogenic-to-antiketogenic macromolecules, which results in approximately 85% fat, 12% protein, and 3% carbohydrates. There can be a diverse mixture of fats. For example, the fats can include those from plants, nuts, and animal products. The diet can be actively managed by dieticians who interact with patients on the diet on a specific time table, for example, on a weekly basis or on a monthly basis. Such a diet can obtain up to 80% compliance, up to 90% compliance, up to 95% compliance, up to 96% compliance, up to 98% compliance, up to 99% compliance, or even up to 100% compliance. For example, 100% compliance over 4 weeks was achieved in an ongoing pilot study in women with endometrial cancer.

Routes of Administration, Formulations, and Dosages

The disclosed methods of treatment can be accomplished via any mode of administration for therapeutic agents. These modes include systemic or local administration such as oral, nasal, parenteral, transdermal, subcutaneous, vaginal, buccal, rectal or topical administration modes.

Depending on the intended mode of administration, the disclosed

compositions can be in sohd, semi-solid or liquid dosage form, such as, for example, injectables, tablets, suppositories, pills, time-release capsules, elixirs, tinctures, emulsions, syrups, powders, liquids, suspensions, or the like, sometimes in unit dosages and consistent with conventional pharmaceutical practices. Likewise, the compositions can also be administered in intravenous (both bolus and infusion), intraperitoneal, subcutaneous or intramuscular form, and all using forms well known to those skilled in the pharmaceutical arts.

Illustrative pharmaceutical compositions are tablets and gelatin capsules comprising a pathway inhibitor (and/or a modulator of glucose metabolism) and a pharmaceutically acceptable carrier, such as a) a diluent, e.g., purified water, triglyceride oils, such as hydrogenated or partially hydrogenated vegetable oil, or mixtures thereof, corn oil, olive oil, sunflower oil, safflower oil, fish oils, such as EPA or DHA, or their esters or triglycerides or mixtures thereof, omega-3 fatty acids or derivatives thereof, lactose, dextrose, sucrose, mannitol, sorbitol, cellulose, sodium, saccharin, glucose and/or glycine; b) a lubricant, e.g. , silica, talcum, stearic acid, its magnesium or calcium salt, sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and/or polyethylene glycol; for tablets also; c) a binder, e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, magnesium carbonate, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, waxes and/or

polyvinylpyrrolidone, if desired; d) a disintegrant, e.g. , starches, agar, methyl cellulose, bentonite, xanthan gum, algic acid or its sodium salt, or effervescent mixtures; e) absorbent, colorant, flavorant and sweetener; i) an emulsifier or dispersing agent, such as Tween 80, Labrasol, HPMC, DOSS, caproyl 909, labrafac, labrafil, peceol, transcutol, capmul MCM, capmul PG-12, captex 355, gelucire, vitamin E TGPS or other acceptable emulsifier; and/or g) an agent that enhances absorption of the compound such as cyclodextrin, hydro xypropyl-cyclodextrin, PEG400, PEG200.

Liquid, particularly injectable, compositions can, for example, be prepared by dissolution, dispersion, etc. For example, the pathway inhibitor and/or modulator of glucose metabolism is dissolved in or mixed with a pharmaceutically acceptable solvent such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to thereby form an injectable isotonic solution or suspension. Proteins such as albumin, chylomicron particles, or serum proteins can be used to solubilize the disclosed compounds.

The disclosed pharmaceutical compositions can be also formulated as a suppository that can be prepared from fatty emulsions or suspensions; using polyalkylene glycols such as propylene glycol, as the carrier.

The disclosed pharmaceutical compositions can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, containing cholesterol, stearylamine or

phosphatidylcholines. In some embodiments, a film of lipid components is hydrated with an aqueous solution of drug to a form hpid layer encapsulating the pathway inhibitor and/or modulator of glucose metabolism, as described in U.S. Pat. No. 5,262,564 which is hereby incorporated by reference in its entirety.

Disclosed pharmaceutical compositions can also be dehvered by the use of monoclonal antibodies as individual carriers to which pathway inhibitor and/or modulator of glucose metabolism are coupled. Pathway inhibitor and/or modulator of glucose metabolism can also be coupled with soluble polymers as targetable drug carriers. Such polymers can include polyvinylpyrrolidone, pyran copolymer, poly(hydroxypropyl)methacrylamide -phenol, poly(hydroxyethyl)-aspanamide phenol, or poly(ethyleneoxide)-polylysine substituted with palmitoyl residues. Furthermore, pathway inhibitor and/or modulator of glucose metabohsm can be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels. In one embodiment, pathway inhibitor and/or modulator of glucose metabohsm are not covalently bound to a polymer, e.g., a polycarboxylic acid polymer, or a polyacrylate.

Parental injectable administration is generally used for subcutaneous, intramuscular or intravenous injections and infusions. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions or sohd forms suitable for dissolving in liquid prior to injection.

Pharmaceutical compositions can be prepared according to conventional mixing, granulating or coating methods, respectively, and the present pharmaceutical compositions can contain from about 0.1 % to about 99%, from about 5% to about 90%, or from about 1% to about 20% of modulators of glucose metabolism and/or kinase inhibitor by weight or volume.

The dosage regimen is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the subject; the severity of the condition to be treated; the route of administration; the renal or hepatic function of the subject; and the particular compound employed. A physician or veterinarian of ordinary skill in the art can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the disease or disorder.

In some cases, effective dosage amounts of pathway inhibitor and/or modulator of glucose metabolism, when used for the indicated effects, range from about 0.5 mg to about 5000 mg of the pathway inhibitor and/or modulator of glucose metabolism as needed to treat the disease or disorder. Compositions for in vivo or in vitro use can contain about 0.5, 5, 20, 50, 75, 100, 150, 250, 500, 750, 1000, 1250, 2500, 3500, or 5000 mg of the pathway inhibitor and/or modulator of glucose metabolism, or, in a range of from one amount to another amount in the list of doses. In one embodiment, the compositions are in the form of a tablet that can be scored.

In some embodiments, the disclosure provides a pharmaceutical composition that includes a modulator of glucose metabolism and an inhibitor of at least one kinase in the insulin-receptor PI3K/AKT/mTOR pathway.

In some embodiments, the disclosure provides a pharmaceutical composition that includes a glucose-uptake inhibitor and an inhibitor of at least one kinase in the insulin-receptor/PI3K/AKT/mTOR pathway.

In some embodiments, the disclosure provides a kit comprising a

pharmaceutical composition comprising a glucose -uptake inhibitor and a pharmaceutical composition comprising an inhibitor of at least one kinase in the insulin-receptor/PBK/AKT/mTOR pathway.

Kits

Also described herein is a kit that includes a packaged pharmaceutical composition for controlling, preventing or treating a cell prohferative disease. The kits of the invention can be designed for controlling, preventing or treating cell proliferation or a cell prohferative disease. In one embodiment, the kit or container holds at least one inhibitor of at least one kinase in the insulin-receptor/PBK/AKT/mTOR pathway. The kit can also include a modulator of glucose metabolism. Each inhibitor or modulator can be packaged separately. Alternatively, one or more inhibitors or modulators can be packaged or formulated together.

Hence, the kit or container can hold at least one inhibitor of at least one kinase in the insulin-receptor/PBK/AKT/mTOR pathway, at least one modulator of glucose metabolism, or a combination thereof.

The kit can also hold instructions for administering the at least one inhibitor of at least one kinase in the insulin-receptor/PBK/AKT/mTOR pathway, the at least one modulator of glucose metabolism, or a combination thereof.

In another embodiment, the kit holds one or more components of a ketogenic diet. Each component of a ketogenic diet can be packaged separately. Alternatively, one or more components of a ketogenic diet can be packaged together.

In another embodiment, the kit or container holds instructions for starting and/or maintaining a ketogenic diet with instructions for using the at least one inhibitor of at least one kinase in the insulin-receptor/PBK/AKT/mTOR pathway, at least one modulator of glucose metabolism, or a combination thereof. For example, the instructions can include methods for mixing components of a ketogenic diet, methods for obtaining supplemental components of a ketogenic diet, time tables for consumption of components of the ketogenic diet, or a combination thereof.

The kits of the invention can also include containers with tools useful for administering the compositions and maintaining a ketogenic diet as described herein. Such tools include syringes, swabs, catheters, antiseptic solutions, package opening devices, forks, spoons, straws, and the like.

The compositions, kits, and/or methods described herein are useful for treatment of cell proliferative diseases such as cancer or cell-proliferative disorder, a metabolic disorder, neurodegenerative disease, or an inflammatory disease. For example, the compositions, kits, and/or methods described herein can reduce the incidence or progression of such diseases by 1% or more, 2% or more, 3% or more, 5% or more, 7% or more, 10% or more, 15% or more, 20% or more, 25% or more, 30% or more, 40% or more, or 50% or more compared to a control. Such a control can be the initial frequency or previous rate of progression of the disease of the subject. The control can also be an average frequency or rate of progression of the disease. For example, when treating cancer, the compositions and/or methods described herein can reduce tumor volume in the treated subject by 1% or more, 2% or more, 3% or more, 5% or more, 7% or more, 10% or more, 15% or more, 20% or more, 25% or more, 30% or more, 40% or more, or 50% or more compared to a control. Such a control can be the initial tumor volume. In some cases, the compositions and/or methods described herein can reduce the incidence or progression of such diseases by at least 2-fold, or at least 3-fold, or at least 5-fold, or at least 10-fold compared to a control.

EXAMPLES

The following Examples are given for the purpose of illustrating various embodiments of the disclosure and are not meant to limit the present disclosure in any fashion. Changes therein and other uses which are encompassed within the spirit of the disclosure, as defined by the scope of the claims, will be recognized by those skilled in the art. Appendix A provides further description of experimental work involved in the development of the methods and compositions described herein.

Overview

Gain of function mutations in PIK3CA, encoding the insulin-activated phosphoinositide-3-kinase (PI3K), and loss of function mutations in PTEN, a phosphatase that degrades the phosphoinositide lipids generated by PI3K, are among the most frequent events in human cancers. Yet, pharmacological inhibition of PI3K, using diverse classes of inhibitors, has resulted in variable clinical responses in humans, raising the possibility of an inherent mechanism of resistance to PI3K inhibition. Here, the inventors show that pharmacologic blockade of PI3K not only elevates serum glucose but also dramatically raises serum insulin. This

hyperinsulinemia re-activates the PI3K and mTOR signaling pathway in tumors within an hour or two of dosing, thereby compromising the effectiveness of blocking PI3K. The inventors demonstrate herein a variety of interventions to reduce serum insulin including metformin, a Na + /Glucose co -transporter inhibitor, and a ketogenic diet. The inventors found that a diverse group of human tumor organoids and cell fines grown as tumors in mice, as well as genetically engineered mouse tumors, exhibited enhanced responses to PI3K inhibitors when the mice were on a ketogenic diet. The enhanced responses were found in tumors with or without PIK3CA mutations. These results demonstrate that maintaining patients on ketogenic diets or administering modulators of glucose metabolism (for example, therapies that lower serum insulin) could enhance patient responses to PI3K inhibitors across a wide spectrum of cancers.

The PI3K pathway is one of the most frequently mutated pathways in human cancer, with mutations in PIK3CA being observed at similar frequency to mutations in KRAS. More than twenty PI3K inhibitors have entered clinical trials but only two (idelalisib and copanlisib) have been approved for use in cancer therapy. These agents are effective for treating lymphomas by primarily targeting the PIK3CD encoded enzyme pl 10A rather than the more broadly mutated PIK3CA-encoded enzyme, pl lOoc. Several drugs that target pl lOoc have entered approval trials, however the toxicity profile, including hyperglycemia, has been a challenge to manage and the responses have not correlated with PIK3CA mutations as would be expected. Since pl lOoc mediates virtually all cellular responses to insulin, hyperglycemia is an expected on-target effect of pl lOoc inhibitors. Blocking insulin signaling promotes glucose release from the liver and prevents glucose uptake into skeletal muscle and fat. The resulting acute hyperglycemia varies from patient to patient and is often resolved within a few hours of drug administration due to compensatory insulin release from the pancreas. Many patients with borderline insulin resistance must discontinue therapy due to the inability to endogenously control serum glucose levels with glucose lowering drugs such as metformin during therapy. Experimental subjects subjected to prolonged PI3K inhibition display reduced glucose tolerance and increased insulin resistance.

Here, the inventors show that the systemic glucose-insulin feedback caused by targeted inhibition of this pathway activates PI3K signaling in several tumors, even in the presence of PI3K inhibitors. The feedback hyperinsulinemia disclosed herein can be prevented using dietary or pharmaceutical approaches, which greatly enhances the efficacy/toxicity ratios of insulin receptor/PI3K/AKT/mTOR pathway inhibitors. These findings have direct clinical implications for the multiple pl lOoc inhibitors that are in clinical trials and provide a means to significantly increase treatment efficacy for patients with a myriad of tumor types. Example 1: Disruption of systemic glucose homeostasis

using therapeutic doses of compounds targeting a variety of kinases

in the insulin receptor/PI3K/mTOR pathway

Hyperglycemia has largely been treated as a treatment-related complication that requires management in only a subset of patients for whom the hyperglycemia becomes persistent. Due to the body’ s normal glycemic regulation, patients treated with these agents experience some degree of systemic hyperinsulinemia as the pancreas attempts to normalize serum glucose levels. Since insulin is a potent stimulator of PI3K signaling in tumors and can have profound effects on cancer progression, the inventors hypothesized that treatment-induced hyperinsulinemia limits the therapeutic potential of agents targeting the PI3K pathway.

Wild-type mice were treated with therapeutic doses of compounds targeting a variety of kinases in the insulin receptor/PI3K/mTOR pathway, including inhibitors of INSR/IGFR, PI3K, AKT, and mTOR, and after treatment their blood glucose levels were monitored over time (FIG. 1A, FIG. 5A, FIG. 5B). The inventors observed that many of these agents cause significant increases in blood glucose levels. Importantly, the inventors noticed that the hyperglycemia resolved after only a few hours without additional intervention, suggesting that PI3K signaling had been reactivated in muscle and liver despite the presence of the drug. For each of the agents that caused an increase in blood glucose, there was also an increase in the amount of insulin released in the serum as measured by ELIS As for insulin over time (FIG. 1B) and c-peptide, which is clinically used as a surrogate for insulin over time (FIG. 1C, FIG. 5C, FIG. 5D).

To assess if these PI3K inhibitor-induced spikes in glucose and insulin were affecting tumors, the inventors performed fluorodeoxyglucose positron emission tomography FDG-PET on mice bearing orthotopic Kras-Tp53-Pdx-Cre (KPC) tumor allografts in the pancreas. The inventors observed an increase in glucose uptake in these tumors in the acute setting after PI3K inhibition as compared to vehicle treated mice, indicating that the spikes in insulin could be causing transient increases in glucose uptake in these tumors (FIG. 1D). Example 2: Insulin stimulates PI3K signaling in the context of PI3K inhibition

To test whether or not these spikes in insulin were stimulating PI3K signaling in the context of PI3K inhibition, KPC cells were treated in vitro with the PI3K inhibitors in the presence or absence of 10 ng/ml insulin, the level observed in the mice within 15-30 min after drug administration (FIG. 1D). This level of insulin was sufficient to partially rescue PI3K signaling in the continued presence of PI3K inhibitors as indicated by partial re-activation of phosphorylated AKT (pAKT) and almost complete reactivation of phosphorylated S6 (pS6), a reporter of growth signaling through the mTORCl complex (FIG. 2A). In addition, this enhanced signaling correlated with a partial recovery of cellular proliferation (FIG. 2B-2C).

Similar effects of insulin stimulating proliferation in the presence of a PI3K inhibitor were observed in a variety of other tumor cell lines and patient derived organoids from breast, endometrium and prostate tumors (FIG. 6A-6G). The amount of stimulation was not uniform across all cell lines, as would be expected in tumors with variable expression of the insulin receptor and differential dependence on PI3K signaling for growth. These observations support the conclusions that insulin is a potent activator of PI3K signaling in certain tumors, and that elevation of serum insulin following PI3K inhibitor administration can reactivate PI3K signaling and potentially other PI3K-independent responses to insulin in both normal tissues and tumors.

Example 3: Metform, SGLT2 Inhibitors, Ketogenic Diet

Research and care for diabetic patients has resulted in the development of numerous approaches to manage blood glucose and insulin levels. Utilizing these tools, the inventors sought to identify approaches to augment PI3K inhibitor therapies by circumventing the acute glucose/insulin feedback. Metformin and sodium-glucose co-transporter 2 (SGLT2) inhibitors were evaluated, as well as a ketogenic diet in our murine models of cancer.

Treatment-naive mice bearing KPC allografts were placed on a ketogenic diet or treated with metformin for 10 days prior to a single treatment with B KM 120. During this treatment, blood glucose was monitored and, after 3 hours, c-peptide (a surrogate for blood insulin) was evaluated (FIG. 3A-3B). In some mice, tumors were harvested at 90 minutes and stained for the pS6 (FIG. 3C-3D). These results demonstrated that pre treatment with metformin had only minimal impact on the PI3K inhibitor-induced elevation in blood glucose and insulin levels or on growth signaling through mTORCl . In contrast, both the SGLT2 inhibitor and the ketogenic diet approaches decreased the resulting hyperglycemia and reduced the total insulin that was released in response to BKM120 treatment, and that these effects correlated with reduced signaling through mTORCl in the tumor. Similar effects were seen in mice treated with the pl lOoc specific inhibitor BYL-719 where the inventors observed an enhanced response of KPC allografts to BYL-719 in a manner concordant with the relative ability of each treatment to reduce serum insulin levels (FIG. 3E-G, FIG. 7A- D).

Example 4: Knockdown or Inhibition of Insulin Receptor

Various hormones and metabolites can reactivate growth in the setting of PI3K inhibition. To test if the enhancement in tumor signaling and growth is directly mediated by insulin, the inventors generated a doxycycline-inducible shRNA to target the insulin receptor in KPC tumors (FIG. 4A). Induction of this hairpin in the absence of a PI3K inhibitor had little effect on tumor growth. Flowever, induction of the hairpin at the time of BYL719 treatment initiation resulted in tumor shrinkage that was almost as effective as the ketogenic diet (FIG. 4A). This result indicates that the insulin receptor is not playing a major role in the growth of this tumor until supra- physiologic amounts of insulin are released following treatment with a PI3K inhibitor. The specificity of this effect was further corroborated by combining the PI3K inhibitor, BKM120, with the insulin receptor/IGFl receptor inhibitor, OSI-906, which resulted in a more effective response on the growth of KPC allografts than either drug alone (FIG. 8B-8G).

Example 5: Exogenous Insulin

To further test whether the improved response to PI3K inhibitors while on a ketogenic diet is a consequence of lowering blood insulin levels, the inventors attempted to“rescue” the PI3K reactivation using exogenous insulin. A cohort of the mice bearing Pik3ca mutant breast allografts were treated with the combination of a ketogenic diet and BYL-719, and then given 0.4mU of insulin 15 minutes after each dose of PI3K inhibitor (FIG. 4B). The addition of insulin dramatically reduced the therapeutic benefit of supplementing PI3K inhibitor therapy with a ketogenic diet, the addition of insulin also rescued tumor growth in allografted KPC tumors (FIG. 8H). It should be noted that the combination of the ketogenic diet, insulin, and BYL-719 was not well-tolerated in young mice so that ethical endpoints were reached due to weight loss in the KPC tumors after only one week of treatment.

Together these data demonstrate modulation of glucose metabolism improves responses to PI3K inhibitors by reducing blood insulin and the consequent ability of insulin to activate the insulin receptor in tumors. As demonstrated herein, modulation of glucose metabolism improves responses to PI3K inhibitors in tumors with a wide range of genetic aberrations. Therapeutic benefit was observed in patient-derived xenograft for advanced endometrial adenocarcinoma (harboring a PTEN deletion and PIK3CA mutation) and bladder cancer (FGFR Amplified) as well as syngeneic allograft for Pik3ca mutant breast cancer and MLL-AF9 driven Acute Myeloid Leukemia (FIG. 4C, FIGs. 9A-9E, FIGs. 10A-10E, FIGs. 11A-11E).

Modulation of glucose metabolism improved drug efficacy with an array of agents that target the PI3K pathway in addition to BKM120 and BYL719, including the pan PI3K inhibitor GDC-0941, the RI3K-b sparing compound GDC-0032, the mTOR/PI3K dual inhibitor GDC-0980, and the recently approved PBK-oc/d inhibitor Copanlisib (FIG. 5). In some cases, the ketogenic diet alone had variable effects in different tumor models indicating that the dietary changes themselves were insufficient to cause the tumor responses observed across the murine models. In some instances, such as the AML model, the ketogenic diet alone accelerated disease progression suggesting that this diet may be detrimental for some cancer patients when used in isolation.

The data shown herein indicates that insulin feedback is limiting the efficacy of PI3K inhibition in various hematological malignancies and sold tumors. By reducing the systemic insulin response, the addition of the ketogenic diet to BKM120 reduced immunohistochemical markers of insulin signaling compared to tumors from mice treated with BKM120 alone in PTEN/PIK3CA mutant endometrial PDX tumors. In these exemplary tumors, the ketogenic diet enhanced the ability of BKM120 to reduce levels of phosphor ylated insulin receptor, phosphorylated AKT and phosphorylated S6 and this reduction in signaling correlated with decreased levels of proliferation as shown by Ki67 staining, and increased levels of apoptosis as indicated by cleaved caspase 3 staining (FIG. 4D-E). While these data do not exclude insulin-independent effects of combining PI3K inhibition with anti-glycemic therapy, they demonstrate this modulation of glucose metabolism significantly increases the therapeutic efficacy of these pathway inhibitors. In light of these results, it may also be important to think about how common clinical practices such as IV glucose administration, glucocorticoid use, or providing patients with glucose-laden nutritional supplements may impact therapeutic responses. Therapeutic agents that target this critical oncogenic pathway should be paired with strategies such as administration of modulators of glucose metabolism or ketogenic diet to limit this self-defeating systemic feedback.

Example 6: Multiple Approaches to Target Glucose/Insulin Feedback

This Example illustrates the in vivo impact of multiple approaches to concurrently target glucose/insulin feedback.

FIG. 12A-12C graphically illustrate blood glucose, ketone, and c-peptide levels in wildtype c57/bl6 mice baring syngeneic K8484 KPC allografted tumors after treatment with a single dose of BKM120 with metformin pretreatment, SGLT2- inhibitor (SGLT2i) pretreatment, a ketogenic diet alone, or combinations of such treatments (N = 4/arm). These data demonstrate that combining these approaches can have an added effect in controlling glucose/insulin feedback and can increase treatment efficacy by profoundly enhancing the response of the systemic metabolism to PI3K inhibition.

Example 7 : Methods

Mice procurement and treatment

All animal studies were conducted following IACUC approved animal protocols (#2013-0116) at Weill Cornell Medicine and (AC-AAAQ5405) at Columbia University. Mice were maintained in temperature- and humidity-controlled specific pathogen- free conditions on a l2-hour light/dark cycle and received a normal chow diet (PicoLab Rodent 20 5053 lab Diet St. Louis, MO) or ketogenic diet (Thermo- Fisher AIN-76A) with free access to drinking water. Diets were composed as indicated in Table 3.

Table 3: Ketogenic Diet compared to a Normal Diet

For solid tumor studies, Nude (genotype) and C57/BL6 mice were purchased at 8 weeks of age from Jackson laboratories (Bar Harbor, ME). They were injected with 0.5-1 x 10 6 cells in a 1 :1 mix of growth media and matrigel (Trevigen, # 3433- 005-R1) and tumors were allowed to grow to a minimum diameter of 0.6cm prior to the initiation of treatment. Tumors that did not meet this criteria at the time of treatment initiation were not utilized for experimentation.

For AML studies, 10-12 weeks old male C57BL/6J mice were used for MLL- AF9 Ds-Red AML study (Approved protocol AC-AAAQ5405). For pre-treatment study with MLL-AF9 Ds-Red cells, keto and Keto/BKM group mice were given a ketogenic diet for 10 days prior to injection with MLL-AF9 Ds-Red cells (2xl0 5 per mouse in 200 ul) via lateral tail vein. The day after iv injection, the mice were given 0.5% carboxymethyl cellulose (CMC) as vehicle control or BKM120 (37.5 mg/kg) by oral gavage for two weeks (5 out of 7 days). The mice were euthanized after two- week treatment to check the bone marrow for AML progress. The tumor progress was also monitored via IVIS spectrum machine.

For co-current treatment studies with MLL-AF9 Ds-Red cells, the mice were injected with MLL-AF9 Ds-Red cells (2x10 5 per mouse in 200 ul) via lateral tail vein. The day after iv injection, the mice were given vehicle or BKM120 (37.5 mg/kg) by oral gavage for two weeks. The Keto or Keto/BKM group were changed to a ketogenic diet on the same day. The mice were euthanized after two-week treatment to check the bone marrow for AML progress.

To check if Keto/BKM treatment affects the AML engraftment, Keto and Keto/BKM group mice were given a ketogenic diet for 10 days, then treated with vehicle or BKM120 by oral gavage for two weeks. The mice were then injected with MLL-AF9 Ds-Red cells (2xl0 5 per mouse in 200 ul) vial lateral tail vein. Two weeks after the iv injection, the mice were euthanized to check the bone marrow for AML burden. The survival study, pre-treatment study, and co-current treatment were conducted at the same time. The mice were treated with vehicle or BKM120 (5 out of 7 days) until spontaneous death, or mice were euthanized when they appeared to be very sick (reduced spontaneous activity, unkempt coat, and dehydrated appearance), achieved body weight loss over 20%, or demonstrated signs of limb paralysis. To check if Keto/BKM treatment affects the bone marrow cell population, C57BL/6J mice were treated with 8 doses of vehicle or BKM120 in 9 days. The mice were euthanized, and one femur and tibia were removed from each mouse. The bone marrow cells were flushed with PBS (2% FBS). The red blood cells were lysed with ACK lysis buffer (Invitrogen).

Antibodies used for flow cytometry were as follows: CD34 (RAM34) from eBioscience, c-Kit (2B8), Sca-l (D7), CD3s (145-2C11), B220 (RA3-6B2), CD150 (TC-15-12F2.2), CD49b (DX5) and CD48 (HM48-1) from Biolegend. The‘lineage cocktail’ included CD3, CD4, Gr-l , Mac-l (CDl lb), B220, and Terr-l l9. DAPI was used to exclude dead cells.

Compounds

GDC-0032, MK2206, BEZ235, BKM-120, GDC-0941, GDC-0980, and Canagliflozin were all procured from medchem express (Monmouth Junction, NJ) and given via oral gavage in lOOul. Metformin was procured from Sigma Aldrich (St. Louis, MO). Bay- 80 6946 and OSI-906 came from Sellechem catalogue #S2802 and #Sl09l respectively. The targeting information for these compounds is displayed in Table 4. IC50 data was obtained from the Selleck Chem website (Selleckchem.com). The canagliflozin was administered 60 minutes before the PI3K pathway inhibitors so that its optimal efficacy lined up with the peak glucose levels. Mice treated with metformin were pretreated for 10 days prior to BKM120 treatment. Ketogenic diet was initiated at the time of initial PI3K inhibitor treatment unless otherwise stated. Doxycycline was procured from Sigma (St. Louis, Missouri) catalogue number D3072-1ML and administered via intraperitoneal injection once daily at a dose of 3mg/kg.

Table 4: Exemplary Pathway Inhibitors

Cell Lines

Murine pancreas cell lines were kindly gifted by Dr. Kenneth Olive, Columbia University. Murine breast lines were kindly gifted by Dr. Ramon Parsons, Mount Sinai School of Medicine. PDX Models were derived by the Englander Institute of Precision Medicine. Cell lines HEK293, HCC-38, MDA-MB-468, PC-3, BT-549 were purchased from ATCC and grown in DMEM supplemented with 10% FBS and 1 % Pen/Strep. HCT-116 and DLD-1 isogenic lines with and without PTEN deletion were kindly provided by the Laboratory of Todd Waldman. A chart of cells/organoids used is provided in Table 5, with known oncogenic alterations as described in publications cited above or as available from the ATCC (see website at

atcc.org/~/media/PDFs/Culture%20Guides/Cell_Lines_by_Gene _Mutation.ashx).

Table 5: Exemplary Cell Lines

Signaling Assays

For signaling assays, cells were washed lx in PBS and placed in starvation media (-FBS) for 6-18 hours depending upon cell line and treated 1 hour prior to harvesting with PI3K inhibitors as indicated alone or in combination with insulin 10 minutes prior to harvesting. Three-dimensional culture and dose response experiments of patient derived organoids were run as previously described. In brief, -1000 cells were plated in lOul of 1 :l matrigel to culture media in 96 well angiogenesis plates and allowed to solidify for 30 min at 37 degrees before 70ul of culture media was added. Organoids were then treated in triplicate in a log scale dose response and CellTiter- Glo assay (Promega) was run at 96 hours to determine the IC50 values. Proliferation assays in two-dimensional culture were performed as indicated in FIG. legends. Knockdown of insulin receptor was achieved using a doxycycline inducible shRNA strategy. For generation of miR-E shRNAs, 97-mer oligonucleotides were purchased (IDT Ultramers) coding for predicted shRNAs using an siRNA predictional tool Splash RNA (see website at splashrna.mskcc.org/).

Oligonucleotides were PCR amplified using the primers miRE-Xho-fw (5’- TGAACTCGAGAAGGTATATTGCTGTTGACAGTGAGCG-3’, SEQ ID NO:l) and miRE-Eco-rev (5’ -TGAACTCGAGAAGGTATATTGCTGTTGACAGT GAGCG-3’, SEQ ID NO:2). PCR products were purified and both PCR product and LT3GEPIR vectors (Fellmann, C. et al. An optimized microRNA backbone for effective single-copy RNAi. Cell Rep 5, 1704-1713) were double digested with EcoRI-FlF and XhoFlI. PCR product and vector backbone were ligated and transformed in Stbl3 competent cells and grown at 32° overnight. Colonies were screened using the primer miRE-fwd (5’- TGTTTGAATGAGGCTTCAGTAC-3’, SEQ ID NOG).

Renilla (SEQ ID NO:4):

TGCTGTTGACAGTGAGCGCAGGAATTATAATGCTTATCTATAGTGA

AGCC AC AGATGT AT AG AT A AGC ATT AT A ATT CCT ATGCCT ACTGCC

TCGGA

INSR4 (SEQ ID NOG):

TGCTGTTGACAGTGAGCGCGGGGTTCATGCTGTTCTACAATAGTGA

AGCCACAGATGTATTGTAGAACAGCATGAACCCCATGCCT ACTGCC

TCGGA

Immunoblotting

Cell lysates were prepared in lx CST Cell Lysis Buffer #9803, (Danvers MA). Total protein concentration was evaluated with the BCA kit (Pierce) 23227). The lysates were run out on 4-20% Tris-Glycine Gels (ThermoFisher, Carlsbad CA). Primary antibodies against pAKT473, pAKT308, pS6, pTYR, AKT, and S6 were procured from Cell Signaling (Danvers, MA), and were used overnight 1 :l000 in 5% bovine serum albumin. Actin and tubulin antibodies came from Sigma Aldrich and were used at 1 :5,000 in 5% Milk. All these antibodies were visualized with HRP conjugated secondary antibodies from Jackson Immuno at 1 :5000 in 5% milk.

Immunohistochemistry

Tumor sections (3 pm) were antigen retrieved with 10 mmol/L citrate acid, 0.05% Tween 20, pH6.0, and incubated with antibodies as indicated (Ki67 (Abeam, abl6667) 1 :500; cleaved caspase-3 (Aspl75; 5A1E; Cell Signaling Technology,

9664) 1 :200; phospho-INSR (Tyr 1162; Thermo Fisher #AHR027l) 1 TOO; phospho- AKT (Ser473; Cell Signaling Technology, 8101) 1 :20; and phospho-S6 ribosomal protein (Ser235/236; Cell Signaling Technology, 2211) 1 :300).

Blood Measurements

For assessment of blood glucose lOul of blood was taken from the tail of mice prior to treatment (time 0) and then again at the indicated time points (15, 30, 60, 90, 120,180 minutes) using a OneTouch Ultra Glucometer. At endpoint >l00ul of blood was drawn from the mice into EDTA tubes (Sarstedt #16.444). Blood was centrifuged (10,000 x g for 10 min at 4 °C), and plasma was stored at -20 °C. Plasma b- hydroxybutyrate, triglyceride (Stanbio Laboratory, Boerne, TX), Serum Insulin, and c-Peptide (APLCO Diagnostics, Salem, NH) levels were quantified by ELISA. FDG-PET

Male c57/bl6 mice (n = 4/arm) bearing orthotopic pancreatic adenocarcinoma allografts were injected with 200-250 pCi [ 89 Zr]liposomes (3-4 pmol lipid) in 200- 250 pL PBS solution into the tail vein. At the time of peak blood insulin feedback 90 minutes post BKM120 injection animals were anesthetized and scans were then performed using an Inveon PET/CT scanner (Siemens Healthcare Global). Whole body PET scans were performed recording a minimum of 50 million coincident events, with duration of 10 min. The energy and coincidence timing windows were 350-750 keV and 6 ns. The data was normalized to correct for non-uniformity of response of the PET, dead-time count losses, positron branching ratio, and physical decay to the time of injection. The counting rates in the reconstructed images were converted to activity concentrations (percentage injected dose [%ID] per gram of tissue) by use of a system calibration factor derived from the imaging of a phantom containing 89 Zr. Images were analyzed using ASIPro VMTM software (Concorde Micro-systems). Quantification of activity concentration was done by averaging the maximum values in at least 5 ranges of interest (ROIs) drawn on adjacent slices of the pancreatic tumors.

Metabolomics

Metabolites were extracted from cells or tissues using 80% methanol. Each sample was transferred to a pre-cooled (dry ice) 2 mL homogenization tube containing a single stainless-steel bead (5 mm). Pre-cooled 80% methanol (1 mL) was added to each sample and homogenization was performed using the Qiagen

TissueLyser II. Samples were then centrifuged at 4 °C for 15 minutes at 14,000 rpm. The supernatants were extracted and normalized based on tissue weight. Targeted LC/MS analyses were performed on a Q Exactive Orbitrap mass spectrometer (Thermo Scientific) coupled to a Vanquish UPLC system (Thermo Scientific). The Q Exactive operated in polarity- switching mode. A Sequant ZIC-HILIC column (2.1 mm i.d. x 150 mm, Merck) was used for separation of metabolites. Flow rate was 150 pL/min. Buffers consisted of 100% acetonitrile for A, and 0.1 % NH 4 OH/2O mM CH 3 COONH 4 in water for B. Gradient ran from 85% to 30% A in 20 min followed by a wash with 30% A and re-equilibration at 85% A. Metabolites were identified on the basis of exact mass within 5 ppm and standard retention times. Relative metabolite quantitation was performed based on peak area for each metabolite. All data analysis was done using scripts written in-house.

References:

1 Kandoth, C. etal. Mutational landscape and significance across 12 major cancer types. Nature 502, 333-339, doi:l0.l038/naturel2634 (2013).

2 Millis, S. Z., Ikeda, S., Reddy, S., Gatalica, Z. & Kurzrock, R. Landscape of Phosphatidylinositol-3-Kinase Pathway Alterations Across 19784 Diverse Solid Tumors. JAMA Oncol 2, 1565-1573, doi:l0.l00l/jamaoncol.20l6.089l (2016).

3 Massacesi, C. et al. PI3K inhibitors as new cancer therapeutics: implications for clinical trial design. Onco Targets Ther 9, 203-210, doi:l0.2l47/OTT.S89967 (2016). Mayer, I. A. et al. A Phase lb Study of Alpelisib (BYL719), a PBKalpha- Specific Inhibitor, with Letrozole in ER+/HER2- Metastatic Breast Cancer. Clin Cancer Res 23, 26-34, doi:l0.H58/l078-0432.CCR-l6-0l34 (2017).

Bendell, J. C. et al. Phase I, dose-escalation study of BKM120, an oral pan- Class I PI3K inhibitor, in patients with advanced solid tumors. J Clin Oncol 30, 282-290, doi:l0.l200/JC0.20l 1.36.1360 (2012).

Juric, D. et al Phase I Dose-Escalation Study of Taselisib, an Oral PI3K Inhibitor, in Patients with Advanced Solid Tumors. Cancer Discov 7, 704-715, doi:l0. H58/2l59-8290.CD-l6-l080 (2017).

Patnaik, A. et al. First-in-human phase I study of copanlisib (BAY 80-6946), an intravenous pan-class I phosphatidylinositol 3-kinase inhibitor, in patients with advanced solid tumors and non-Hodgkin's lymphomas. Ann Oncol 27, 1928- 1940, doi: 10.1093/annonc/mdw282 (2016).

Gallagher, E. J. et al. Inhibiting PI3K reduces mammary tumor growth and induces hyperglycemia in a mouse model of insulin resistance and hyperinsulinemia. Oncogene 31, 3213-3222, doi: 10. l038/onc.20l 1.495 (2012). Hopkins, B. D., Goncalves, M. D. & Cantley, L. C. Obesity and Cancer Mechanisms: Cancer Metabolism. J Clin Oncol 34, 4277-4283, doi:l0. l200/JC0.20l6.67.9712 (2016).

Fruman, D. A. et al. The PI3K Pathway in Human Disease. Cell 170, 605-635, doi:l0. l0l6/j.cell.20l7.07.029 (2017).

Belardi, V., Gallagher, E. J., Novosyadlyy, R. & LeRoith, D. Insulin and IGFs in obesity-related breast cancer. J Mammary Gland Biol Neoplasia 18, 277-289, doi: 10.1007 /s 10911 -013-9303 -7 (2013).

Gallagher, E. J. & LeRoith, D. Minireview: IGF, Insulin, and Cancer. Endocrinology 152, 2546-2551, doi:l0. l2l0/en.20l l-023l (2011).

Klil-Drori, A. J., Azoulay, L. & Poliak, M. N. Cancer, obesity, diabetes, and antidiabetic drugs: is the fog clearing? Nat Rev Clin Oncol 14, 85-99, doi: 10.1038/nrclinonc.2016.120 (2017).

Ma, J. et al. A prospective study of plasma C-peptide and colorectal cancer risk in men. J Natl Cancer Inst 96, 546-553 (2004). Xu, J. et al. Association between markers of glucose metabolism and risk of colorectal cancer. BMJ Open 6, e0H430, doi:l0.H36/bmjopen-20l6-0H430 (2016).

Ma, J. et al. Prediagnostic body-mass index, plasma C-peptide concentration, and prostate cancer- specific mortality in men with prostate cancer: a long-term survival analysis. Lancet Oncol 9, 1039-1047, doi:10.1016/S1470-

2045(08)70235-3 (2008).

Olive, K. P. et al Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 324, 1457-1461, doi:lO. H26/science.l 171362 (2009).

Pauli, C. et al. Personalized In Vitro and In Vivo Cancer Models to Guide Precision Medicine. Cancer Discov 7, 462-477, doi:10.1158/2159-8290.CD- 16-1154 (2017).

Komoroski, B. et al. Dapagliflozin, a novel, selective SGLT2 inhibitor, improved glycemic control over 2 weeks in patients with type 2 diabetes mellitus. Clin Pharmacol Ther 85, 513-519, doi:l0.l038/clpt.2008.250 (2009). Demin, O., Jr., Yakovleva, T., Kolobkov, D. & Demin, O. Analysis of the efficacy of SGLT2 inhibitors using semi-mechanistic model. Front Pharmacol 5, 218, doi:l0.3389/fphar.20l4.002l8 (2014).

Poliak, M. Metformin and other biguanides in oncology: advancing the research agenda. Cancer prevention research 3, 1060-1065, doi: 10.1158/1940- 6207.CAPR- 10-0175 (2010).

Poliak, M. Potential applications for biguanides in oncology. J Clin Invest 123, 3693-3700, doi:10.1172/JCI67232 (2013).

Saura, C. et al. Phase lb study of Buparlisib plus Trastuzumab in patients with HER2-positive advanced or metastatic breast cancer that has progressed on Trastuzumab-based therapy. Clin Cancer Res 20, 1935-1945, doi:10.1158/1078-0432.CCR- 13-1070 (2014).

Juvekar, A. et al. Combining a PI3K inhibitor with a PARP inhibitor provides cer. Cancer Discov 2, 1048-

25 Puchalska, P. & Crawford, P. A. Multi-dimensional Roles of Ketone Bodies in Fuel Metabolism, Signaling, and Therapeutics. Cell Metab 25, 262-284, doi:l0. l0l6/j.cmet.20l6.12.022 (2017).

26 Sampaio, L. P. Ketogenic diet for epilepsy treatment. Arq Neuropsiquiatr 74,

842-848, doi:l0.1590/0004-282X20160116 (2016).

27 Xia, J., Sinelnikov, I. V., Han, B. & Wishart, D. S. Metabo Analyst 3.0— making metabolomics more meaningful. Nucleic Acids Res 43, W251-257, doi:l0. l093/nar/gkv380 (2015).

28 Xia, J. & Wishart, D. S. Using MetaboAnalyst 3.0 for Comprehensive Metabolomics Data Analysis. Curr Protoc Bioinformatics 55, 14 10 11-14 10 91, doi : 10.1002/cpb i.11 (2016).

29 Xia, J. & Wishart, D. S. MetPA: a web-based metabolomics tool for pathway analysis and visualization. Bioinformatics 26, 2342-2344, doi: 10. l093/bioinformatics/btq4l 8 (2010).

30 Xia, J. & Wishart, D. S. Metabolomic data processing, analysis, and interpretation using MetaboAnalyst. Curr Protoc Bioinformatics Chapter 14, Unit 14 10, doi:l0.l002/047l250953.bil4l0s34 (2011).

31 Douris, N. et al. Adaptive changes in amino acid metabolism permit normal longevity in mice consuming a low-carbohydrate ketogenic diet. Biochim Biophys Acta 1852, 2056-2065, doi:l0.l0l6/j.bbadis.20l5.07.009 (2015).

32 Pauli, C. et al. An emerging role for cytopathology in precision oncology.

Cancer Cytopathol 124, 167-173, doi:l0.l002/cncy.2l647 (2016).

33 Lee, C., Kim, J. S. & Waldman, T. PTEN gene targeting reveals a radiation- induced size checkpoint in human cancer cells. Cancer Res 64, 6906-6914, doi: 10.1158/0008 -5472. CAN-04- 1767 (2004).

34 Pelossof, R. et al. Prediction of potent shRNAs with a sequential classification algorithm. Nat Biotechnol 35, 350-353, doi:l0. l038/nbt.3807 (2017).

35 Fellmann, C. et al. An optimized microRNA backbone for effective single-copy RNAi. Cell Rep 5, 1704-1713, doi:l0.l0l6/j.celrep.20l3.11.020 (2013).

All patents and publications referenced or mentioned herein are indicative of the levels of skill of those skilled in the art to which the invention pertains, and each such referenced patent or publication is hereby specifically incorporated by reference to the same extent as if it had been incorporated by reference in its entirety individually or set forth herein in its entirety. Applicants reserve the right to physically incorporate into this specification any and all materials and information from any such cited patents or publications.

The following statements are intended to describe and summarize various embodiments of the invention according to the foregoing description in the specification.

Statements:

1. A method of treating a disease or disorder associated with PI3K signaling, comprising administering to a subject in need thereof an effective amount of a modulator of glucose metabolism; and administering to the subject an effective amount of a pathway inhibitor of the msuhn-receptor/PBK/AKT/mTOR pathway.

2. The method of statement 1, wherein the modulator of glucose metabolism is a glucose-uptake inhibitor, optionally selected from the group consisting of a sodium- glucose-linked transport protein 1 (SGLT1) inhibitor, a sodium-glucose-linked transport protein 2 (SGLT2) inhibitor, or a dual SGLT1/SGLT2 inhibitor.

3. The method of statement 2, wherein the glucose-uptake inhibitor is selected from the group consisting of dapagliflozin, empagliflozin, canagliflozin, ipragliflozin, tofogliflozin, sergliflozin etabonate, remogliflozin etabonate, ertugliflozin, sotagliflozin, and conagliflozin.

4. The method of statement 1, wherein the modulator of glucose metabolism is metformin.

5. The method of statement 1, wherein the modulator of glucose metabolism is an insulin receptor/IGFl receptor inhibitor, wherein optionally the insulin receptor/IGFl receptor inhibitor is linsitinib (OSI-906). 6. The method of any of statements 1-5, wherein the pathway inhibitor is capable of inhibiting one or more kinases selected from the group consisting of INSR/IGFR,

PI3K, AKT, and mTOR.

7. The method of statement 6, wherein the pathway inhibitor is selected from the group consisting of idelalisib, copanlisib, buparlisib (BKM120), alpelisib (BYL719), taselisib (GDC-0032), pictilisib (GDC-0941), apitolisib (GDC-0980), serabelisib (TAK-117), dactolisib, apelisib, MK2206, and linsitinib (OSI-906).

8. The method of any of statements 1-7, wherein the disease or disorder is associated with PI3K signaling is a cancer or cell- proliferative disorder, a metabolic disorder, a neurodegenerative disease, or an inflammatory disease.

9. The method of any of statements 1-8, wherein the disease or disorder associated with PI3K signaling is a neurodegenerative disease, optionally brain trauma, spinal cord trauma, trauma to the peripheral nervous system, Alzheimer’s disease, Pick’s disease, diffuse Lewy body disease, progressive supranuclear palsy (Steel-Richardson syndrome), multisystem degeneration (Shy-Drager syndrome), motor neuron diseases including amyotrophic lateral sclerosis, degenerative ataxias, cortical basal degeneration, A LS- P rkinson’s- Dementi a complex of Guam, subacute sclerosing panencephalitis, Huntington’s disease, Parkinson’s disease,

synucleinopathies, primary progressive aphasia, striatonigral degeneration, Machado- Joseph disease/spinocerebellar ataxia type 3 and olivopontocerebellar degenerations, Gilles De La Tourette’s disease, bulbar and pseudobulbar palsy, spinal and spinobulbar muscular atrophy (Kennedy’s disease), primary lateral sclerosis, familial spastic paraplegia, Werdnig-Hoffman disease, Kugelberg-Welander disease, Tay- Sach’s disease, Sandhoff disease, familial spastic disease, Wohlfart-Kugelberg- Welander disease, spastic paraparesis, progressive multifocal leukoencephalopathy, and prion diseases (including Creutzfeldt- Jakob, Gerstmann-Straussler-Scheinker disease, Kuru and fatal familial insomnia, age-related dementia, vascular dementia, diffuse white matter disease (Binswanger’s disease), dementia of endocrine or metabolic origin, dementia of head trauma and diffuse brain damage, dementia pugilistica or frontal lobe dementia, neurodegenerative disorders resulting from cerebral ischemia or infraction including embolic occlusion and thrombotic occlusion as well as intracranial hemorrhage of any type, intracranial and intravertebral lesions, hereditary cerebral angiopathy, hereditary amyloid, Down’s syndrome,

macroglobulinemia, secondary familial Mediterranean fever, Muckle- Wells syndrome, multiple myeloma, pancreatic- related amyloidosis, cardiac-related amyloidosis, chronic hemodialysis arthropathy, Finnish amyloidosis, Iowa amyloidosis, or a combination thereof.

10. The method of any of statements 1-8, wherein the disease or disorder associated with PI3K signaling is an inflammatory disorder, optionally Type II diabetes, insulin resistance cardiovascular disease, arrhythmia, atherosclerosis, coronary artery disease, hypertriglyceridemia, dyslipidemia, retinopathy, nephropathy, neuropathy, obesity, and macular edema, ileitis, ulcerative colitis, Barrett’s syndrome, or Crohn’s disease.

11. The method of any of statements 1-8, wherein the disease or disorder associated with PI3K signaling is a metabolic disease, optionally Type II diabetes, insulin resistance cardiovascular disease, arrhythmia, atherosclerosis, coronary artery disease, hypertriglyceridemia, dyslipidemia, retinopathy, nephropathy, neuropathy, obesity, or macular edema.

12. The method of any of statements 1-10 or 11, wherein the subject consumes or is administered a ketogenic diet during treatment.

13. A method of treating a disease or disorder associated with PI3K signaling, comprising administering an effective amount of at least one pathway inhibitor of at least one kinase in the insulin-receptor/PI3K/AKT/mTOR pathway, wherein the subject consumes or is administered a ketogenic diet during treatment.

14. The method of any of the preceding statements, which disrupts systemic glucose homeostasis and improves efficacy of pathway-inhibitor treatment compared to pathway inhibitor alone. 15. Use of a modulator of glucose metabolism and/or an inhibitor of the insulin- receptor/PBK/AKT/mTOR pathway for treating a disease or disorder associated with PI3K signaling in a subject.

16. The use of statement 15 combined with use of a ketogenic diet by the subject. 17. A pharmaceutical composition comprising a modulator of glucose metabolism and a pathway inhibitor that inhibits at least one kinase in the insulin- receptor/PBK/AKT/mTOR pathway.

18. The pharmaceutical composition of statement 17, wherein the modulator of glucose metabolism is a glucose-uptake inhibitor, a sodium-glucose-linked transport protein 1 (SGLT1) inhibitor, a sodium- glucose-linked transport protein 2 (SGLT2) inhibitor, or a dual SGLT1/SGLT2 inhibitor.

19. The pharmaceutical composition of statement 17 or 18, wherein the glucose- uptake inhibitor is selected from the group consisting of dapagliflozin, empagliflozin, canagliflozin, ipragliflozin, tofogliflozin, sergliflozin etabonate, remogliflozin etabonate, ertugliflozin, sotagliflozin, and conagliflozin.

20. The pharmaceutical composition of statement 17, 18, or 19, wherein the modulator of glucose metabolism is metformin.

21. The pharmaceutical composition of statement 17-19 or 20, wherein the modulator of glucose metabolism is an insulin receptor/IGFl receptor inhibitor, wherein optionally the insulin receptor/IGFl receptor inhibitor is linsitinib (OSI-906).

22. The pharmaceutical composition of any of statement 17-20 or 21, wherein the pathway inhibitor is capable of inhibiting one or more kinases selected from the group consisting of INSR/IGFR, PI3K, ART, and mTOR. 23. The pharmaceutical composition of statement 17-21 or 22, wherein the pathway inhibitor is selected from the group consisting of idelalisib, copanlisib, buparlisib (BKM120), alpelisib (BYL719), taselisib (GDC-0032), pictilisib (GDC- 0941), apitolisib (GDC-0980), serabelisib (TAK-117), dactolisib, apelisib, MK2206, and linsitinib (OSI-906).

24. A method of inhibiting cell proliferation or a cell-proliferative disease, comprising administering to a subject in need thereof an effective amount of a glucose-uptake inhibitor; and administering to the subject an effective amount of a PI3K inhibitor.

25. The method of statement 24, wherein the glucose -uptake inhibitor is selected from the group consisting of dapagliflozin, empagliflozin, canagliflozin, ipragliflozin, tofogliflozin, sergliflozin etabonate, remogliflozin etabonate, ertugliflozin, sotagliflozin, and conagliflozin.

26. The method of statement 24 or 25, wherein the PI3K inhibitor is selected from the group consisting of idelalisib, copanlisib, buparlisib (BKM120), alpelisib (BYL719), taselisib (GDC-0032), pictilisib (GDC-0941), apitolisib (GDC-0980), serabelisib (TAK-117), dactolisib, MK2206, linsitinib (OSI-906), and apelisib.

27. The method of any of statements 24-25 or 26, wherein the inhibition of cell proliferation or cell-proliferative disease is enhanced compared to administration of the PI3K inhibitor without a glucose-uptake inhibitor.

The specific compositions and methods described herein are representative, exemplary and not intended as limitations on the scope of the invention. Other objects, aspects, and embodiments will occur to those skilled in the art upon consideration of this specification and are encompassed within the spirit of the invention as defined by the scope of the claims. It will be readily apparent to one skilled in the art that varying substitutions and modifications may be made to the invention disclosed herein without departing from the scope and spirit of the invention. The terms and expressions that have been employed are used as terms of description and not of limitation, and there is no intent in the use of such terms and expressions to exclude any equivalent of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the invention as claimed. Thus, it will be understood that although the present invention has been specifically disclosed by embodiments and optional features, modification and variation of the concepts herein disclosed may be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this invention as defined by the appended claims and statements of the invention.

The invention illustratively described herein may be practiced in the absence of any element or elements, or limitation or limitations, which is not specifically disclosed herein as essential. The methods and processes illustratively described herein may be practiced in differing orders of steps, and the methods and processes are not necessarily restricted to the orders of steps indicated herein or in the claims.

As used herein and in the appended claims, the singular forms“a,”“an,” and “the” include plural reference unless the context clearly dictates otherwise. Thus, for example, a reference to“a compound” or“a drug” or“an inhibitor” includes a plurality of such compounds, or drugs, or inhibitors, and so forth. In this document, the term“or” is used to refer to a nonexclusive or, such that“A or B” includes“A but not B,”“B but not A,” and“A and B,” unless otherwise indicated.

Under no circumstances may the patent be interpreted to be limited to the specific examples or embodiments or methods specifically disclosed herein. Under no circumstances may the patent be interpreted to be limited by any statement made by any Examiner or any other official or employee of the Patent and Trademark Office unless such statement is specifically and without qualification or reservation expressly adopted in a responsive writing by Applicants.

The invention has been described broadly and generically herein. Each of the narrower species and subgeneric groupings falling within the generic disclosure also form part of the invention. This includes the generic description of the invention with a proviso or negative limitation removing any subject matter from the genus, regardless of whether or not the excised material is specifically recited herein. In addition, where features or aspects of the invention are described in terms of Markush groups, those skilled in the art will recognize that the invention is also thereby described in ter s of any individual member or subgroup of members of the Markush group.

The Abstract is provided to comply with 37 C.F.R. § 1.72(b) to allow the reader to quickly ascertain the nature and gist of the technical disclosure. The Abstract is submitted with the understanding that it will not be used to interpret or limit the scope or meaning of the claims.