Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ANTI-METHANOGENIC LOVASTATIN ANALOGS OR DERIVATIVES AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2016/161085
Kind Code:
A1
Abstract:
The present invention relates to, in part, methods for the treatment of methanogen-associated disorders such as, for example, Irritable Bowel Syndrome (IBS) using at least one anti-methanogenic lovastatin analog or derivative. In addition, modified-release formulations comprising at least one anti-methanogenic lovastatin analog or derivative are provided which release the anti-methanogenic lovastatin analog or derivative in the gastrointestinal tract.

Inventors:
PIMENTEL MARK (US)
WACHER VINCE (US)
Application Number:
PCT/US2016/025214
Publication Date:
October 06, 2016
Filing Date:
March 31, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CEDARS SINAI MEDICAL CENTER (US)
SYNTHETIC BIOLOGICS INC (US)
International Classes:
A61K31/351; A61K31/366; A61P1/04
Domestic Patent References:
WO2005058861A12005-06-30
WO2010088633A22010-08-05
WO2014152754A22014-09-25
WO2010004273A12010-01-14
Foreign References:
US20100048595A12010-02-25
US6201014B12001-03-13
US4231938A1980-11-04
US4294846A1981-10-13
US4450171A1984-05-22
US4444784A1984-04-24
US4661483A1987-04-28
US4864038A1989-09-05
US4937264A1990-06-26
US4847306A1989-07-11
US5049696A1991-09-17
US4766145A1988-08-23
US4876279A1989-10-24
US4876366A1989-10-24
US4937263A1990-06-26
US4771071A1988-09-13
US4916162A1990-04-10
US4940727A1990-07-10
US4841074A1989-06-20
US4894465A1990-01-16
US4894466A1990-01-16
US4855456A1989-08-08
USRE36520E2000-01-18
USRE36481E2000-01-04
US5116870A1992-05-26
US9235487A1987-09-02
US4866090A1989-09-12
US4857547A1989-08-15
US4857546A1989-08-15
US4921974A1990-05-01
US4963538A1990-10-16
US4738982A1988-04-19
US4782084A1988-11-01
US4885314A1989-12-05
US4654363A1987-03-31
US4198425A1980-04-15
US4255444A1981-03-10
US4604472A1986-08-05
EP0024348A11981-03-04
US6818638B22004-11-16
US5284858A1994-02-08
US6414016B12002-07-02
US6583174B12003-06-24
US7304036B22007-12-04
US6344566B12002-02-05
US5602179A1997-02-11
US20130116334A12013-05-09
US8110177B22012-02-07
US6274320B12001-08-14
US6258568B12001-07-10
US6210891B12001-04-03
Other References:
GHOSHAL, UC ET AL.: "Irritable bowel syndrome and small intestinal bacterial overgrowth: Meaningful association or unnecessary hype.", WORLD JOURNAL OF GASTROENTEROLOGY, vol. 20, no. 10, 14 March 2014 (2014-03-14), pages 2482 - 2491, XP055317104
FERNANDES ET AL.: "Possible association between statin use and bowel dysmotility", BMJ CASE REPORTS, 2012
MOHAMMAD FASELEH JAHROMI ET AL., BIOMED RES. INT., 1 January 2013 (2013-01-01), pages 11 - 10
MILLER ET AL., J. DAIRY SCI., vol. 84, no. 6, 2001, pages 1445 - 1448
FRANCIS ET AL., ALIMENT PHARMACOL THER, vol. 11, 1997, pages 395 - 402
ASAKURA, CLINICAL DIGESTIVE INTERNAL MEDICINE, vol. 8, no. 8, 1993, pages 1373 - 1381
GASTROENTEROLOGY, vol. 87, pages 1 - 7
WONGDROSSMAN, EXPERT REV. GASTROENTEROL. HEPATOL., vol. 4, no. 3, 2010
G. R. SWARTJ. W. VAN DEN BERG: "C breath test in gastrointestinal practice", SCAND. J. GASTROENTEROL., vol. 225, 1998, pages 13 - 18
S. F. DELLERT ET AL.: "The C-xylose breath test for the diagnosis of small bowel bacterial overgrowth in children", J. PEDIATR. GASTROENTEROL. NUTR., vol. 25, no. 2, 1997, pages 153 - 58
C. E. KINGP. P. TOSKES: "Breath tests in the diagnosis of small intestinal bacterial overgrowth", CRIT. REV. LAB. SCI., vol. 21, no. 3, 1984, pages 269 - 81
C. S. CHANG ET AL.: "Increased accuracy of the carbon-14 D-xylose breath test in detecting small-intestinal bacterial overgrowth by correction with the gastric emptying rate", EUR. J. NUCL. MED., vol. 22, no. 10, 1995, pages 1118 - 22
C. E. KINGP. P. TOSKES: "Comparison of the 1-gram [14C]xylose, 10-gram lactulose-H , and 80-gram glucose-H breath tests in patients with small intestine bacterial overgrowth", GASTROENTEROL, vol. 91, no. 6, 1986, pages 1447 - 51, XP009140180
A. SCHNEIDER ET AL.: "Value of the C -D-xylose breath test in patients with intestinal bacterial overgrowth", DIGESTION, vol. 32, no. 2, 1985, pages 86 - 91, XP000981761, DOI: 10.1159/000199223
INNIS ET AL.: "A Guide to Methods and Applications", 1990, ACADEMIC PRESS, INC., article "PCR Protocols"
GINZONGER ET AL., CANCER RESEARCH, vol. 60, 2000, pages 5405 - 5409
WUWALLACE, GENOMICS, vol. 4, 1989, pages 560
LANDEGREN ET AL., SCIENCE, vol. 241, 1988, pages 1077
BARRINGER ET AL., GENE, vol. 89, 1990, pages 117
KWOH ET AL., PROC. NATL. ACAD. SCI. USA, vol. 86, 1989, pages 1173
GUATELLI ET AL., PROC. NAT. ACAD. SCI. USA, vol. 87, 1990, pages 1874
BENNETT ET AL., PHARMACOGENOMICS, vol. 6, no. 373-20, 2005, pages 382
MARGULIES ET AL., NATURE, vol. 437, 2005, pages 376 - 380
See also references of EP 3277274A4
Attorney, Agent or Firm:
HUBER, Linda, B. et al. (300 South Grand Avenue Suite 410, Los Angeles California, US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A method of inhibiting or reducing methanogenesis, comprising administering an effective amount of an anti-methanogenic lovastatin analog or derivative to a subject in need thereof. 2. The method of claim 1, wherein the subject suffers from irritable bowel syndrome. 3. The method of any one of claims 1 or 2, wherein the subject suffers from constipation-associated IBS (IBS-C) and/or chronic idiopathic constipation (CIC). 4. The method of any one of claims 1-3, wherein the subject suffers from obesity. 5. The method of any one of claims 1-4, wherein the subject suffers from diabetes. 6. A method of reducing or eliminating enteric methane production, comprising administering an effective amount of an anti-methanogenic lovastatin analog or derivative to a subject in need thereof. 7. A method of treating constipation, comprising administering an effective amount of an anti- methanogenic lovastatin analog or derivative to a subject in need thereof. 8. The method of any one of claims 1-7, wherein the anti-methanogenic lovastatin is a compound of one or more of the Formulae Ia-e, IIa-e, IIIa-e, IVa-e, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XVI, XVII, XVIII, XVIX, XXX, XXXI, XXXII, XXXIII, XXXIV, XXXV and XXXVI, as described herein. 9. The method of claim 1, wherein the anti-methanogenic lovastatin analog is lovastatin diol lactone. 10. The method of claim 6, wherein the anti-methanogenic lovastatin analog is lovastatin diol lactone. 11. The method of claim 7, wherein the anti-methanogenic lovastatin analog is lovastatin diol lactone. 12. The method of any one of claims 1-8, wherein the anti-methanogenic lovastatin analog or derivative is predominantly in the lactone form. 13. The method of any one of claims 1-12, wherein the anti-methanogenic lovastatin analog or derivative comprises one or more modifications relative to lovastatin in the lactone ring. 14. The method of any one of claims 1-13, wherein the anti-methanogenic lovastatin analog or derivative comprises one or more modifications relative to lovastatin that cause the anti-methanogenic lovastatin analog or derivative to be predominantly in the in the lactone form.

15. The method of any one of claims 1-14, wherein the anti-methanogenic lovastatin analog or derivative is predominantly in the lactone form in the subject’s GI tract. 16. The method of any one of claims 1-15, wherein the anti-methanogenic lovastatin analog or derivative is not an effective inhibitor of HMG-CoA reductase. 17. A modified-release formulation comprising at least one anti-methanogenic lovastatin analog or derivative, wherein the formulation releases at least 60% of the anti-methanogenic lovastatin analog or derivative after the stomach and into one or more regions of the intestinal tract. 18. The modified-release formulation of claim 17, wherein the anti-methanogenic lovastatin is a compound of one or more of the Formulae Ia-e, IIa-e, IIIa-e, IVa-e, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XVI, XVII, XVIII, XVIX, XXX, XXXI, XXXII, XXXIII, XXXIV, XXXV and XXXVI, as described herein. 19. The modified-release formulation of claim 17, wherein the anti-methanogenic lovastatin is lovastatin diol lactone. 20. The modified-release formulation of any one of claims 17 or 18, wherein the anti-methanogenic lovastatin analog or derivative is predominantly in the lactone form. 21. The modified-release formulation of any one of claims 17-20, wherein the anti-methanogenic lovastatin analog or derivative comprises one or more modifications relative to lovastatin in the lactone ring. 22. The modified-release formulation of any one of claims 17-21, wherein the anti-methanogenic lovastatin analog or derivative comprises one or more modifications relative to lovastatin that cause the anti-methanogenic lovastatin analog or derivative to be predominantly in the in the lactone form. 23. The formulation of any one of claims 17-22, wherein the anti-methanogenic lovastatin analog or derivative is not an effective inhibitor of HMG-CoA reductase. 24. The formulation of any of claims 17-23, wherein the anti-methanogenic lovastatin analog or derivative is released in the small intestine. 25. The formulation any of claims 17-24, wherein the anti-methanogenic lovastatin analog or derivative is released in the duodenum and/or jejunum. 26. The formulation any of claims 17-25, wherein the anti-methanogenic lovastatin analog or derivative is released in the ileum and/or at the ileocecal junction.

27. The formulation any of claims 17-26, wherein the anti-methanogenic lovastatin analog or derivative is released in the large intestine. 28. The formulation any of claims 17-27, wherein the anti-methanogenic lovastatin analog or derivative is released one or more of the cecum, ascending, transverse, descending or sigmoid portions of the colon, and rectum. 29. The formulation of any one of claims 17-28, wherein the formulation is administered orally. 30. The formulation any of claims 17-29, wherein the formulation is in the form of a capsule or a tablet. 31. The formulation of any one of claims 17-30, wherein the formulation comprises a modified-release coating that is substantially stable in gastric fluid. 32. The formulation of any one of claims 17-31, wherein the formulation comprises a modified-release coating that is degraded by a microbial enzyme present in the gut flora. 33. The formulation of any one of claims 17-32, wherein the formulation comprises a modified-release coating having a solubility that is pH-dependent. 34. The formulation of any one of claims 17-33, wherein the formulation comprises a modified-release coating having a time-dependent erosion profile. 35. The formulation of any one of claims 17-34, comprising: a first dose of lovastatin or at least one anti-methanogenic lovastatin analog or derivative; and a second dose of at least one anti-methanogenic lovastatin analog or derivative; and wherein the first dose and the second dose are released at different times and/or at different pHs. 36. The formulation of claim 35, wherein the first and/or second dose is encapsulated in a core particle. 37. The formulation of any one of claim 35 or 36, wherein a modified-release coating is disposed over the core particle to form a modified-release particle. 38. The formulation of any one of claims 35-37, wherein the formulation comprises a plurality of modified- release particles. 39. The formulation of any one of claims 35-38, wherein the formulation is in the form of a capsule. 40. The formulation of any one of claims 35-39, wherein the first and/or second dose are encapsulated in a layer.

41. The formulation of any one of claims 35-40, wherein a modified-release coating is disposed over the layer to form a modified-release layer. 42. The formulation of any one of claims 35-41, wherein the formulation comprises a plurality of layers. 43. The formulation of any one of claims 35-42, wherein the formulation is in the form of a multi-layered tablet. 44. The formulation of any one of claims 35-43, wherein the first dose is released at the duodenum, and the second dose is released at the ileum. 45. The formulation of any one of claims 35-44, wherein the first is released at the small intestine, and the second dose is released at the large intestine. 46. The formulation of any one of claims 35-45, wherein the modified-release coating is substantially stable in gastric fluid. 47. The formulation of any one of claims 35-46, wherein the modified-release coating is degraded by a microbial enzyme present in the gut flora. 48. The formulation of any one of claims 35-47, wherein the modified-release coating has a solubility that is pH-dependent. 49. The formulation of any one of claims 35-48, wherein the modified-release coating has a time-dependent erosion profile. 50. The formulation of any one of claims 35-49, further comprising a pharmaceutically acceptable excipient. 51. The formulation of any one of claims 35-50, further comprising an agent which prevents or reduces lactone ring-opening. 52. The formulation of claim 51, wherein the agent which prevents or reduces lactone ring-opening is an esterase inhibitor and/or a paraoxonase inhibitor. 53. The formulation of claim 52, wherein the esterase inhibitor and/or a paraoxonase inhibitor is grapefruit and/or strawberry extract. 54. The formulation of any one of claims 35-53, further comprising an additional therapeutic agent. 55. The formulation of claim 54, wherein the additional therapeutic agent is a prokinetic agent.

56. A method of inhibiting or reducing methanogenesis, comprising administering an effective amount of a formulation of any one of claims 17-55 to a subject in need thereof. 57. The method of claim 56, wherein the subject suffers from irritable bowel syndrome. 58. The method of any one of claims 56 or 57, wherein the subject suffers from constipation-associated IBS (IBS-C) and/or chronic idiopathic constipation (CIC). 59. The method of any one of claims 56-58, wherein the subject suffers from obesity. 60. The method of any one of claims 56-59, wherein the subject suffers from diabetes. 61. A method of treating constipation, comprising administering an effective amount of a formulation of any one of claims 17-55 to a subject in need thereof. 62. A method of reducing or eliminating enteric methane production, comprising administering an effective amount of a formulation of any one of claims 17-55 to a subject in need thereof. 63. The method of any one of claims 56-62, wherein the subject is human. 64. A method of treating IBS-C and/or CIC, comprising administering an effective amount of the formulation of any one of claims 17-55 to a subject in need thereof.

65. A method of treating IBS-C and/or CIC, comprising administering an effective amount of a compound of one or more of the Formulae Ia-e, IIa-e, IIIa-e, IVa-e, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XVI, XVII, XVIII, XVIX, XXX, XXXI, XXXII, XXXIII, XXXIV, XXXV and XXXVI as described herein to a subject in need thereof.

66. A method of treating IBS-C and/or CIC, comprising administering an effective amount of is lovastatin diol lactone to a subject in need thereof

67. A method of making an agent effective for the treatment of constipation-associated IBS (IBS-C) and/or chronic idiopathic constipation (CIC), comprising:

(a) identifying the agent by:

(i) measuring a subjects breath levels of methane;

(ii) administering an effective amount of a test compound to the subject;

(ii) measuring the subjects breath levels of methane;

(iii) comparing the subjects breath levels of methane with and without the test compound; and (iv) selecting the test compound as a candidate compound if the result of the comparison of step (iii) indicates that the test compound is useful for the treatment of IBS-C and/or CIC and (b) formulating the identified agent for administration to the GI tract.

68. A method of claim 67, wherein the test compound is one or more of a compound of Formulae Ia-e, IIa- e, IIIa-e, IVa-e, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XVI, XVII, XVIII, XVIX, XXX, XXXI, XXXII, XXXIII, XXXIV, XXXV and XXXVI, as described herein to a subject in need thereof.

Description:
ANTI-METHANOGENIC LOVASTATIN ANALOGS OR DERIVATIVES AND USES THEREOF

PRIORITY

[001] This application claims the benefit of and priority to U.S. Provisional Patent Application No. 62/141,413, filed April 1, 2015, the entire contents of all of which are incorporated by reference herein.

FIELD OF THE INVENTION

[002] The present invention relates to, in part, methods and compositions for the treatment of methanogen-associated disorders such as, for example, Irritable Bowel Syndrome (IBS) using anti-methanogenic lovastatin analogs and derivatives thereof.

BACKGROUND

[003] The human microbiome is proving to be a vital component of both human health and disease. For example, the microbiome of the gastrointestinal (GI) tract underlies central processes of nutrient capture and metabolism; however, disruption of this microbiome is believed to be causative of a number of disorders that may severely reduce the quality of an afflicted subject’s life, or worse.

[004] Irritable Bowel Syndrome (IBS) affects an estimated 30 million people in the United States alone. IBS is a functional GI disorder that results in abdominal pain and/or discomfort, along with changes in bowel habits. It is often classified into four subtypes: constipation-associated IBS (IBS-C); diarrhea-associated IBS (IBS-D); mixed (or alternating) IBS (IBS-M or IBS-A); and unsubtyped (or unspecified) IBS (IBS-U).

[005] Recent studies suggest a link between intestinal methane (CH 4 ) production and constipation in IBS- C as well as chronic idiopathic constipation (CIC). Methane (CH 4 ) production in humans, which is due to methanogenic archaea in the intestine (including Methanobrevibacter smithii (M. smithii)), likely reflects a microbiome disruption.

[006] Antibiotic therapy may not be a feasible treatment for this affliction, as methanogens such as M. smithii are highly resistant to most classes of antibiotics and the likely chronic administration of antibiotics comes with a number of therapeutic risks, including further microbiome disruption.

[007] Recent studies have suggested that certain statin drugs, including lovastatin, are promising candidates for the treatment of disorders such as IBS-C and CIC. Accordingly, the field would benefit from further related agents that may be able to provide treatment effects with benefits such as reduced doses and/or more extensive benefits. SUMMARY OF THE INVENTION

[008] Accordingly, the present invention provides, inter alia, improved compositions, formulations and methods for the treatment of various methanogen-associated disorders. In one aspect, the present invention provides for methods of inhibiting or reducing methanogenesis and/or methane accumulation by administering an anti-methanogenic lovastatin analog or derivative to a subject in need thereof. In some embodiments, the subject suffers from IBS, such as IBS-C. In other embodiments, the subject suffers from obesity. In yet another embodiment, the subject suffers from diabetes. In various embodiments, the present invention provides for methods of treating or preventing a methanogen-associated disorder optionally selected from one or more of IBS, such as IBS-C, diabetes, and obesity by administering an anti-methanogenic lovastatin analog or derivative to a subject in need thereof. In an embodiment, methods are provided for treating constipation. In another embodiment, methods are provided for reducing or eliminating enteric methane production. In various embodiments, the anti-methanogenic lovastatin analog or derivative is a compounds described herein, such as lovastatin diol lactone.

[009] In another aspect, the present invention provides, in part, modified-release formulations comprising anti-methanogenic lovastatin analogs or derivatives. In some embodiments, the modified-release formulations release at least 60% of the anti-methanogenic lovastatin analog or derivative, after the stomach and into one or more regions of the intestinal tract. In certain embodiments, the formulation releases the anti-methanogenic lovastatin analog or derivative in the small intestine, including one or more of the duodenum, jejunum, and ileum. In other embodiments, the formulation releases the anti-methanogenic lovastatin analog or derivative in the large intestine (e.g., one or more of the cecum, ascending, transverse, descending or sigmoid portions of the colon, and rectum).

[010] In various embodiments, the anti-methanogenic lovastatin analog or derivative is in either the lactone or ß-hydroxyacid form. In some embodiments, the anti-methanogenic lovastatin analog or derivative is in the lactone form. In some embodiments, the anti-methanogenic lovastatin analog or derivative is lovastatin diol lactone.

[011] In various embodiments, the modified-release formulation is administered orally to a subject in need thereof. In one embodiment, the formulation may be in the form of a capsule or a tablet. In an embodiment, the formulation comprises a modified-release coating that is substantially stable in gastric fluid. In another embodiment, the modified-release coating may be degraded by a microbial enzyme present in the gut flora. In a further embodiment, the modified-release coating may have a solubility and/or stability that is pH dependent. In other embodiments, the modified-release coating may have a time-dependent erosion profile.

[012] In various embodiments, the modified-release formulation comprises a first dose of lovastatin or at least one anti-methanogenic lovastatin analog or derivative and a second dose of at least one anti-methanogenic lovastatin analog or derivative (e.g. the first and second doses may be the same or different anti-methanogenic lovastatin analog or derivative at a given dose, or the first and second doses may be the same anti- methanogenic lovastatin analog or derivative at the same or different doses, or the first or second doses may comprise lovastatin). In various embodiments, the first dose and the second dose are released at different times and/or at different pHs and in different regions of the gastrointestinal tract. In some embodiments, the first and/or second dose of at least one anti-methanogenic lovastatin analog or derivative is encapsulated in a core particle. A modified-release coating may be disposed over the core particle to form a modified-release particle. In certain embodiments, the formulation comprises a plurality of modified-release particles. In an illustrative embodiment, the formulation maybe in the form of a capsule. In another embodiment, the first and second dose of at least one anti-methanogenic lovastatin analog or derivative is encapsulated in a layer. A modified-release coating may be disposed over the layer to form a modified-release layer. In certain embodiments, the formulation comprises a plurality of modified-release layers. In an illustrative embodiment, the formulation maybe in the form of a multilayer tablet.

[013] In some embodiments, the first dose and second dose of anti-methanogenic lovastatin analog or derivative are released at different times and or at different pHs. In illustrative embodiments, the first dose may release the anti-methanogenic lovastatin analog or derivative at the duodenum while the second dose may release the anti-methanogenic lovastatin analog or derivative at the ileum. In other embodiments, the first dose may release the anti-methanogenic lovastatin analog or derivative at the small intestine while the second dose may release the anti-methanogenic lovastatin analog or derivative at the large intestine.

[014] The formulations of the present invention may further include a pharmaceutically acceptable excipient. In some embodiments, the formulation may further include an agent which prevents or reduces lactone ring-opening, such as an esterase inhibitor (e.g. grapefruit juice; , including flavonoid components such as, for example, naringenin, kaempferol, morin, galangin, and quercetin; flavoring ester mixtures in, for example, strawberry juice (e.g. phenyl benzoate, propyl paraben, phenethyl isobutyrate, bacampicillin, talampicillin, p-tolyl benzoate, ethyl paraben, diethyl phthalate, octyl acetate, and pivampicillin) and/or a paraoxonase inhibitor (e.g. PON1 or PON3 inhibitor). In some embodiments, the formulation may further include an inhibitor of the organic anion transporting polypeptide (OATP) transporter, such as one or more of green tea extract, epicatechin gallate (ECG) and epigallocatechin gallate (EGCG). In some embodiments, the OATP inhibitor is released prior to release of the lovastatin analog or derivative. The formulations of the present invention may also further include an additional therapeutic agent such as, by way of non-limiting example, a prokinetic agent.

[015] In various embodiments, the methods and formulations described herein eradicate or reduce methane production, which is causative of, or correlative with, various methanogen-associated disorders, including, for example, IBS (e.g. IBS-C), diabetes and obesity. In various embodiments, the methods and formulations described herein target the gastrointestinal (GI) tract and therefore provide for specific delivery to a site of methanogen colonization and/or methane production and/or accumulation while avoiding or reducing systemic exposure to anti-methanogenic lovastatin analogs or derivatives and minimizing their systemic effects. As such, the present invention provides for effective treatments that avoid side effects associated with chronic systemic administration (e.g. muscle pain, abnormalities in liver enzyme tests, etc.). Further, in some embodiments, the present invention surprisingly treats bowel-disorders despite reports linking statin use to, for example, constipation (see, e.g., Fernandes et al. Possible association between statin use and bowel dysmotility. BMJ Case Reports 2012; 10.1136/bcr.10.2011.4918 and Merck Global Medical Information. Professional Information Response UK11-010274, the contents of which are hereby incorporated by reference in their entireties). Further, in some embodiments, the present invention surprisingly treats diabetes despite reports linking statin use to this disorder (see, e.g., Circ Cardiovasc Qual Outcomes 6 (4): 390–9, the contents of which are hereby incorporated by reference in their entirety).

[016] In an embodiment, the present invention provides for methods of inhibiting or reducing methanogenesis and/or methane accumulation by administering formulations disclosed herein to a subject in need thereof. In various embodiments, the present invention provides for methods of treating or preventing a methanogen-associated disorder optionally selected from one or more of IBS, such as IBS-C, diabetes, and obesity by administering a formulations described herein to a subject in need thereof. In an embodiment, methods are provided for treating constipation using the formulations described herein. In another embodiment, methods are provided for reducing or eliminating enteric methane production using the formulations described herein.

DESCRIPTION OF THE FIGURES

[017] Figures 1A-1B depict some embodiments of a modified-release formulation in the form of encapsulated beads which releases a first dose of anti-methanogenic lovastatin analog or derivative at the duodenum and a second dose of anti-methanogenic lovastatin analog or derivative at the ileum.

[018] Figure 2 depicts embodiments of modified-release formulations as multi-layer capsules or tablets for delivery of anti-methanogenic lovastatin analogs or derivatives to the intestines (an illustrative commercial material is shown, related materials are known in the art).

[019] Figures 3A-3B depict embodiments of modified-release formulations for colonic delivery of anti- methanogenic lovastatin analogs or derivatives (an illustrative commercial material is shown, related materials are known in the art).

[020] Figure 4 shows illustrative methane versus time profiles for human stool samples treated with the different lovastatin species.

[021] Figure 5 depicts various embodiments of modified-release formulations in the form of capsules that delivers either one or two doses of anti-methanogenic lovastatin analogs or derivatives to the intestines.

[022] Figure 6 depicts a clinical study design with the present compositions. DETAILED DESCRIPTION OF THE INVENTION

[023] The present invention is based, in part, of the surprising discovery of methods and formulations that are useful in effectively treating or preventing methanogen-associated disorders while avoiding side effects. The present invention provides, inter alia, methods of treating or preventing methanogen-associated disorders using one or more anti-methanogenic lovastatin analogs or derivatives, including, without limitation, lovastatin diol lactone. The present invention additionally provides, inter alia, modified-release formulations comprising one or more anti-methanogenic lovastatin analogs or derivatives, including, without limitation, lovastatin diol lactone, which are useful in, for example, the treatment of methanogen-associated disorders such as, for example, IBS (including, for example, IBS-C).

[024] As used herein,“lovastatin analogs or derivatives” refer to a class of compounds including pharmaceutically acceptable analogs, derivatives, esters, prodrugs, salts, solvates, enatiomers, stereoisomers, active metabolites, co-crystals, and other physiologically functional derivatives of lovastatin. Such lovastatin analogs or derivatives may be used as lipid lowering agents. However, the prior use of the lovastatin analogs or derivatives does not necessarily imply a mechanism of action in the treatment of methanogenesis. That is, in some embodiments, the lovastatin analog or derivative may inhibit the enzyme HMG-CoA reductase while in others it may have another manner of causing an effect. For example, the lovastatin analog or derivative may target a methanogenic enzyme, such as, for example, one or more of adh alcohol dehydrogenase; fdh formate dehydrogenase; fno F420-dependent NADP oxidoreductase; ftr formyl-MF:H4MPT formyltransferase; fwd formyl- MF dehydrogenase; hmd methylene-H4MPT dehydrogenase; mch methenyl-H4MPT cyclohydrolase; mtd F420- dependent methylene-H4MPT dehydrogenase; mer F420-dependent methylene-H4MPT reductase; mtr methyl- H4MPT:CoM-methyltransferase; mcr methyl-CoM reductase; and the mtaB methanol:cobalamin methyltransferase heterodisulfide reductase system. In some embodiments, the lovastatin analog or derivative does not substantially inhibit the enzyme HMG-CoA reductase. In some embodiments, the lovastatin analog or derivative is present in the lactone form, for example, as the active form (e.g. in a subject’s GI tract). In some embodiments, the lovastatin analog or derivative is lovastatin diol lactone.

[025] Illustrative“lovastatin analogs or derivatives” are compounds of Formulae Ia-e, IIa-e, IIIa-e, IVa-e, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XVI, XVII, XVIII, XVIX, XXX, XXXI, XXXII, XXXIII, XXXIV, XXXV and XXXVI, as described herein. In some embodiments, the lovastatin analog or derivative is lovastatin diol lactone.

[026] Systemic statin usage has been associated with adverse side effects such as elevation in hepatic enzyme levels and muscle problems (e.g., myalgias, rhabdomyolysis, and severe myopathy). Further, systemic statin usage has been linked to digestive disorders in some patients. The present invention also provides modified release formulations comprising anti-methanogenic lovastatin analogs or derivatives that minimize absorption of the administered lovastatin analogs or derivatives from the intestine into the systemic circulation and reduce potential side effects or disease exacerbating effects. Additionally, not all patients with IBS-C or CIC will requ ire lipid lowe ring therapy, so systemic absorption of the anti-m ethanogenic lovastatin a nalogs or derivative s from the m odified releas e formulations of the prese nt invention w ill ideally be insufficient to provide a clinically- meaningful re duction in to tal cholestero l (total-C), o r low-density lipoprotein c holesterol (L DL-C), or apolipopr otein B (Apo B), or triglyc erides (TG), or a clinicall y-meaningful increase in h igh-density l ipoprotein cholester ol (HDL-C) (fo r example, a reduction of le ss than 5% i n serum LDL- C levels at 6 w eeks).

Anti-Meth anogenic Lov astatin Analo gs or Derivati ves

[027] The pres ent invention contemplate s the use of lovastatin an alogs or deriv atives, which includes pharmace utically acce ptable analo gs, esters, prodrugs, sa lts, solvates, enatiomers, stereoisome rs, active metabolit es, co-crystal s, and other physiologically functional d erivatives of l ovastatin. Lov astatin, also known as mevinolin , was describ ed, for examp le, in U.S. Pa tent Applicati on No.4,231, 938, the entir e contents of which are incorpora ted by refer ence herein. Various anti -methanogen ic lovastatin analogs or d erivatives th ereof are contempl ated. In vario us embodime nts, any of th ese anti-meth anogenic lov astatin analog s or derivativ es are in the lacto ne form (e.g . substantial ly in the lact one for, or in an equilib rium in whic h the lactone form is predomin ant of the bet a-hydroxy for m), where app licable.

[028] In variou s embodime nts, the an ti-methanoge nic lovastatin analog or derivative c omprises polyhydro naphthyl mo ieties and v arious 8-acyl oxy groups attached the reto. In an e mbodiment, the anti- methanog enic lovasta tin analog o r derivative may be a g roup of 6(R) -[2-(8'-acyloxy -2'-methyl-6'- methyl(or hydrogen )-polyhydrona phthyl-1')-eth yl]-4(R)-hydro xy-3,4,5,6-te trahydro-2H-p yran-2-ones or the hydr oxy acid form of s aid pyranones , the pharma ceutically acc eptable salts of said hydro xy acids and to the lower a lkyl (e.g., C 1-4 alkyl ester) and ph enyl, dimethyl amino or ace tylamino subs tituted lower a lkyl esters of the lactone o r hydroxy acid form . In some em bodiments, t he anti-metha nogenic lova statin analog or derivative may be a dih ydro and tetrahydr o analog of lo vastatin. Suc h analogs an d derivatives are describe d, for examp le, in U.S. Pa tent Nos. 4,294,846 , 4,450,171, and 4,444,784, the enti re contents of which are incorporated by referenc e herein (including , by way of illustration, di sclosures of specific comp ounds and s ynthetic sche mes). For ex ample, in some em bodiments, th e anti-methan ogenic lovast atin analog o r derivative m ay have the fo rmula:

or pharm aceutically ac ceptible salts thereof, whe rein R‘ repre sents H or C H 3 ; X, Y, and Z represen t possible double b onds; the se ries a-e are defined in the table above and, where applicable, R represents: (1) C 1 - 10 straight, or branched chain alkyl except (S)-2- butyl, (2) C 3 - 10 cycloalkyl , (3) C 2 - 10 al lkenyl, (4) C 1 - 10 CF 3 - substitute d alkyl, (5) p henyl, (6) hal ophenyl, whe rein halo is ch loro, fluoro, b romo or iodo , (7) phenyl-C 1 -3 alkyl, or (8) sub stituted phen yl-C 1 -3 alkyl in which the s ubstituent is halo, such as fluoro, chloro , bromo, or i odo, C 1 -3 alkyl or C 1 -3 alkoxy.

[029] In anothe r embodimen t, the anti-m ethanogenic lovastatin ana log or deriva tive may hav e one or more 8-a cyloxy groups as described , for example , in U.S. Pate nt No.4,661, 483, the entire contents of which are incorpora ted by refere nce herein (including, by way of illus tration, discl osures of sp ecific compo unds and synthetic schemes). Fo r example, th e anti-methan ogenic lovas tatin analog o r derivative m ay have the fo rmula:

wherein: n is 0 to 5; R 1 is C 1 - 3 alky l; R 2 is hydro gen or C 1 -3 a lkyl; R 3 and R 4 independe ntly are hydro gen, C 1 - 3 alkyl, C 3 - 7 cycloalkyl, p henyl or sub stituted pheny l in which the substituents are X and Y and when n is 2 to 5, each of t he R 3 s and R 4 s are indep endently hydr ogen or C 1 - 3 a lkyl, c 3 - 7 cycl oalkyl or only one of the R 3 s and R 4 s is phen yl or substitut ed phenyl; R 5 is hydrogen o r hydroxy; R 6 and R 7 indep endently are hydrogen, or C 1 -3 alkyl or R 6 an d R 7 togethe r with the nit rogen atom t o which they are attached form a hete rocycle sele cted from

piperidiny l, morpholiny l, pyrrolidinyl, piperazinyl o r thiomorpho linyl; A is in w hich R 8 is

hydrogen or hydroxy; B is , in which R 9 is hydrogen or hydroxy; a, b, c and d r epresent sing le bonds, one of a, b, c or d rep resents a dou ble bond, or both a and c or both b and d represent double bonds provided

that when a is a doub le bond, A is and wh en d is a dou ble bond, B is and X and Y

independ ently are hyd rogen, haloge n, trifluorome thyl, C 1 -3 alk yl, nitro, cyan o or a group selected from : (a) R 10 O(CH 2 ) m in which m is 0 to 3 and R 10 is hydro gen, C 1 -3 alkyl or hy droxy-C 1 -3 a lkyl; (b) , in which R 11 is hydrogen, C 1 - 3 alkyl, h ydroxy-C 2 - 3 a lkyl, phenyl, naphthyl, amino-C 2 - 3 alkyl, C 1 - 3 alkylamino-C 1 - 3 alkyl, di(C 1 - 3 alkyl)amin o-C 1 - 3 alkyl, h ydroxy-C 2 - 3 a lkylamino-C 1 - 3 alkyl or di(hydrox y-C

2- 3 alkyl)-a mino-C 1 - 3 al kyl; (c) , in which R 12 is hydrogen, C 1 - 3 alkyl, hydr oxy-C 1 - 3 -

alkyl, C 1 - 3 alkoxy-C 1 - 3 a lkyl, phenyl o r naphthyl; (d ) R 13 R 14 N(C H 2 ) m , , in

which R 13 and R 14 inde pendently are hydrogen, C 1 - 3 alkyl, hydr oxy-C 2 - 3 alkyl or together w ith the nitroge n atom to which the y are attache d form a hete rocycle group selected fro m piperidinyl, pyrrolidinyl, p iperazinyl, m orpholinyl or thiomo rpholinyl; (e) R 15 S(O) p ( CH 2 ) m in whi ch p is 0 to 2 and R 15 is hydrogen, C 1 - 3 alkyl, am ino, C 1 - 3 alkylamin o or di(C 1 - 3 a lkyl)-amino; Z is hydrogen, C 1 - 5 alkyl or C 1 - 5 alkyl sub stituted with a member of the group consisting of phenyl, d imethylamino or acetylamin o; and pharm aceutically a cceptable salt s of the com pounds of the Form ula VI in whic h Z is hydrog en.

[030] In certai n embodime nts, the ant i-methanogen ic lovastatin analog or derivative m ay have functiona lized 8-acylox y groups as d escribed, for example, in U .S. Patent No s.4,864,038 , 4,937,264, 4 ,847,306, 5,049,696 , 4,766,145, 4,876,279, 4 ,876,366, 4,937263, 4,771 ,071, and 4,916,162, the e ntire contents of which are incor porated by re ference here in (including, by way of ill ustration, disc losures of sp ecific compo unds and synthetic schemes).

[031] In an em bodiment, the anti-methano genic lovasta tin analog or derivative may be any one described in U.S. P atent No. 4,864,038, the e ntire content s of which ar e incorporate d by referenc e herein (inc luding, by way of illu stration, disc losures of spe cific compou nds and synth etic schemes ) and have the formula:

wherein: m is 0 to 2; n is 0 to 5; R 1 is C 1 - 3 alk yl; R 2 is hydr ogen or C 1 - 3 alkyl; R 3 and R 4 independ ently are hydrogen , C 1 - 3 alkyl, C 3 - 7 cycloalkyl, phenyl or sub stituted phen yl in which the substituents are X and Y and when n is 2 to 5, each of the R 3 s and R 4 s are indepen dently hydrog en or C 1 - 3 alk yl, c 3 - 7 cycloa lkyl or only o ne of the R 3 s and R 4 s is phenyl or substituted phenyl; R 5 is hydrogen or h ydroxy;R 6 is phenyl or sub stituted pheny l wherein

the subst ituents are X and Y;A is , in whic h R 7 is hydro gen or hydrox y; B is CHR 8 which R 8 is hydrog en or hydroxy ; and a, b, c a nd d represe nt single bond s, one of a, b , c or d repres ents a double bond, or both a a nd c or bot h b and d represent do uble bonds provided that when a is a double bo nd, A is , and when d is a double bond, B is and X a nd Y indepe ndently are hydrogen, halogen, trifluoromethy l, C 1 - 3 alkyl, n itro, cyano or a group sele cted from (a) R 9 O(CH 2 ) m in which m is 0 to 3 and R 9 is hyd rogen, C 1 - 3 al kyl or hydroxy -C 1 - 3 alkyl; (b ) in whi ich R 10 is hydr ogen, C 1 - 3 alkyl, hy droxy-C 2 - 3 a lkyl, phenyl, n aphthyl, amin o-C 1 - 3 alkyl, C 1 - 3 alkylamin o-C 1 - 3 alkyl, d i(C 1 - 3 alkyl)a mino-C 1 - 3 alkyl, hyd roxy-C

2- 3 alky lamino-C 1 - 3 a lkyl or di(hydr oxy-C 2 - 3 alky l)amino-C 1 - 3 a lkyl; (c) om which R 11 is hydrog en, C 1 - 3 alky l, hydroxy-C 1 - 3 alkyl, C 1 - 3 alkoxy-C 1 - 3 alkyl, phenyl or naphthyl; (d) R 12 R 13 N(CH 2 ) m , in which R 12 and R 13 in dependently a re hydrogen, C 1 - 3 alkyl, hy droxy-C 2 - 3 alkyl o r together wit h the nitroge n atom to wh ich they are attached form a heterocyc le group sele cted from piperidiny l, pyrrolidinyl , piperazinyl, morpholinyl o r thiomorphol inyl; (e) R 14 S (O) p (CH 2 ) m in which p is 0 to 2 and R 14 is hyd rogen, C 1 - 3 a lkyl, amino, C 1 - 3 alkylamin o or di (C 1 - 3 a lkyl) amino; Z is hydrogen , C 1 - 5 alkyl or C 1 - 5 alkyl substitute d with a m ember of the group c onsisting of phenyl, dim ethylamino or acetylam ino; and pharmace utically acce ptable salts of the compoun ds of the For mula VIII in w hich Z is hydr ogen.

[032] In an em bodiment, the anti-methano genic lovasta tin analog or derivative may be any one described in U.S. P atent No. 4,847,306, the e ntire content s of which ar e incorporate d by referenc e herein (inc luding, by way of illu stration, disc losures of spe cific compou nds and synth etic schemes ) and have the formulae:

X, wherein, q is 0 to 5; n is 2 to 7; R 1 is C 1 - 6 alk yl, C 1 - 6 alkyl substituted w ith a group s elected from halogen, hydroxy, C 1 - 3 alkylthio , C 1 - 3 alkylsul finyl and C 1 - 3 alkylsulfonyl, C 1 - 6 alkylthio , C 1 - 6 alkylsul lfinyl, C 1 - 6 alk ylsulfonyl, C 3 - 7 cycl oalkylthio, C 3 - 7 cycloalkylsu lfinyl, C 3 - 7 cy cloalkylsulfon yl, C 3 - 7 cyclo alkyl, phenyl l, or phenyl s ubstituted with X a nd Y; R 2 is h ydrogen or h ydroxy; R 3 a nd R 4 indepe ndently are h ydrogen, C 1 - 3 alkyl, C 3 - 7 c ycloalkyl, phenyl o r phenyl subs tituted with X and Y and w hen q is 2 t o 5, each of the R 3 s and R 4 s are inde pendently hydrogen , C 1 - 3 alkyl, C 3 - 7 cycloalky l or only one of the R 3 s and R 4 s is phenyl or su bstituted phe nyl; A is , in whic h R 5 is hydro gen or hydro xy; B is CHR 6 in which R 6 i is hydrogen o r hydroxy; a, b, c an d d represent single bonds , one of a, b, c or d represe nts a double bond or both a and c or bot h b and d

represen t double bond s, provided th at when a is a double bon d, A is a nd when d is a double bond, B i s an d X and Y ind ependently a re hydrogen, halogen, triflu oromethyl, C 1 - 3 alkyl, nitro , cyano or a group s elected from: (a) R 7 O(CH 2 ) m in which m is 0 to 3 and R 7 is hydroge n, C 1 - 3 alkyl o r hydroxy-C 1 - 3 alkyl;(b) in which R 8 is h ydrogen, C 1 - 3 alkyl, hydroxy -C 2 - 3 alkyl, ph enyl, naphth yl, amino- C 1 - 3 alky l, C 1 - 3 alkyla mino-C 1 - 3 a lkyl, di(C 1 - 3 alkyl)amino-C 1 - 3 alkyl, hy droxy-C 2 - 3 al lkylamino-C 1 - 3 alkyl or di(hydrox y-C

2- 3 alkyl)a mino-C 1 - 3 alk yl; (c) in wh ich R 9 is hydr ogen, C 1 - 3 al lkyl, hydroxy- C 1 - 3 alkyl, C 1 - 3 alkox y-C 1 - 3 alkyl, p henyl, or nap hthyl; (d) R 10 R 11 N(CH 2 ) m , in whic h R 10 and R 11 independen tly are hydrog en, C 1 - 3 alkyl , hydroxy- C 2 - 3 alkyl or together w ith the nitrog en atom to w hich they are attached form a heterocyc le group sele cted from piperidiny l, pyrrolidinyl , piperazinyl, morpholinyl o r thiomorpho linyl; (e) R 12 S (O) p (CH 2 ) m in which p is 0 to 2 and R 12 is hy drogen, C 1 - 3 a lkyl, amino, C 1 - 3 alkylamin o or di(C 1 - 3 a lkyl)amino; Z is hydrogen, C 1 - 5 alkyl or C 1 - 5 alkyl substitute d with a grou p selected fr om phenyl, d imethylamino or acetylamin o; and pharm aceutically a cceptable salts of th e compounds of the Form ula X in which Z is hydroge n.

[033] In some e mbodiments , the anti-meth anogenic lov astatin analog or derivative may be as d ecribed in U.S. Pate nt No.4,766 ,145, the enti re contents of which are in corporated by reference he rein (includin g, by way of illustra tion, disclosur es of specific compounds a nd synthetic schemes) and be of formula e:

wherein, q is 0 to 5; n is 2 to 7; R 1 is C 1 - 6 alk yl; R 2 is hyd rogen or hyd roxy; R 3 and R 4 independ ently are hydrogen , C 1 - 3 alkyl, C 3 - 7 cycloalkyl , phenyl or ph enyl substitut ed with X and Y and when q is 2 to 5, ea ch of the R 3 s and R 4 s are indep endently hydr ogen, C 1 - 3 alk yl, C 3 - 7 cyclo alkyl or only o ne of the R 3 s and R 4 s is phenyl or substitute d phenyl; A i s , in which R 5 is hydrog en or hydroxy ; B is -CHR 6 - in which R 6 is hyd rogen or hydr oxy; a, b, c a nd d represen t single bond s, one of a, b , c or d repres ents a doubl e bond or both a a nd c or bot h b and d represent dou ble bonds, provided tha t when a is a double bo nd, A is and when d is a d ouble bond, B is a nd X and Y in dependently are hydrogen , halogen, trifluorom ethyl, C 1 - 3 al kyl, nitro, cya no or a group selected fro m: (a) R 7 O(C H 2 ) m in which m is 0 to 3 and R 7 is hydrogen , C 1 - 3 alkyl o r hydroxy-C 1 - 3 alkyl; (b) in which R 8 is hydro gen, C 1 - 3 alkyl, hyd roxy-C 2 - 3 alk yl, phenyl, na phthyl, amino -C 1 - 3 alkyl, C 1 - 3 alkylamin o-C 1 - 3 alkyl, d i(C 1 - 3 alkyl)a mino-C 1 - 3 alkyl, hyd roxy-C 2 - 3 alky lamino-C 1 - 3 a lkyl or di(hyd roxy-C 2 - 3 alky l)amino-C 1 - 3 alkyl; (c) in w hich R 9 is hydrogen , C 1 - 3 alkyl, hydroxy-C 1 - 3 alkyl, C 1 - 3 a lkoxy-C 1 - 3 a lkyl, phenyl, or naphthyl; (d) R 10 R 11 N(CH 2 ) m , in whic h R 10 and R 11 independen tly are hydrog en, C 1 - 3 alkyl , hydroxy- C 2 - 3 alkyl or together w ith the nitrog en atom to w hich they are attached form a heterocyc le group sele cted from piperidiny l, pyrrolidinyl , piperazinyl, morpholinyl o r thiomorpho linyl; (e) R 12 S (O) p (CH 2 ) m in which p is 0 to 2 and R 12 is hy drogen, C 1 - 3 a lkyl, amino, C 1 - 3 alkylamin o or di(C 1 - 3 a lkyl)amino; Z is hydrogen, C 1 - 5 alkyl or C 1 - 5 alkyl substitute d with a grou p selected fr om phenyl, d imethylamino or acetylamin o; and pharm aceutically a cceptable salts of th e compounds of the Form ula XII in whic h Z is hydrog en.

[034] In some embodiments , the anti-met hanogenic lov astatin analo g or derivativ e may be as described in U.S. P atent No. 4,876,366, the e ntire content s of which ar e incorporate d by referenc e herein (inc luding, by way of illu stration, disc losures of spe cific compou nds and synth etic schemes ) and may be of formulae:

wherein n is 0 to 5; R 1 is C 1 - 3 alky l; R 2 is hydro gen or C 1 - 3 a lkyl; R 3 and R 4 independen tly are hydro gen, C 1 - 3 alkyl, C 3 - 7 cycloalkyl, p henyl or sub stituted pheny l wherein the substituents are X and Y and when n is 2 to 5, each of t he R 3 s and R 4 s are indepe ndently hydro gen, C 1 - 3 alky l, C 3 - 7 cycloa lkyl or only on e of the R 3 s a nd R 4 s is phenyl or substituted p henyl; R 5 is h ydrogen or h ydroxy; X and Y independe ntly are hydro gen, halogen (F, Cl or Br), trifluo romethyl, C 1 - 3 alkyl, nitro, cyano or a gr oup selected from: (a) R 6 O (CH 2 ) m in whic h m is 0 to 3 and R 6 is hydrogen , C 1 - 3 alkyl or hydroxy-C 1 - 3 alkyl; (b) in which R 7 is hydrogen, C 1 - 3 alkyl, hydroxy-C 2 - 3 alkyl, phe nyl, naphthy l, amino-C 1 - 3 alkyl, C 1 - 3 al kylamino-C 1 - 3 alkyl, di(C 1 - 3 alkyl)amino- C 1 - 3 alkyl, hydroxy-C 2 - 3 alkylamin o-C 1 - 3 alkyl or di(hydroxy -C 2 - 3 alkyl)am ino-C 1 - 3 alky l; (c) in wh ich R 8 is hydrogen , C 1 - 3 alkyl, hydroxy-C 2 - 3 -alkyl, C 1 - 3 alkoxy-C 1 - 3 alkyl, phenyl or naphthyl; (d) R 9 R 10 N(CH 2 ) m , in which R 9 and R 10 inde pendently are hydrogen, C 1 - 3 alkyl, hyd roxy-C 2 - 3 alkyl or t ogether with the nitrogen atom to whic h they are a ttached form a heterocycli ic group sele cted from piperidiny l, pyrrolidinyl , piperazinyl, morpholinyl o r thiomorphol inyl; (e) R 11 S (O) p (CH 2 ) m in which p is 0 to 2 and

R 11 is hyd rogen, C 1 - 3 a lkyl, amino, C 1 - 3 alkylamino or di(C 1 - 3 al kyl)amino; A is in which R 12 is hydrog en or hydrox y; B is CHR 13 in which R 13 is hydrogen or hydroxy; and a, b, c a nd d repres ent single bonds, o ne of a, b, c or d represe nts a double bond, both a and c or bo th b and d re present doub le bonds

provided that when a i s a double bo nd, A is a nd when d is a double bon d, B is and Z is hydrog en, C 1 - 5 alkyl or C 1 - 5 alkyl substituted w ith a member of the group consisting of phenyl, dime thylamino or acetyl amino; and p harmaceutica lly acceptabl e salts of the compounds of the Form ula XIV in w hich Z is hydrogen .

[035] In some embodiments , the anti-met hanogenic lov astatin analo g or derivativ e may be as described in U.S. P atent No. 4,771,071, the e ntire content s of which ar e incorporate d by referenc e herein (inc luding, by way of illu stration, disc losures of spe cific compou nds and synth etic schemes ) and may be of formulae:

wherein, n is 0 to 5; R 1 is C 1 - 3 alky l; R 2 is hydro gen or C 1 - 3 a lkyl; R 3 and R 4 independe ntly are hydro gen, C 1 - 3 alkyl, C 3 - 7 cycloalkyl, p henyl or sub stituted pheny l in which the substituents are X and Y and when n is 2 to 5, each of t he R 3 s and R 4 s are indepe ndently hydro gen, C 1 - 3 alky l, C 3 - 7 cycloa lkyl or only on e of the R 3 s a nd R 4 s is phenyl o r substitute d phenyl; R 5 is hydro gen or hyd roxy; R 6 is hydrogen or C 1 -3 alk yl; A is in which R 7 is hydro gen or hydro xy; B is in wh ich R 8 is hyd rogen or hydroxy; and a, b, c an d d represen t single bonds , one of a, b , c or d repres ents a double bond, or bot h a and c

or both b and d repres ent double b onds provide d that when a is a double bond, A is when d is a double bo nd, B is and X a nd Y indepen dently are hyd rogen, haloge n, trifluorome thyl, C 1 - 3 alkyl, nitr o, cyano or a group selecte d from: (a) R 9 O(CH 2 ) m in w hich m is 0 t o 3 and R 9 is hydrogen, C 1 - 3 alkyl or

hydroxy-C 1 - 3 alkyl; (b) in which R 10 is hydrogen, al lkyl, hydroxy- C 2 - 3 alkyl, phenyl, naphthyl, am ino-C 1 - 3 alky l, C 1 - 3 alkyla mino-C 1 - 3 a lkyl, di(C 1 - 3 a lkyl)amino-C 1 - 3 alkyl, hyd roxy-C 2 - 3 alkylamin o-C 1 - 3 alkyl o r di(hydroxy-C 2 - 3 alkyl)ami no-C 1 - 3 alkyl; (c) in which R 11 is hydro gen, C 1 - 3 alkyl, h ydroxy-C 1 - 3 alkyl, C 1 - 3 alkoxy-C 1 - 3 alkyl, phe nyl or na phthyl; (d) R 12 R 13 N(CH 2 ) m , in whic h R 12 and R 13 independent ly are hydrog en, C 1 - 3 alkyl , hydroxy- C 2 - 3 alkyl or together w ith the nitrog en atom to w hich they are attached form a heterocyc le group sele cted from piperidiny l, pyrrolidinyl , piperazinyl, morpholinyl o r thiomorphol inyl; (e) R 14 S (O) p (CH 2 ) m in which p is 0 to 2 and R 14 is hy drogen, C 1 - 3 a lkyl, amino, C 1 - 3 alkylamin o or di(C 1 - 3 a lkyl)amino; Z is hydrogen, C 1 - 5 alkyl or C 1 - 5 alkyl substitute d with a m ember of the group c onsisting of phenyl, dim ethylamino or acetylam ino; and pharmace utically acce ptable salts of the compoun ds of the For mula XVI in w hich Z is hyd rogen.

[036] In certai n embodime nts, the anti- methanogeni c lovastatin analog or d erivative may have a hydroxym ethyl group, acyloxymeth yl group, car bamoyloxyme thyl group, a carboxy gro up, an alkox ycarbonyl group or a carbamoyl group substit uted on the 6 -position of t he polyhydro naphthyl moie ty as describ ed in, for example, U.S. Patent Nos. 4,940,727, 4,841,074 , 4,894,465, 4,894,466, 4 ,855,456, RE 36520, RE36481, and 5,116,870 , the entire c ontents of w hich are inco rporated by r eference here in (including , by way of il lustration, disclosur es of specific compounds and syntheti c schemes). In various e mbodiments, the anti-meth anogenic lovastatin analog or de rivative may possess a su bstituent in th e 6-position i n either the 6α or 6β stereo chemical position.

[037] In an em bodiment, the anti-methano genic lovasta tin analog or derivative m ay have a hy droxyalkyl group, ac yloxyalkyl or carbamoylox yalkyl group, for instance as described in U.S. Pate nt No. 4,940 ,727, the entire co ntents of whi ch are incorp orated by ref erence herein (including, b y way of illu stration, discl osures of specific c ompounds an d synthetic sc hemes) and may be of form ulae:

wherein, R is

R 1 an d R 5 are ind ependently: (1) C 1 - 10 alky l; (2) substitu ted C 1 - 10 alkyl in w hich one or m ore substitue nt(s) is: halog en, hydroxy, C 1 - 10 alkoxy, C 1 - 5 alkoxycar bonyl, C 1 - 5 ac yloxy, C 3 - 8 cycloalk yl, phenyl, su bstituted phen yl in which th e substituent s are X and Y , C 1 - 10 alkylS( O) n in which n is 0 to 2, C 3 - 8 cyclo alkylS(O) n , p henylS(O) n , s ubstituted ph enylS(O) n in which the sub stituents are X and Y, an d oxo; (3) C 1 - 10 alko xy; (4) C 2 - 10 alkenyl; (5) C 3 - 8 cycloalkyl; (6) substitute d C 3 - 8 cycloa lkyl in which o ne substituen t is: C 1 - 10 alkyl, sub stituted C 1 - 10 alkyl in which the substitue nt is: haloge n, hydroxy, C 1 - 10 alkoxy, C 1 - 5 alkoxycarb onyl, C 1 - 5 acyloxy, phenyl, subst ituted phenyl in which the s ubstituents a re X and Y, C 1 - 10 alkylS(O) n , C 3 - 8 cycloal kylS-(O) n , phenylS( O) n , substitut ed phenylS(O ) n in which t he substituen ts are X and Y, and oxo , C 1 - 10 alkylS (O) n , C 3 - 8 cycloalky lS(O) n , pheny lS(O) n , subst ituted phenylS (O) n in which the substitue nts are X an d Y, halogen , hydroxy, C 1 - 10 alko xy, C 1 - 5 alko xycarbonyl, C 1 - 5 acyloxy, p henyl, and s ubstituted phe nyl in which the substitue nts are X and Y; (7 ) phenyl; (8) substituted ph enyl in which the substitue nts are X an d Y; (9) amino ; (10) C 1 - 5 a lkylamino; (11) di(C 1 - 5 alkyl)amino ; (12) pheny lamino; (13) s ubstituted ph enylamino in which the sub stituents are X and Y; (14) phen yl C 1 - 10 alkyla mino; (15) su bstituted phe nyl C 1 - 10 alky lamino in whic h the substitu ents are X a nd Y; (16) a membe r selected fro m: piperidiny l, pyrrolidinyl, piperazinyl, m orpholinyl, a nd thiomorph olinyl; and (17) R 9 S in which R 9 is: C 1 - 10 alky l, phenyl, an d substituted phenyl in wh ich the subst ituents are X and Y; R 2 a nd R 6 are independ ently (1) hyd rogen; (2) C 1 - 5 alkyl; (3 ) substituted C 1 - 5 alkyl in which the substituent i s phenyl, dimethyla mino, and ac etylamino, an d (4) 2,3-dihy droxypropyl; R 3 and R 4 are independentl ly: (1) hydroge n; (2) C 1 - 1 0 alkyl; ( 3) substituted C 1 -10 alkyl in which one or more sub stituent(s) is halogen, hydr oxy, C 1 - 10 alk oxy, C 1 - 5 alkoxyca rbonyl, C 1 - 5 a cyloxy, C 3 - 8 c ycloalkyl, phe nyl, substitut ed phenyl in which the sub stituents are X and Y, C 1 - 10 alky lS(O) n , C 3 - 8 c ycloalkylS(O) n , phenylS(O) n , substituted phenylS(O) n in which the substituents a re X and Y, and o xo; (4) C 2 - 10 alkenyl; (5) substituted C 2 - 10 alkenyl i n which one or more sub stituent(s) is: halogen, hydroxy, C 1 - 10 alkoxy, C 1 - 5 alkoxyca rbonyl, C 1 - 5 a cyloxy, C 3 - 8 c ycloalkyl, phe nyl, substitut ed phenyl in which the substitue nts are X and Y, C 1 - 10 alky lS(O) n , C 3 - 8 c ycloalkylS(O ) n , phenylS(O ) n , substituted phenylS(O) n in which the subst ituents are X and Y, and ox o; (6) C 3 - 8 cy cloalkyl; (7) s ubstituted C 3 - 8 cycloalkyl in which one s ubstituent is: C 1 - 10 alkyl, subst ituted C 1 - 10 alkyl in whic h the subst ituent is: ha logen, hydrox y, C 1 - 10 alk oxy, C 1 - 5 alkoxyca rbonyl, C 1 - 5 a cyloxy, pheny l, substituted phenyl in wh ich the substit uents are X a nd Y, C 1 - 10 a lkylS(O) n , C 3 - 8 cyclo alkylS(O) n , p henylS(O) n , s ubstituted phe nylS(O) n in w hich the subs tituents are X and Y, and oxo, C 1 - 10 alkylS(O) n , C 3 - 8 cycloa lkylS(O) n , ph enylS(O) n , su bstituted phe nylS(O) n in w hich the sub stituents are X and Y, halogen, hydroxy, C 1 - 10 alkoxy, C 1 - 5 alkoxycarbo nyl, C 1 - 5 acy loxy, phenyl, and substitute d phenyl in which the substitue nts are X and Y; (8) pheny l; (9) substitut ed phenyl in w hich the sub stituents are X and Y; R 7 a nd R 8 are independ ently:(1) hydr ogen; (2) C 1 - 10 alkyl; (3) substituted C 1 -10 alkyl in which one or more substit uent(s) is halogen, hydroxy, C 1 - 10 alkoxy, C 1 - 10 alkoxycarbo nyl, C 1 - 5 acy loxy, C 3 - 8 cyc loalkyl, pheny l, substituted phenyl in which the substituents are X and Y , C 1 - 10 alkyl S (O) n in which n is 0 to 2, C 3 - 8 cycloalky l S(O) n , phen yl S(O) n ; substitute d phenyl S(O ) n in which th e substituents are X and Y , and oxo; (4) C 2 - 10 alkenyl l; (5) C 3 - 8 cycl oalkyl; (6) aminocar bonyl; (7) su bstituted amin ocarbonyl in which one or more substit uent(s) is C 1 - 5 alkyl, C 3 - 8 c ycloalkyl, phenyl, s ubstituted phe nyl in which the substituen ts are X and Y; (8) pheny l; (9) substitut ed phenyl in which the substitue nts are X an d Y; (10) C 1 - 10 alkylcarb onyl; (11) C 3 - 8 cycloalkylc arbonyl; (12 ) phenylcarbo nyl; (13) substitute d phenylcarb onyl in which the substitu ents are X an d Y; and (14 ) a nitrogen- containing he terocyclic group se lected from piperidinyl, pyrrolidinyl, p iperazinyl, m orpholinyl a nd thiomorph olinyl; and X and Y independ ently are hyd rogen, haloge n, trifluorome thyl, C 1 -3 al kyl, nitro, cya no or a group selected fro m:(1) R 10 O(CH 2 ) m in which m is 0 to 3 and R 10 is hydro gen, C 1 - 3 alkyl or hy droxy-C 2 - 3 a lkyl; (2) in which R 11 i s hydrogen, C 1 - 3 alkyl, hy droxy-C 2 - 3 a lkyl, phenyl, naphthyl, amino-C 1 - 3 alkyl, C 1 - 3 alkylamino-C 1 - 3 alkyl, di(C 1 - 3 alkyl)amin o-C 1 - 3 alkyl, h ydroxy-C 2 - 3 a lkylamino-C 1 - 3 alkyl or

di(hydrox y-C 2 - 3 alkyl) amino-C 1 -3 a lkyl; (3) i n which R 12 i s hydrogen, C 1 - 3 alkyl, hyd roxy-C 2 - 3 alkyl, C 1 - 3 alkoxy-C 1 - 3 alkyl, pheny l or naphthy l; (4) in which R 13 and R 14 inde pendently are hydrogen, C 1 - 3 alkyl, hydr oxy-C 2 - 3 alkyl or together w ith the nitroge n atom to which the y are attache d form a hete rocyclic grou p selected fro m piperidinyl, pyrrolidinyl, p iperazinyl, m orpholinyl or thiomo rpholinyl; (5) R 15 S(O) n (C H 2 ) m in which R 15 is hydro gen, C 1 - 3 alk yl, amino, C 1 - 3 alkylamino or di(C 1 - 3 alkyl)ami no; and a, b a nd c each rep resent single bonds or one of a, b and c represents a double bond or both a and c rep resent double bonds; or a p harmaceutica lly acceptabl e salt thereof.

[038] In certain embodiment s, the anti-me thanogenic l ovastatin ana log or derivat ive may have a methyl group in the 6-position in the 6β ste reochemical position as d escribed, for example, in U .S. Patent A pplication No. 07/092,354 filed September 2 , 1987, the e ntire contents of which ar e incorporate d by referen ce herein (including , by way of ill ustration, disc losures of sp ecific compou nds and synth etic schemes ). [039] In certain embodimen ts, the anti-m ethanogenic lovastatin ana log or deriva tive may hav e contain gem-disu bstitution at t he 6,6 positio ns of the poly hydronaphthy l moiety as de scribed, for e xample, in U .S. Patent No.4,866 ,090, the ent ire contents o f which are in corporated by reference he rein (including , by way of il lustration, disclosur es of specific compounds and synthet ic schemes). For exampl e, the anti-m ethanogenic lovastatin analog or derivative ma y have the fo rmula

wherein, R 1 is selecte d from: (1) C 1 - 10 alkyl; (2) substituted C 1 - 10 alkyl in which one or more substit uent(s) is selected from (a) hal ogen, (b) hyd roxy, (c) C 1 - 10 alkoxy, (d ) C 1 - 5 alkoxy carbonyl, (e) C 1 - 5 acyloxy , (f) C 3 - 8 cycloalky l, (g) phenyl, (h) substitute d phenyl in w hich the subs tituents are X and Y, (i) C 1 - 10 alkyls(0) n i n which n is 0 to 2, (j) C 3 - 8 cycloa lkylS(0) n , (k) phenylS(0) n , (1) substitute d phenylS(0) n in which the substituents are X and Y, and (m ) oxo; (3) C 1 - 10 alkoxy; (4) C 2 - 10 alkenyl ; (5) C 3 - 8 cycl oalkyl; (6) sub stituted C 3 - 8 cycloalkyl in w hich one substitue nt is selected from (a) C 1 - 10 alkyl (b) sub stituted C 1 - 10 alkyl in whic h the substitu ent is selecte d from (i) halogen, (ii) hydroxy, (iii) C 1 - 10 alko xy, (iv) C 1 - 5 alk oxycarbo nyl, (v) C 1 - 5 a cyloxy, (vi) p henyl, (vii) s ubstituted phenyl in which the su bstituents are X and Y (vii i) C 1 - 10 alkylS (0) n , (ix) C 3 - 8 cycloalkylS(0 ) n , (x) phenyl S(0) n , (xi) substitute d phenylS(0 ) n in which t he substituen ts are X and Y, and (xii ) oxo, (c) C 1 - 10 alkylS(0) n , (d) C 3 - 8 cycloalky lS(0)n, (e) ph enylS(0) n , (f) substituted p henylS(0) n in which the su bstituents are X and Y, (g) halogen, (h) hydro xy, (i) C 1 - 10 a lkoxy, (j) C 1 - 5 alkoxycarbo nyl, (k) C 1 - 5 a cyloxy, (l) ph enyl, and (m ) substituted phenyl in which the substituents are X and Y; (7) phenyl; (8 ) substituted phenyl in wh ich the substi tuents are X a nd Y; (9) amino; (10) C 1 - 5 alkyla mino; (11) d i(C 1 - 5 alkyl)am ino; (12) phe nylamino; (13) substituted phenylamino in which the subst ituents are X and Y; (14) p henyl C 1 - 10 a lkylamino; (15 ) substituted phenyl C 1 - 10 a lkylamino in which the substitue nts are X an d Y; (16) a member sele cted from (a ) piperidinyl, (b) pyrrolidin yl, (c) pipera zinyl, (d) morpholin yl, and (e) th iomorpholinyl ; and (17) R 6 S in which R 6 is selected fr om(a) C 1 - 10 a lkyl, (b) pheny l, and (c) substitute d phenyl in w hich the sub stituents are X and Y; R 2 and R 3 are in dependently selected from :(1) C 1 - 10 alkyl; and (2) substitute d C 1 - 10 alkyl in which one o r more subst ituent(s) is se lected from (a ) halogen, (b ) hydroxy, (c) C 1 - 10 alkoxy, (d) C 1 - 5 alkoxycarb onyl, (e) C 1 - 5 acyloxy, (f) C 3 - 8 cycloalky l, (g) phenyl,o r (3) togethe r with the carbon a tom to which they are atta ched form a C 3 - 8 carbocyc lic ring;R 4 is selected from :(1) hydrogen ; (2) C 1 - 5 alkyl; (3) substituted C 1 - 5 alkyl in wh ich the subs tituent is sele cted from (a) phenyl, (b) d imethylamino , and (c) acetylam ino, and (4) 2 ,3-dihydroxy propyl; X and Y independe ntly are hyd rogen, haloge n, trifluorome thyl, C 1 - 3 alkyl, nitr o, cyano or g roup selected from:(1) R 8 O(CH 2 ) m in w hich m is 0 to 3 and R 8 is hydrogen, C 1 - 3 alkyl or hydroxy C 2 - 3 alkyl;(2) in whi ch R 9 is hydr ogen, C 1 - 3 al kyl, hydroxy C 2 - 3 alkyl, phenyl, n aphthyl, am ino-C 1 - 3 alky l, C 1 - 3 alkyla mino-C 1 - 3 alk yl, di(C 1 - 3 a lkyl)amino-C 1 - 3 alkyl, hyd roxy C 2 - 3 alkylamin o-C 1 - 3 alkyl o r di(hydroxy-C 2 - 3 alkyl) am ino-C 1 - 3 alkyl ; (3) in which R 10 is hydro gen, C 1 - 3 alkyl, hydroxy-C 2 - 3 alkyl, C 1 - 3 alk oxy C 1 - 3 alkyl, phenyl o r naphth yl; (4) in whic h R 11 and R 12 independen tly are hydro gen, C 1 - 3 alkyl, hyd roxy-C 2 - 3 alky l or together with the nitro gen atom to w hich they are attached for m a heterocy clic group selected from piperidin yl, pyrrolidiny l, piperazinyl, morpholinyl o r thiomorphol inyl; (5) R 13 S(0) n (CH 2 ) m in which R 13 is hy drogen, C 1 - 3 alkyl, amino, C 1 - 3 alkylamin o or di(C 1 - 3 alkyl)amino;o r a pharmace utically accep table salt thereof.

[040] In certain embodimen ts, the anti-m ethanogenic lovastatin ana log or deriva tive may hav e contain two doub le bonds in th e 4,4a- and 5 ,6- positions o r one double bond in the 5 ,6- position of the polyhydr onaphthyl moiety as described, fo r example, in U.S. Patent No.4,857,547 , the entire c ontents of wh ich are incorp orated by reference herein (inclu ding, by way o f illustration, disclosures o f specific com pounds and s ynthetic sche mes). For example, the anti-meth anogenic lova statin analog or derivative may have the formula:

wherein, R is CH 2 OH , or CO 2 R 4 ;R 1 and R 3 a re independe ntly selected from:(1) C 1 - 10 alkyl; (2)

substitute d C 1 - 10 alkyl in which one or more sub stituent(s) is selected from : (a) halogen , (b) hydroxy , (c) C 1 - 10 alkoxy, (d ) C 1 - 5 alkoxyc arbonyl, (e) C 1 - 5 acyloxy, ( f) C 3 - 5 cycloa lkyl, (g) pheny l, (h) substitu ted phenyl in which the substitue nts are X and Y, and (i) oxo ; (3) C 3 - 8 cyc loalkyl; (4) su bstituted C 3 - 8 cycloalkyl in w hich one sub stituent is selected from:(a) C 1 -10 alkyl, (b) su bstituted C 1 -10 alkyl in whic h the substitu ent is selecte d from: (i) ha logen, (ii) hydroxy, (iii) C 1 - 10 alko xy (iv) C 1 - 5 a cyloxy, (v) C 1 - 5 alkoxycarb onyl, (vi) phe nyl, (vii) subs tituted pheny l in which the subst ituents are X and Y, and (v iii) oxo, (c) ha logen, (d) hy droxy, (e) C 1 - 10 alkoxy, (f) C 1 - 5 alkoxycar bonyl, (g) C 1 - 5 acyl oxy, (h) phen yl, (i) substit uted phenyl in which the substituents are X and Y ; (5) phenyla mino; (6) substitute d phenylamin o in which th e substituent s are X and Y ; (7) phenyl C 1 - 10 alkylami ino; and (8) s ubstituted phenyl C 1 - 10 alkylamin o in which th e substituents are X and Y . R 2 and R 4 are independ ently selected from:(a) hydrogen ; (b) C 1 -5 alk yl; (c) substitu ted-C 1 -5 alky l in which the substituent i s selected fro m PA4 (i) ph enyl, PA4 (ii) dimet hylamino, and PA4 (iii) ace tylamino; and (d) 2,3-dihyd roxypropyl; X and Y indepe ndently are hydrogen, halogen, trifluoromethy l, C 1 -3 alkyl, nitro, cyano o r group selec ted from:(1) R 8 O(CH 2 ) m in which m is 0 to 3 and R 8 is hyd rogen, C 1 - 3 a lkyl or hydrox y-C 2 - 3 alkyl;( 2) in whic h R 9 is hydro gen, C 1 - 3 alkyl, hyd roxy-C 2 - 3 alk yl, phenyl, na phthyl, amino -C 1 - 3 alkyl, C 1 - 3 alkylamin o-C 1 - 3 alkyl, d i(C 1 - 3 alkyl)a mino-C 1 - 3 alkyl, hyd roxy-C 2 - 3 alk ylamino-C 1 - 3 alkyl or di(hy droxy-C 2 - 3 al kyl)amino-C 1 - 3 alkyl;(3) in which R 10 is hy drogen, C 1 - 3 alkyl, hydroxy -C 2 - 3 alkyl, C 1 - 3 alkoxy-C 1 - 3 alkyl, phen yl or naphthy l;(4) R 11 R 12 N(CH 2 ) m , in which R 11 and R 12 ind ependently a re hydrogen, C 1 - 3 alkyl, hyd roxy-C 2 - 3

alkyl or t ogether with the nitrogen atom to whic h they are a ttached form a heterocycli ic group sele cted from piperidiny l, pyrrolidinyl , piperazinyl, m orpholinyl or thiomorpholi nyl;(5) R 13 S(O ) n (CH 2 )m in which R 13 is hydrogen, C 1 - 3 alky l, amino, C 1 - 3 alkylamino o r di(C 1 -3 alk yl)amino; a r epresents a s ingle bond o r a double b ond; or a pharmace utically acce ptable salt the reof.

[041] In certain embodiment s, the anti-me thanogenic lo vastatin analo g or derivativ e may have c ontain an aminoalk yl group, or substituted a minoalkyl gr oup, on the 6-position o f the polyhyd ronaphthyl m oiety as described , for example , in U.S. Pa tent No. 4,857,546 (includ ing, by way o f illustration, disclosures o f specific compoun ds and synth etic schemes ), the entire contents of w hich are inc orporated by reference he rein. For example, the anti-meth anogenic lova statin analog or derivative may have the formula:

[042] wherein, R 1 is select ed from:(1) C 1 - 10 alkyl; (2 ) substituted C 1 - 10 alkyl i in which one or more substitue nt(s) is select ed from:(a) ha logen, (b) hy droxy, (c) C 1 - 10 alkoxy, (d) C 1 - 5 alkoxycar bonyl, (e) C 1 - 5 acyloxy, (f) C 3 - 8 cy cloalkyl, (g) p henyl, (h) su bstituted phen yl in which th e substituent s are X and Y , (i) C 1 - 10 alky lS (O) n in which n i s 0 to 2, (j) C 3 - 8 cycloalkyl(O ) n , (k) pheny lS(O) n , (l) sub stituted phen ylS(O) n in whi ich the substit uents are X and Y, and (m) oxo ; (3) C 1 - 10 al koxy; (4) C 2 - 10 alkenyl; (5) C 3 - 8 cycloalk yl; (6) substit uted C 3 - 8 cyc loalkyl in which on e substituent is selected from (a) C 1 - 10 alkyl (b) s ubstituted C 1 - 10 alkyl in w hich the subs tituent is selected from:(i) halog en, (ii) hydrox y, (iii) C 1 - 10 a lkoxy, (iv) C 1 - 5 alkoxycarbo nyl, (v) C 1 - 5 a cyloxy (vi) ph enyl, (vii) substitute d phenyl in which the su bstituents are X and Y,(v iii) C 1 - 10 alky lS(O) n , (ix) C 3 - 8 cycloalkyl S(O) n , (x) phenylS( O) n , (xi) subst ituted phenylS (O) n in which the substitue nts are X and Y, and (xii) o xo, (c) C 1 - 10 a lkylS(O) n (d) C 3 - 8 c ycloalkylS(O) n , (e) phenylS (O) n , (f) subs tituted pheny lS(O) n in whi ch the substit uents are X a nd Y, (g) halogen, (h) hydroxy, (i) C 1 - 10 alkox y, (j) C 1 - 5 al koxycarbonyl, (k) C 1 - 5 acy loxy, (l) phen yl, and (m) s ubstituted phenyl in which the su bstituents are X and Y; (7) phenyl; (8) s ubstituted ph enyl in which the substitue nts are X and Y; (9 ) amino; (10) C 1 - 5 alkylami no; (11) di(C 1 - 5 alkyl)amino ; (12) phenyl amino; (13) s ubstituted phe nylamino in which the substituen ts are X and Y; (14) phen yl C 1 - 10 alkyla mino; (15) s ubstituted phe nyl C 1 - 10 alky lamino in which the substituents are X and Y; (16) a memb er selected fr om: (a) piper idinyl, (b) pyrr olidinyl, (c) pi perazinyl, (d) morph olinyl, and (e ) thiomorphol inyl; and (17) R 6 S in which R 6 is selecte d from:(a) C 1 - 10 alkyl, (b) ph enyl, and (c) subst ituted phenyl in which the substituents are X and Y ; R 2 and R 3 are independ ently selected from:(1) hydrogen ; (2) C 1 - 10 al kyl; (3) subst ituted C 1 - 10 a lkyl in which one or more substituent(s ) is selected from: (a) halogen, (b) hydroxy, ( c) C 1 - 10 alkox y, (d) C 1 - 5 alk oxycarbonyl, (e) C 1 - 5 acylo xy, (f) C 3 - 8 cy cloalkyl, (g) p henyl, (h) substitute d phenyl in w hich the sub stituents are X and Y, and (i) oxo; (4) C 1 - 10 acyl; (5) s ubstituted C 1 - 10 acyl in which on e or more substituent(s) is selected from (a) ha logen, (b) h ydroxy, (c) C 1 - 10 alkoxy, (d) C 1 - 5 alkoxyca rbonyl, (e) C 1 - 5 acyloxy, (f) C 3 - 8 cycloalk yl, (g) pheny l, (h) substitu ted phenyl in which the su bstituents are X and Y, and (i) ox o; (6) phenyl carbonyl; (7) substituted ph enylcarbonyl in which the substituents a re X and Y; (8) C 1 - 5 alkylamin ocarbonyl; (9 ) di(C 1 - 5 alky l)aminocarbo nyl; (10) phe nylaminocarb onyl; (11) s ubstituted phenylam inocarbonyl in which the substituents are X and Y; (12) phe nyl C 1 - 10 alk ylaminocarbo nyl; (13) substitute d phenyl C 1 - 10 alkylamino carbonyl in w hich the su bstituents are X and Y; R 4 is selected from:(1) hydrogen ; (2) C 1 - 5 alk yl; (3) subst ituted C 1 - 5 a lkyl in which the substitue nt is selecte d from:(a) p henyl, (b) dimethyla mino, and (c ) acetylamino , and (4) 2,3-d ihydroxyprop yl;X and Y in dependently a re hydrogen , halogen, trifluorom ethyl, C 1 - 3 al kyl, nitro, cya no or group selected from :(1) R 8 O(CH 2 ) m in which m is 0 to 3 and R 8 is hydrogen , C 1 - 3 alkyl or hydroxy-C 2 - 3 alkyl;(2)

hydroxy-C 2 - 3 alkyl, phe nyl, naphthy l, amino-C 1 - 3 alkyl, C 1 - 3 al kylamino-C 1 - 3 alkyl, di(C 1 - 3 alkyl)amino- C 1 - 3 alkyl, hydroxy-C 2 - 3 alkylamin o-C 1 - 3 alkyl o r di(hydroxy- C 2 - 3 alkyl; am ino-C 1 - 3 alkyl ;(3) in wh ich R 10 is hydrogen , C 1 - 3 alk Yl, hydrox y-C 2 - 3 alkyl , C 1 - 3 alk oxy-C 1 - 3 al kyl, phenyl or napht hyl; (4) in whic h R 11 and R 12 independen tly are hydro gen, C 1 - 3 alkyl, hyd roxy-C 2 - 3 alky l or together with the nitro gen atom to w hich they are attached for m a heterocy clic group selected from piperidin yl, pyrrolidiny l, piperazinyl , morpholinyl or thiomorph olinyl; (5) R 13 S(O) n (CH 2 ) m in which R 13 is hyd rogen, C 1 - 3 a lkyl, amino, C 1 - 3 alkylamino or di(C 1 - 3 al kyl)amino; and a, b, c and d each repres ent single bonds or one of a, b, c and d repres ents a double bond or bot h a and c or b and d repre sent double b onds;or a pharmace utically acce ptable salt the reof. [043] In certai n embodimen ts, the anti- methanogenic lovastatin a nalog or de rivative may be a 5'- oxygenat ed derivative of lovastati n and analo g thereof at the 8'-acyl side chain a nd 6'-positio n of the polyhydro naphthyl ring as described , for example , in U.S. Pate nt No.4,921, 974, the entire contents of which are incorpora ted by refere nce herein (including, by way of illus tration, discl osures of sp ecific compo unds and synthetic schemes). Fo r example, th e anti-methan ogenic lovas tatin analog o r derivative m ay have the fo rmula:

or pharm aceutically ac ceptible salts thereof, whe rein, R 1 is s elected from: (1) C 1 - 10 alky l; (2) substit uted C 1 - 10 alkyl in w hich one or more substitu ent(s) is sele cted from: (a ) halogen, (b ) hydroxy, (c ) C 1 - 10 alkoxy , (d) C 1 - 5 alkoxyca rbonyl, (e) C 1 - 5 acyloxy, (f) C 3 - 8 cycloalk yl, (g) pheny l, (h) substitu ted phenyl in which the su bstituents are X an d Y, (i) C 1 - 10 alkylS(0) n in which n is 0 to 2, (j) C 3 - 8 cycloalkylS (0) n , (k) phen ylS(0) n , (l) s ubstituted phenylS( 0) n in which the substituen ts are X and Y, and (m) oxo; (3) C 1 - 10 alkoxy; (4) C 2 - 10 alkeny l; (5) C 3 - 8 cycloalky l; (6) substitu ted C 3 - 8 cyclo alkyl in which one substitu ent is selecte d from (a) C 1 - 10 alkyl (b) s ubstituted C 1 - 10 alk yl in which the substitue nt is selecte d from (i) h alogen, (ii) h ydroxy, (iii) C 1 - 10 alkoxy, (iv) C 1 - 5 alkoxyca rbonyl, (v) C 1 - 5 acyloxy, (vi ) phenyl, (vii) substituted p henyl in whic h the substitu ents are X a nd Y (viii) C 1 - 10 alky lS(0) n , (ix) C 3 - 8 cycloalkylS (0) n , (x) phe nylS(0) n , (xi) substituted p henylS(0) n in which the su bstituents are X and Y, and (xii) o xo, (c) C 1 - 10 a lkylS(0) n , (d) C 3 - 8 cycloalky lS(0) n , (e) ph enylS(0) n , (f) substituted ph enylS(0) n in which the substituen ts are X and Y, (g) haloge n, (h) hydrox y, (i) C 1 - 10 alk oxy, (j) C 1 - 5 a lkoxycarbony l, (k) C 1 - 5 acyloxy, (l) phenyl, and (m) substitu ted phenyl in which the su bstituents are X and Y; (7) phenyl; (8) s ubstituted phenyl in which the s ubstituents a re X and Y; (9) amino; (10) C 1 - 5 alkyla mino; (11) d i(C 1 - 5 alkyl)am ino; (12) phenylam ino; (13) sub stituted pheny lamino in wh ich the subst ituents are X and Y; (14) p henyl C 1 - 10 a lkylamino; (15) subs tituted pheny l C 1 - 10 alkylam ino in which the substitue nts are X and Y; (16) a me mber selecte d from (a) piperidiny l, (b) pyrrolid inyl, (c) piper azinyl, (d) mo rpholinyl, and (e) thiomorp holinyl; and (17) R 5 S in w hich R 5 is selected from (a) C 1 - 10 alkyl, (b) phe nyl, and (c) s ubstituted ph enyl in which the substitue nts are X and Y; R 4 is; (1) hydro gen; (2) C 1 - 10 alkyl; and (3) substituted C 1 - 10 alkyl in w hich one or m ore substituen ts is selected from: (a) halogen, (b) hydroxy, (c) C 1 - 10 alk oxy (d) C 1 - 5 alkoxycarbo nyl, (e) C 1 - 5 alkylacyloxy, (f) phenylac yloxy, (g) phenoxyc arbonyl, (h) phenyl C 1 - 5 a lkylacyloxy, ( i) phenyl C 1 - 5 alkoxy, (j) a mino, (k) C 1 - 5 alkylamino, (l) di(C 1 - 5 alkyl)ami no, (m) pheny lamino, (n) su bstituted phe nylamino in w hich the subs tituents are X and Y; (o) p henyl C 1 - 5 alkylamin o, (p) substit uted phenyl C 1 - 5 alkylamino in which the substituents are X and Y , (q) C 3 - 8 cyc loalkyl, (r) phenyl, (s ) substituted phenyl in wh ich the substit uents are X a nd Y, (t) phe nylS(0) n , (u) s ubstituted ph enyl S(0) n in which t he substituen ts are X and Y, (v) phenyl C 1 - 5 alkyl S(0 ) n , (w) C 1 - 5 alk ylS(0) n ; (x) p henylaminoac yloxy, (y) C 1 - 5 alkyl aminoacyloxy , (z) C 1 - 5 alky lacylamino, ( aa) di(phenylC 1 - 5 alkyl)pho sphonyl (bb) d i(C 1 - 5 alkyl)p hosphinyl (4) R 4 to gether with t he carbon at om to which it is attache d represents a C 3 - 8 carbo cyclic ring; R 5 and R 6 independ ently are H, O H, OR 7 or R 5 and R 6 toge ther with the carbon to whi ch they are a ttached repre sent C=O or R 5 and R 6 together with the carb on to which t hey are attac hed represen t a carbocycli ic ring of 4 to 7 atoms; provided that when R 5 is H, R 6 is O H or OR 7 , an d when R 5 is OH, R 6 is H, and when R 5 is OR 7 , R 6 i s H; R 7 is phenylC 1 -3 alkyl, C 1 -5 al kyl; R 8 and R 9 independen tly are H, C 1 -3 alky l, phenylC 1 -3 alkyl or aryl wherein aryl is phenyl nap hthyl, pyridyl, furanyl, thien yl or phenyl, naphthyl, pyridyl, fu ranyl or thien yl substituted with groups X and Y prov ided that wh en R 7 is R 8 is not H and whe n R 7 is neith er R 8 nor R 9 is H; X and Y are indep endently sele cted from: (a ) OH, (b) halogen, (c) trifluoro methyl, (d) C 1 - 3 alkoxy, (e) C 1 - 3 alk ylcarbonyloxy , (f) phenyl lcarbonyloxy, (g) C 1 - 3 alkyoxyca rbonyl, (h) ph enyloxycarbo nyl, (i) hydrog en; (j) C 1 - 5 al kyl; a is a sing le bond or a double bond.

[044] In certai n embodimen ts, the anti- methanogenic lovastatin a nalog or de rivative may be a 5'- oxygenat ed derivative of lovastatin and analog th ereof wherein position 5 of the polyhydr onaphthyl ring is singly or doubly bonded to ox ygen or incor porated into a C 3 -7 carbocy clic ring as d escribed, for e xample, in U .S. Patent No.4,963 ,538, the ent ire contents o f which are in corporated by reference he rein (including , by way of il lustration, disclosur es of specific compounds and synthet ic schemes). For exampl e, the anti-m ethanogenic lovastatin analog or derivative ma y have the fo rmula:

wherein, R 1 is selecte d from:(1) C 1 - 10 alkyl; (2) substituted C 1 - 10 alkyl in w hich one or more substit uent(s) is selected from: (a) ha logen, (b) hy droxy, (c) C 1 - 10 alkoxy, (d ) C 1 - 5 alkoxy carbonyl, (e) C 1 - 5 acyloxy , (f) C 3 - 8 cycloalky l. (g) phenyl, (h) substituted phenyl in wh ich the subst ituents are X and Y, (i) C 1 - 10 alkylS(O) n i n which n is 0 to 2, (j) C 3 - 8 cycloa lkylS(O) n , (k) phenylS(O) n , (l) substitute d phenylS(O) n in which the substituents are X and Y, and (m ) oxo; (3) C 1 - 10 alkoxy; (4) C 2 - 10 alkenyl ; (5) C 3 - 8 cycl oalkyl; (6) sub stituted C 3 - 8 cycloalkyl in w hich one substitue nt is selected from: (a) C 1 - 10 alkyl (b) sub stituted C 1 - 10 alkyl in whic h the substitu ent is selecte d from: (i) halogen, (ii) hydroxy, (iii) C 1 - 10 alko xy, (iv) C 1 - 5 alkoxycarbon yl, (v) C 1 - 5 a cyloxy, (vi) p henyl, (vii) s ubstituted phenyl in which the su bstituents are X and Y (viii ) C 1 - 10 alkylS( O) n , (ix) C 3 - 8 cycloalkylS(O ) n , (x) phenylS (O) n , (xi) substitute d phenylS(O ) n in which t he substituen ts are X and Y, and (xii) oxo, (c) C 1 - 10 alkylS(O) n , (d) C 3 - 8 cycloalky lS(O) n , (e) ph enylS(O) n , (f) substituted p henylS(O) n in which the su bstituents are X and Y, (g) halogen, (h) hydro xy, (i) C 1 - 10 a lkoxy, (j) C 1 - 5 alkoxycarbo nyl, (k) C 1 - 5 a cyloxy, (l) ph enyl, and (m ) substituted phenyl in which the substituents are X and Y; (7) phenyl; (8 ) substituted phenyl in wh ich the substi tuents are X a nd Y; (9) amino; (10) C 1 - 5 alkyla mino; (11) d i(C 1 - 5 alkyl)am ino; (12) phe nylamino; (13) substituted phenylamino in which the subst ituents are X and Y; (14) p henyl C 1 - 10 a lkylamino; (15 ) substituted phenyl C 1 - 10 a lkylamino in which the substitue nts are X an d Y; (16) a member sele cted from: (a ) piperidinyl, (b) pyrrolidin yl, (c) pipera zinyl, (d) morpholin yl, and (e) th iomorpholinyl ; and (17) R 10 S in which R 10 is selected from (a) C 1 - 10 alkyl, (b) ph enyl, and (c) subst ituted pheny l in which th e substituent s are X and Y; R 4 is:(1) hydrogen; (2 ) C 1 - 10 alkyl ; and (3) substitute d C 1 - 10 alkyl in which one or more su bstituents is selected from (a) halogen, (b) hydroxy , (c) C 1 - 10 alkoxy (d ) C 1 - 5 alkoxy carbonyl, (e) C 1 - 5 alkylacy loxy, (f) phe nylacyloxy, (g ) phenoxycar bonyl, (h) ph enyl C 1 - 5 alkylacylo xy, (i) pheny l C 1 - 5 alkoxy, (j) amino, (k ) C 1 - 5 alkylam ino, (l) di(C 1 - 5 alkyl)amino , (m) phenyla mino, (n) substitute d phenylamin o in which th e substituents are X and Y ; (o) phenyl C 1 - 5 alkylamino , (p) substitut ed phenyl C 1 - 5 alky lamino in whi ch the substi tuents are X and Y, (q) C 3 - 8 cycloalkyl, (r) phenyl, (s ) substituted phenyl in which the substituents are X and Y, (t) phenylS(O ) n , (u) substit uted phenyl S (O) n in which the substitue nts are X and Y, (v ) phenyl C 1 - 5 alkyl S(O) n , (w) C 1 - 5 alkylS (O) n ; (x) ph enylaminoacy loxy, (y) C 1 - 5 alkylaminoac yloxy, (z) C 1 - 5 alky lacylamino, ( aa) di(phenyl C -5 alkyl)phos phonyl (bb) di(C 1 - 5 alkyl)p hosphinyl (4 ) R 4 together with the carbon a tom to which it is attached represents a C 3 - 8 carbocyc lic ring; R 5 an d R 6 indepen dently are H, OH, OR 7 or R 5 and R 6 together with the carb on to which th ey are attach ed represent C=O or R 5 a nd R 6 togethe r with the carbon to which they a re attached re present a ca rbocyclic ring of 3 to 7 atom s; provided t hat when R 5 is H, R 6 is OH or OR 7 , and when R 5 is OH, R 6 is H, a nd when R 5 is OR 7 , R 6 is H ; R 7 is phenylC 1 -3 al kyl. C 1 -5 alkyl ; R 8 and R 9 in dependently are H, C 1 - 3 alkyl, p henylC 1 -3 al kyl or aryl wh erein aryl is phenyl naph thyl, pyridyl, furanyl, thien yl or phenyl, naphthyl,

pyridyl, fu ranyl or thien yl substituted with groups X and Y prov ided that wh en R 7 is R 8 is not H and whe n R

7 is nei ther R 8 nor R 9 is H;X and Y are indep endently sele cted from:(a ) OH, (b) halogen, (c) trifluorom ethyl, (d) C 1 -3 alkoxy, (e) C 1 -3 -al kylcarbonylox y, (f) phenyl lcarbonyloxy, (g) C 1 -3 alkoxyca rbonyl, (h) ph enyloxycarbo nyl, (i) hydrog en; (j) C 1 -5 a lkyl; Z is sel ected from: (1 ) hydrogen; (2) C 1 -5 - alkyl; (3) substituted C 1 -5 alkyl in w hich the subs tituent is sele cted from (a ) phenyl, (b) dimethylamin o, and (c) acetylam ino, and (4) 2,3 hydroxyp ropyl; Halog en is Cl or F; a is a sin gle bond or a double bo nd; or a pharmace utically acce ptable salt the reof. [045] In certain embodiment s, the anti-me thanogenic lo vastatin anal og or derivativ e may be a m etabolite of lovasta tin which po ssess a 2,3,5 ,6,7,8-hexah ydronapthyl m oiety and a 3-hydroxy gr oup, as desc ribed, for example, in U.S. Pat ent No. 4,738,982, the en tire contents of which ar e incorporate d by referen ce herein (including , by way of i llustration, dis closures of s pecific comp ounds and sy nthetic schem es). For exa mple, the anti-meth anogenic lova statin analog or derivative may have the formula:

wherein R is hydroge n, C 1 -5 alkyl , C 1 -5 alkyl s ubstituted wi th a member of the group consisting o f phenyl, dimethyla mino, or ace tylamino, an d pharmaceu tically accep table salts of the Formul a XXIX in w hich R is hydrogen .

[046] In certain embodiment s, the anti-me thanogenic lo vastatin anal og or derivativ e may be a m etabolite of mevin olin and rel ated compou nds which possess a 1 ,2,7,8,8a-pen tahydronapht hyl moiety a nd a 6- exomethy lene group, as described , for exampl e, in U.S. P atent Nos. 4 ,782,084 and 4,885,314, t he entire contents of which are incorporated by reference herein (includ ing, by way of illustration, disclosures o f specific compoun ds and synth etic schemes ). For examp le, the anti-m ethanogenic lovastatin an alog or deriv ative may have the formula:

wherein; R is C 1 - 10 alk yl; R 1 is hydro gen, C 1 - 5 alk yl, C 1 - 5 alkyl s ubstituted wi th a member of the group c onsisting of pheny l, dimethylam ino, or acetyla mino, and ph armaceutical ly acceptable salts of the F ormula XXX I in which R 1 is hyd rogen.

[047] In an em bodiment, the anti-methano genic lovasta tin analog or derivative ma y be a synthe tic analog of lovast astin as des cribed, for ex ample, in U .S. Patent N o. 4,654,363 , the entire c ontents of w hich are incorpora ted by refere nce herein (including, by way of illus tration, discl osures of sp ecific compo unds and synthetic schemes). Fo r example, th e anti-methan ogenic lovas tatin analog o r derivative m ay have the fo rmula:

wherein A is: , C H 2 CH 2 - or -CH=CH-; R 2 and R 4 independ ently are chlo ro, fluoro, or C 1 - 3 alkyl, esp ecially methy l; R 6 is hydrog en or chloro; R 7 is hydrog en, chloro or fluoro provided that when R 6 is ch loro then R 7 i s hydrogen an d when R 6 is hydrogen, R 7 is chloro or f luoro; X is hydrogen , C 1 - 5 alkyl or C 1 - 5 alkyl su bstituted with a member of the group co nsisting of ph enyl, dimethy lamino or acetylam ino; and pha rmaceutically acceptable s alts of the c ompounds of the Formula XXXII in w hich X is hydrogen .

[048] Additiona l synthetic an ti-methanoge nic lovastatin analogs or de rivatives are disclosed, for example, in U.S. P atent Nos. 4 ,198,425, 4,255,444, 4,604 ,472, and EP publication No. 0024348, the entire co ntents of which are incorporated by reference herein (inclu ding, by way of illustration , disclosures of specific co mpounds and synth etic schemes ).

[049] In certain embodiment s, the anti-me thanogenic lo vastatin analo g or derivativ e may have a modified lactone r ing, as desc ribed, for ex ample, in U. S. Patent N o. 6,818,638 , the entire c ontents of w hich are incorpora ted by refere nce herein (including, by way of illus tration, discl osures of sp ecific compo unds and synthetic schemes). Fo r example, th e anti-methan ogenic lovas tatin analog o r derivative m ay have the fo rmula:

XXXIII

wherein, each of a - - - b and α - - - β inde pendently, is either a sin gle bond or a double bo nd; R 1 is wherein R a is H, C1-6a lkyl optionally substituted b y OH or C 1-4 a lkoxy, C 2- 6 alkenyl o r aryl-C 1-4 alk yl; R 2 is OH;—O—CO—R 5 wherein R 5 is C 1-8 alkyl, C 3-7 cycloalkyl, C 3-7 cycloalky l-C 1-4 alkyl, aryl or ar yl-C 1-4 alkyl; o r—O—R 6 wh erein R 6 is the residue of a n α-amino-ac id attached to O through its carbonyl residue o r—CHR 7 —C OR 8 wherein R 7 is H, C 1-4 a lkyl, heteroC 1-4 alkyl, C 3-7 cy cloalkyl, C 3-7 c ycloalkyl-C 1-4 alkyl, aryl or aryl-C 1 -4 alkyl and R 6 is OH, C 1-4 al koxy or NR 9 R 10 wherein eac h of R 9 and R 10 independe ntly is H, C 1-4 a lkyl or R 9 and R 10 f orm together with the nitro gen to which they are bou nd, a hetero aryl group; R 3 is a substitu ted linear

amino alc ohol or cyclic carbamate o f formula (c

2): (c2 )

wherein each of R 50 , independent ly is H; C 1-8 a lkyl; C 3-7 cyclo alkyl; aryl; C 3-7 cycloalkyl- C 1-4 alkyl; heteroary l; heteroaryl- C 1-4 alkyl; C 1- 4 alkylcarbony l; aryl-carbon yl; heteroary l-carbonyl; ar yl-C 1-4 alkyl-ca rbonyl or heteroary l-C 1-4 alkyl-car bonyl, and e ach of R 51 , in dependently is H; C 1-4 alky l; hydroxy-C 1 -4 alkyl; amino -C 1-4 alkyl; C 1-4 alkox y-C 1-4 alkyl; C 1 -4 alkoxy-carbo nyl-C 1-4 alkyl wherein C 1-4 a lkoxy is optio nally substitut ed by amino, C 1-4 alkyl- amino or di-(C 1-4 alkyl)a mino; HOOC—C 1-4 alkyl; o r R 23 R 24 N—C O—C 1-4 alkyl wherein R 23 is H, C 1-4 alkyl, hydroxy- C 1-4 alkyl, polyhydroxy- C 1-8 alkyl, het eroaryl, heter oaryl-C 1-4 alky l, amino-C 1-4 a lkyl, C 1-4 alkyl lamino-C 1-4 alk yl, di-(C 1- 4 alkyl)am ino-C 1-4 alkyl o r aryl-C 1-4 alky l and R 24 is H , C 1-4 alkyl or hydroxy-C 1-4 a lkyl, at least o ne of R 50 and R 51 being other tha n H,each of X and Y is H or X and Y fo rm together e ach of a - - - b and α - - - β being a si ngle bond

when eac h of R

1 3 or R 14 is OH; and R 4 is H or O R 19 wherein R 19 is C 1-6 alky l, hydroxy-C 1- 6 alkyl, C 1- 4 alkoxy-C 1-6 alkyl, aryl- C 1-4 alkyl or C 1-4 alkoxycarb onyl-C 1-4 alky l. and where ver“aryl” ap pears as is or in the significan ces of “aryl- C 1-4 alkyl” in t he above de finition, it is“phenyl” or“naphthyl” op tionally subs tituted by halogen, OH, NR 11 R 12 , COOH, C F 3 , C 1-4 alkox y, C 1-4 alkyl, hydroxy-C 1-4 a lkyl, hydroxy -C 1 alkoxy, C 1-4 alkoxy- carbonyl, cyano or CO NR 11 R 12 , each of R 11 and R 12 independen tly being H, C 1-4 alkyl, phen yl, naphthyl, p henyl-C 1- 4 alkyl or naphthyl-C 1-4 a lkyl or R 11 an d R 12 togethe r with the nitr ogen to which they are bou nd forming h eteroaryl; and wher ever“heteroa ryl” appears, it is a 5- or 6- membered he teroaryl optio nally fused to a benzene ri ng; in free form or in salt form.

[050] In certain embodiment s, the anti-me thanogenic lo vastatin analo g or derivativ e may be a c ompound of the foll owing Formu la:

and pharmaceutically acceptable salts of the compound.

[051] In certain embodiments, the anti-methanogenic lovastatin analog or derivative may be a compound of the following Formula:

wherein X is selected from O, N, and S, and Z is selected from hydrogen, substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted straight or branched C 1-10 alkyl, C 2-10 alkenyl, or C 2-10 alkynyl, substituted or unsubstituted C 3-10 cycloalkyl, substituted or unsubstituted C 5-8 cycloalkenyl, substituted or unsubstituted C 7-20 alkyl, substituted or unsubstituted C 1-10 aminoalkyl, substituted or unsubstituted C 5-10 aryl, substituted or unsubstituted saturated or unsaturated 3-11 member heteroaryl or heteroarylalkyl containing 1, 2, 3, or 4 heteroatoms selected independently from N, O, S, or S(O) 2 or R 2 and R 3 may be combined with the nitrogen to which they are attached to form a 3, 4, 5, 6, or 7 membered heterocyclein which one or more of the carbons may be substituted with a heteratom selected from O, N, or S and in which any of the hydrogens of the heterocycle may be substituted with C 1-6 alkyl, F, Cl, Br, I, cyano, C 1-6 alkoxy, or C 6-10 aryloxy, or a stereoisomer thereof, tautomer thereof, solvate thereof, or a pharmaceutically acceptable salt thereof.

[052] In certain embodiments, the anti-methanogenic lovastatin analog or derivative may be a compound of the following Formula:

wherein X is selected from O, N, and S, and Z is selected from substituted or unsubstituted C 1-6 alkyl, substituted or unsubstituted straight or branched C 1-10 alkyl, C 2-10 alkenyl, or C 2-10 alkynyl, substituted or unsubstituted C 3-10 cycloalkyl, substituted or unsubstituted C 5-8 cycloalkenyl, substituted or unsubstituted C 7-20 alkyl, substituted or unsubstituted C 1-10 aminoalkyl, substituted or unsubstituted C 5-10 aryl, substituted or unsubstituted saturated or unsaturated 3-11 member heteroaryl or heteroarylalkyl containing 1, 2, 3, or 4 heteroatoms selected independently from N, O, S, or S(O) 2 or R 2 and R 3 may be combined with the nitrogen to which they are attached to form a 3, 4, 5, 6, or 7 membered heterocyclein which one or more of the carbons may be substituted with a heteratom selected from O, N, or S and in which any of the hydrogens of the heterocycle may be substituted with C 1-6 alkyl, F, Cl, Br, I, cyano, C 1-6 alkoxy, or C 6-10 aryloxy, or a stereoisomer thereof, tautomer thereof, solvate thereof, or a pharmaceutically acceptable salt thereof.

[053] Except where specifically defined to the contrary, the terms "alkyl", "alkenyl", "acyl" "aryloxy" and "alkoxy" include both the straight chain and branched-chain species of the term.

Modified Release Profile

[054] In one aspect, the present invention provides modified release formulations comprising at least one anti-methanogenic lovastatin analog or derivative, wherein the formulation releases at least about 60% of the anti-methanogenic lovastatin analog or derivative after the stomach and into one or more regions of the intestinal tract.

[055] In various embodiments, the anti-methanogenic lovastatin analog or derivative can inhibit the production of methane, inhibit methanogenesis, or inhibit the growth and/or proliferation of methanogens. In some aspects, the anti-methanogenic lovastatin analog or derivative is in a hydroxyacid form which typically is, without wishing to be bound by theory, an effective inhibitor of HMG-CoA reductase, or in a lactone form which typically is, without wishing to be bound by theory, an ineffective HMG-CoA inhibitor.

[056] In one aspect, the present invention provides modified release formulations comprising at least one anti-methanogenic lovastatin analog or derivative, wherein the formulation releases at least 60% of the anti- methanogenic lovastatin analog or derivative after the stomach into one or more regions of the intestinal tract.

[057] In various embodiments, the anti-methanogenic lovastatin analog or derivative is in the lactone form, including substantially in the lactone form, at the site of delivery by the present formulations. For example, in some embodiments, the amount of GI tract-delivered anti-methanogenic lovastatin analog or derivative which is in the lactone form is more than about 95%, or more than about 90%, or more than about 85%, or more than about 80%, or more than about 75%, or more than about 70%, or more than about 65%, or more than about 60%, or more than about 55%, or more than about 50%, or more than about 25%.

[058] In various embodiments, the modified-release formulations of the present invention are designed for immediate release (e.g. upon ingestion). In various embodiments, the modified-release formulations may have sustained-release profiles, i.e. slow release of the active ingredient(s) in the body (e.g., GI tract) over an extended period of time. In various embodiments, the modified-release formulations may have a delayed-release profile, i.e. not immediately release the active ingredient(s) upon ingestion; rather, postponement of the release of the active ingredient(s) until the composition is lower in the gastrointestinal tract; for example, for release in the small intestine (e.g., one or more of duodenum, jejunum, ileum) or the large intestine (e.g., one or more of cecum, ascending, transverse, descending or sigmoid portions of the colon, and rectum). For example, a composition can be enteric coated to delay release of the active ingredient(s) until it reaches the small intestine or large intestine. In some embodiments, there is not a substantial amount of the active ingredient(s) of the present formulations in the stool.

[059] In various embodiments, the modified-release formulation of the present invention releases (optionally as a first release) at least 60% of the anti-methanogenic lovastatin analog or derivative after the stomach into one or more regions of the intestine. For example, the modified-release formulation releases at least 60%, at least 61%, at least 62%, at least 63%, at least 64%, at least 65%, at least 66%, at least 67%, at least 68%, at least 69%, at least 70%, at least 71%, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% of the anti-methanogenic lovastatin analog or derivative in the intestine.

[060] In various embodiments, the modified-release formulation releases (optionally as a first release) the anti-methanogenic lovastatin analog or derivative in the small intestine. In various embodiments, the modified- release formulation of the present invention releases at least 60% of the anti-methanogenic lovastatin analog or derivative in the small intestine. For example, the modified-release formulation releases at least 60%, at least 61%, at least 62%, at least 63%, at least 64%, at least 65%, at least 66%, at least 67%, at least 68%, at least 69%, at least 70%, at least 71%, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% of the anti- methanogenic lovastatin analog or derivative in the small intestine.

[061] In one embodiment, the formulation releases (optionally as a first release) the anti-methanogenic lovastatin analog or derivative in the duodenum. In various embodiments, the modified-release formulation of the present invention releases at least 60% of the anti-methanogenic lovastatin analog or derivative in the duodenum. For example, the modified-release formulation releases at least 60%, at least 61%, at least 62%, at least 63%, at least 64%, at least 65%, at least 66%, at least 67%, at least 68%, at least 69%, at least 70%, at least 71%, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% of the anti-methanogenic lovastatin analog or derivative in the duodenum. [062] In another embodiment, the formulation releases (optionally as a first release) the anti- methanogenic lovastatin analog or derivative in the jejunum. In various embodiments, the modified-release formulation of the present invention releases at least 60% of the anti-methanogenic lovastatin analog or derivative in the jejunum. For example, the modified-release formulation releases at least 60%, at least 61%, at least 62%, at least 63%, at least 64%, at least 65%, at least 66%, at least 67%, at least 68%, at least 69%, at least 70%, at least 71%, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% of the anti- methanogenic lovastatin analog or derivative in the jejunum.

[063] In a further embodiment, the formulation releases (optionally as a first release) the anti- methanogenic lovastatin analog or derivative in the ileum and/or the ileocecal junction. In various embodiments, the modified-release formulation of the present invention releases at least 60% of the anti-methanogenic lovastatin analog or derivative in the ileum and/or the ileocecal junction. For example, the modified-release formulation releases at least 60%, at least 61%, at least 62%, at least 63%, at least 64%, at least 65%, at least 66%, at least 67%, at least 68%, at least 69%, at least 70%, at least 71%, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% of the anti-methanogenic lovastatin analog or derivative in the ileum and/or the ileocecal junction.

[064] In other embodiments, the modified-release formulation releases (optionally as a first release) the anti-methanogenic lovastatin analog or derivative in the large intestine. In various embodiments, the modified- release formulation of the present invention releases at least 60% of the anti-methanogenic lovastatin analog or derivative in the large intestine. For example, the modified-release formulation releases at least 60%, at least 61%, at least 62%, at least 63%, at least 64%, at least 65%, at least 66%, at least 67%, at least 68%, at least 69%, at least 70%, at least 71%, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% of the anti- methanogenic lovastatin analog or derivative in the large intestine.

[065] In an embodiment, the modified-release formulation releases (optionally as a first release) the anti- methanogenic lovastatin analog or derivative in the cecum. In various embodiments, the modified-release formulation of the present invention releases at least 60% of the anti-methanogenic lovastatin analog or derivative in the cecum. For example, the modified-release formulation releases at least 60%, at least 61%, at least 62%, at least 63%, at least 64%, at least 65%, at least 66%, at least 67%, at least 68%, at least 69%, at least 70%, at least 71%, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% of the anti- methanogenic lovastatin analog or derivative n in the cecum.

[066] In another embodiment, the modified-release formulation releases (optionally as a first release) the anti-methanogenic lovastatin analog or derivative in the ascending colon. In various embodiments, the modified- release formulation of the present invention releases at least 60% of the anti-methanogenic lovastatin analog or derivative in the ascending colon. For example, the modified-release formulation releases at least 60%, at least 61%, at least 62%, at least 63%, at least 64%, at least 65%, at least 66%, at least 67%, at least 68%, at least 69%, at least 70%, at least 71%, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% of the anti- methanogenic lovastatin analog or derivative in the ascending colon.

[067] In yet another embodiment, the anti-methanogenic lovastatin analog or derivative is released (optionally as a first release) in the transverse colon. In various embodiments, the modified-release formulation of the present invention releases at least 60% of the anti-methanogenic lovastatin analog or derivative in the transverse colon. For example, the modified-release formulation releases at least 60%, at least 61%, at least 62%, at least 63%, at least 64%, at least 65%, at least 66%, at least 67%, at least 68%, at least 69%, at least 70%, at least 71%, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% of the anti-methanogenic lovastatin analog or derivative in the transverse colon.

[068] In a further embodiment, the anti-methanogenic lovastatin analog or derivative is released (optionally as a first release) in the descending colon. In various embodiments, the modified-release formulation of the present invention releases at least 60% of the anti-methanogenic lovastatin analog or derivative in the descending colon. For example, the modified-release formulation releases at least 60%, at least 61%, at least 62%, at least 63%, at least 64%, at least 65%, at least 66%, at least 67%, at least 68%, at least 69%, at least 70%, at least 71%, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% of the anti-methanogenic lovastatin analog or derivative in the descending colon. [069] In another embodiment, the anti-methanogenic lovastatin analog or derivative is released (optionally as a first release) in the sigmoid colon. In various embodiments, the modified-release formulation of the present invention releases at least 60% of the anti-methanogenic lovastatin analog or derivative in the sigmoid colon. For example, the modified-release formulation releases at least 60%, at least 61%, at least 62%, at least 63%, at least 64%, at least 65%, at least 66%, at least 67%, at least 68%, at least 69%, at least 70%, at least 71%, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% of the anti-methanogenic lovastatin analog or derivative in the sigmoid colon.

[070] In certain embodiments, the modified-release formulation does not substantially release the anti- methanogenic lovastatin analog or derivative in the stomach.

[071] In certain embodiments, the modified-release formulation releases the anti-methanogenic lovastatin analog or derivative at a specific pH. For example, in some embodiments, the modified-release formulation is substantially stable in an acidic environment and substantially unstable (e.g., dissolves rapidly or is physically unstable) in a near neutral to alkaline environment. In some embodiments, stability is indicative of not substantially releasing while instability is indicative of substantially releasing. For example, in some embodiments, the modified-release formulation is substantially stable at a pH of about 7.0 or less, or about 6.5 or less, or about 6.0 or less, or about 5.5 or less, or about 5.0 or less, or about 4.5 or less, or about 4.0 or less, or about 3.5 or less, or about 3.0 or less, or about 2.5 or less, or about 2.0 or less, or about 1.5 or less, or about 1.0 or less. In some embodiments, the present formulations are stable in lower pH areas and therefore do not substantially release in, for example, the stomach. In some embodiments, modified-release formulation is substantially stable at a pH of about 1 to about 4 or lower and substantially unstable at pH values that are greater. In these embodiments, the modified-release formulation is not substantially released in the stomach. In these embodiments, the modified-release formulation is substantially released in the small intestine (e.g. one or more of the duodenum, jejunum, and ileum) and/or large intestine (e.g. one or more of the cecum, ascending colon, transverse colon, descending colon, and sigmoid colon). In some embodiments, modified-release formulation is substantially stable at a pH of about 4 to about 5 or lower and consequentially is substantially unstable at pH values that are greater and therefore is not substantially released in the stomach and/or small intestine (e.g. one or more of the duodenum, jejunum, and ileum). In these embodiments, the modified-release formulation is substantially released in the large intestine (e.g. one or more of the cecum, ascending colon, transverse colon, descending colon, and sigmoid colon). In various embodiments, the pH values recited herein may be adjusted as known in the art to account for the state of the subject, e.g. whether in a fasting or postprandial state.

[072] In some embodiments, the modified-release formulation is substantially stable in gastric fluid and substantially unstable in intestinal fluid and, accordingly, is substantially released in the small intestine (e.g. one or more of the duodenum, jejunum, and ileum) and/or large intestine (e.g. one or more of the cecum, ascending colon, transverse colon, descending colon, and sigmoid colon).

[073] In some embodiments, the modified-release formulation is stable in gastric fluid or stable in acidic environments. These modified-release formulations release about 30% or less by weight of the anti- methanogenic lovastatin analog or derivative and/or additional therapeutic agent in the modified-release formulation in gastric fluid with a pH of about 4 to about 5 or less, or simulated gastric fluid with a pH of about 4 to about 5 or less, in about 15, or about 30, or about 45, or about 60, or about 90 minutes. Modified-release formulations of the of the invention may release from about 0% to about 30%, from about 0% to about 25%, from about 0% to about 20%, from about 0% to about 15%, from about 0% to about 10%, about 5% to about 30%, from about 5% to about 25%, from about 5% to about 20%, from about 5% to about 15%, from about 5% to about 10% by weight of the anti-methanogenic lovastatin analog or derivative and/or additional therapeutic agent in the modified-release formulation in gastric fluid with a pH of 4-5, or less or simulated gastric fluid with a pH of 4-5 or less, in about 15, or about 30, or about 45, or about 60, or about 90 minutes. Modified-release formulations of the invention may release about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10% by weight of the total anti-methanogenic lovastatin analog or derivative and/or additional therapeutic agent in the modified-release formulation in gastric fluid with a pH of 5 or less, or simulated gastric fluid with a pH of 5 or less, in about 15, or about 30, or about 45, or about 60, or about 90 minutes.

[074] In some embodiments, the modified-release formulation is unstable in intestinal fluid. These modified-release formulations release about 70% or more by weight of the anti-methanogenic lovastatin analog or derivative and/or additional therapeutic agent in the modified-release formulation in intestinal fluid or simulated intestinal fluid in about 15, or about 30, or about 45, or about 60, or about 90 minutes. In some embodiments, the modified-release formulation is unstable in near neutral to alkaline environments. These modified-release formulations release about 70% or more by weight of the anti-methanogenic lovastatin analog or derivative and/or additional therapeutic agent in the modified-release formulation in intestinal fluid with a pH of about 4-5 or greater, or simulated intestinal fluid with a pH of about 4-5 or greater, in about 15, or about 30, or about 45, or about 60, or about 90 minutes. A modified-release formulation that is unstable in near neutral or alkaline environments may release 70% or more by weight of anti-methanogenic lovastatin analog or derivative and/or additional therapeutic agent in the modified-release formulation in a fluid having a pH greater than about 5 (e.g., a fluid having a pH of from about 5 to about 14, from about 6 to about 14, from about 7 to about 14, from about 8 to about 14, from about 9 to about 14, from about 10 to about 14, or from about 11 to about 14) in from about 5 minutes to about 90 minutes, or from about 10 minutes to about 90 minutes, or from about 15 minutes to about 90 minutes, or from about 20 minutes to about 90 minutes, or from about 25 minutes to about 90 minutes, or from about 30 minutes to about 90 minutes, or from about 5 minutes to about 60 minutes, or from about 10 minutes to about 60 minutes, or from about 15 minutes to about 60 minutes, or from about 20 minutes to about 60 minutes, or from about 25 minutes to about 90 minutes, or from about 30 minutes to about 60 minutes. [075] In one embodiment, the modified-release formulation may remain essentially intact, or may be essentially insoluble, in gastric fluid. The stability of the delayed-release coating can be pH dependent. Delayed- release coatings that are pH dependent will be substantially stable in acidic environments (pH of about 5 or less), and substantially unstable in near neutral to alkaline environments (pH greater than about 5). For example, the delayed-release coating may essentially disintegrate or dissolve in near neutral to alkaline environments such as are found in the small intestine (e.g. one or more of the duodenum, jejunum, and ileum) and/or large intestine (e.g. one or more of the cecum, ascending colon, transverse colon, descending colon, and sigmoid colon).

[076] Examples of simulated gastric fluid and simulated intestinal fluid include, but are not limited to, those disclosed in the 2005 Pharmacopeia 23NF/28USP in Test Solutions at page 2858 and/or other simulated gastric fluids and simulated intestinal fluids known to those of skill in the art, for example, simulated gastric fluid and/or intestinal fluid prepared without enzymes.

[077] Alternatively, the stability of the modified-release formulation can be enzyme-dependent. Delayed- release coatings that are enzyme dependent will be substantially stable in fluid that does not contain a particular enzyme and substantially unstable in fluid containing the enzyme. The delayed-release coating will essentially disintegrate or dissolve in fluid containing the appropriate enzyme. Enzyme-dependent control can be brought about, for example, by using materials which release the active ingredient only on exposure to enzymes in the intestine, such as galactomannans. Also, the stability of the modified-release formulation can be dependent on enzyme stability in the presence of a microbial enzyme present in the gut flora.

[078] In some embodiments, a dual pulse formulation is provided. In various embodiments, the present invention provides for modified-release formulations that release multiple doses of the anti-methanogenic lovastatin analog or derivative, at different locations along the intestines, at different times, and/or at different pH. In an illustrative embodiment, the modified-release formulation comprises a first dose of the anti-methanogenic lovastatin analog or derivative and a second dose of the anti-methanogenic lovastatin analog or derivative, wherein the first dose and the second dose are released at different locations along the intestines, at different times, and/or at different pH. For example, the first dose is released at the duodenum, and the second dose is released at the ileum. In another example, the first dose is released at the jejunum, and the second dose is released at the ileum. In other embodiments, the first dose is released at a location along the small intestine (e.g., the duodenum), while the second dose is released along the large intestine (e.g., the ascending colon). In various embodiments, the modified-release formulation may release at least one dose, at least two doses, at least three doses, at least four doses, at least five doses, at least six doses, at least seven doses, or at least eight doses of the anti-methanogenic lovastatin analog or derivative at different locations along the intestines, at different times, and/or at different pH. Each individual dose may comprise the same lovastatin analog or derivative or may comprise different lovastatin analog or derivative.

[079] In some embodiments, the dual pulse formulation is an enteric-coated capsule comprising beads that comprise an anti-methanogenic lovastatin analog or derivative and optionally an additional therapeutic agent. In some embodiments, the enteric-coated capsule dissolves in a first area of GI tract to release the beads and/or a first population of beads releases in a second area of the GI tract (and that is not the same as the first area of the GI tract) and a second population of beads releases in a third area of the GI tract (and that is not the same as the first or second areas of the GI tract). In some embodiments, the dose/release ratio (e.g. how much agent is released in various locations) can be tuned as needed. In some embodiments, the enteric-coated capsule dissolves in the duodenum to release the beads and/or a first population of beads releases in the duodenum and/or a second population of beads releases in the ileocecal junction (see, e.g. Figures 1-4).

Modified Release Formulation and Dosage Forms

[080] The modified-release formulation of the present invention may further comprise a pharmaceutically acceptable carrier or excipient. As one skilled in the art will recognize, the formulations can be in any suitable form appropriate for the desired use and route of administration. Examples of suitable dosage forms include, for example, oral and parenteral dosage forms.

[081] Suitable dosage forms for oral use include, for example, solid dosage forms such as tablets, dispersible powders, granules, and capsules. In one embodiment, the modified-release formulation is in the form of a tablet. In another embodiment, the modified-release formulation is in the form of a capsule. In yet another embodiment, the modified-release formulation is in the form of a soft-gel capsule. In a further embodiment, the modified-release formulation is in the form of a gelatin capsule.

[082] In such dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate, dicalcium phosphate, etc., and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, silicic acid, microcrystalline cellulose, and Bakers Special Sugar, etc., b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, acacia, polyvinyl alcohol, polyvinylpyrrolidone, methylcellulose, hydroxypropyl cellulose, and hydroxymethyl cellulose etc., c) humectants such as glycerol, etc., d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, sodium carbonate, cross-linked polymers such as crospovidone (cross-linked polyvinylpyrrolidone), croscarmellose sodium (cross-linked sodium carboxymethylcellulose), sodium starch glycolate, etc., e) solution retarding agents such as paraffin, etc., f) absorption accelerators such as quaternary ammonium compounds, etc., g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, etc., h) absorbents such as kaolin and bentonite clay, etc., and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, glyceryl behenate, etc., and mixtures of such excipients. One of skill in the art will recognize that particular excipients may have two or more functions in the oral dosage form. In the case of an oral dosage form, for example, a capsule or a tablet, the dosage form may also comprise buffering agents.

[083] The modified release formulation can additionally include a surface active agent. Surface active agents suitable for use in the present invention include, but are not limited to, any pharmaceutically acceptable, non-toxic surfactant. Classes of surfactants suitable for use in the compositions of the invention include, but are not limited to polyethoxylated fatty acids, PEG-fatty acid diesters, PEG-fatty acid mono- and di-ester mixtures, polyethylene glycol glycerol fatty acid esters, alcohol-oil transesterification products, polyglycerized fatty acids, propylene glycol fatty acid esters, mixtures of propylene glycol esters-glycerol esters, mono- and diglycerides, sterol and sterol derivatives, polyethylene glycol sorbitan fatty acid esters, polyethylene glycol alkyl ethers, sugar esters, polyethylene glycol alkyl phenols, polyoxyethylene-olyoxypropylene block copolymers, sorbitan fatty acid esters, lower alcohol fatty acid esters, ionic surfactants, and mixtures thereof. In some embodiments, compositions of the invention may comprise one or more surfactants including, but not limited to, sodium lauryl sulfate, polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80, and triethyl citrate.

[084] The modified-release formulation can also contain pharmaceutically acceptable plasticizers to obtain the desired mechanical properties such as flexibility and hardness. Such plasticizers include, but are not limited to, triacetin, citric acid esters, phthalic acid esters, dibutyl sebacate, cetyl alcohol, polyethylene glycols, polysorbates or other plasticizers.

[085] The modified-release formulation can also include one or more application solvents. Some of the more common solvents that can be used to apply, for example, a delayed-release coating composition include isopropyl alcohol, acetone, methylene chloride and the like.

[086] The modified-release formulation can also include one or more alkaline materials. Alkaline material suitable for use in compositions of the invention include, but are not limited to, sodium, potassium, calcium, magnesium and aluminum salts of acids such as phosphoric acid, carbonic acid, citric acid and other aluminum/magnesium compounds. In addition the alkaline material may be selected from antacid materials such as aluminum hydroxides, calcium hydroxides, magnesium hydroxides and magnesium oxide.

[087] The solid oral dosage forms can be prepared by any conventional method known in the art, for example granulation (e.g., wet or dry granulation) of the active compound (e.g., statins) with one or more suitable excipients. Alternatively, the active compound can be layered onto an inert core (e.g., a nonpareil/sugar sphere or silica sphere) using conventional methods such as fluidized bed or pan coating, or extruded and spheronized using methods known in the art, into active compound-containing beads. Such beads can then be incorporated into tablets or capsules using conventional methods.

[088] Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethyl formamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, etc., and mixtures thereof.

[089] Besides inert diluents, the oral compositions can also include adjuvants such as sweetening, flavoring, and perfuming agents. [090] Suspensions, in addition to the active compounds, may contain suspending agents such as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, tragacanth, etc., and mixtures thereof.

[091] The formulations comprising the therapeutic agents of the present invention may conveniently be presented in unit dosage forms and may be prepared by any of the methods well known in the art of pharmacy. Such methods generally include the step of bringing the therapeutic agents into association with a carrier, which constitutes one or more accessory ingredients. Typically, the formulations are prepared by uniformly and intimately bringing the therapeutic agent into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product into dosage forms of the desired formulation (e.g., wet or dry granulation, powder blends, etc., followed by tableting using conventional methods known in the art).

[092] In various embodiments, the modified-release formulation of the present invention may utilize one or more modified-release coatings such as delayed-release coatings to provide for effective, delayed yet substantial delivery of the anti-methanogenic lovastatin analog or derivative to the GI tract together with, optionally, other therapeutic agents.

[093] In one embodiment, the delayed-release coating includes an enteric agent that is substantially stable in acidic environments and substantially unstable in near neutral to alkaline environments. In an embodiment, the delayed-release coating contains an enteric agent that is substantially stable in gastric fluid. The enteric agent can be selected from, for example, solutions or dispersions of methacrylic acid copolymers, cellulose acetate phthalate, hydroxypropylmethyl cellulose phthalate, polyvinyl acetate phthalate, carboxymethylethylcellulose, and EUDRAGIT®-type polymer (poly(methacrylic acid, methylmethacrylate), hydroxypropyl methylcellulose acetate succinate, cellulose acetate trimellitate, shellac or other suitable enteric coating polymers. The EUDRAGIT®-type polymer include, for example, EUDRAGIT® FS 30D, L 30 D-55, L 100- 55, L 100, L 12,5, L 12,5 P, RL 30 D, RL PO, RL 100, RL 12,5, RS 30 D, RS PO, RS 100, RS 12,5, NE 30 D, NE 40 D, NM 30 D, S 100, S 12,5, and S 12,5 P. In some embodiments, one or more of EUDRAGIT® FS 30D, L 30 D-55, L 100-55, L 100, L 12,5, L 12,5 P RL 30 D, RL PO, RL 100, RL 12,5, RS 30 D, RS PO, RS 100, RS 12,5, NE 30 D, NE 40 D, NM 30 D, S 100, S 12,5 and S 12,5 P is used. The enteric agent may be a combination of the foregoing solutions or dispersions.

[094] In another embodiment, the delayed-release coating may degrade as a function of time when in aqueous solution without regard to the pH and/or presence of enzymes in the solution. Such a coating may comprise a water insoluble polymer. Its solubility in aqueous solution is therefore independent of the pH. The term“pH independent” as used herein means that the water permeability of the polymer and its ability to release pharmaceutical ingredients is not a function of pH and/or is only very slightly dependent on pH. Such coatings may be used to prepare, for example, sustained release formulations. Suitable water insoluble polymers include pharmaceutically acceptable non-toxic polymers that are substantially insoluble in aqueous media, e.g., water, independent of the pH of the solution. Suitable polymers include, but are not limited to, cellulose ethers, cellulose esters, or cellulose ether-esters, i.e., a cellulose derivative in which some of the hydroxy groups on the cellulose skeleton are substituted with alkyl groups and some are modified with alkanoyl groups. Examples include ethyl cellulose, acetyl cellulose, nitrocellulose, and the like. Other examples of insoluble polymers include, but are not limited to, lacquer, and acrylic and/or methacrylic ester polymers, polymers or copolymers of acrylate or methacrylate having a low quaternary ammonium content, or mixture thereof and the like. Other examples of insoluble polymers include EUDRAGIT RS®, EUDRAGIT RL®, and EUDRAGIT NE®. Insoluble polymers useful in the present invention include polyvinyl esters, polyvinyl acetals, polyacrylic acid esters, butadiene styrene copolymers, and the like. In one embodiment, colonic delivery is achieved by use of a slowly-eroding wax plug (e.g., various PEGS, including for example, PEG6000).

[095] In a further embodiment, the delayed-release coating may be degraded by a microbial enzyme present in the gut flora. In one embodiment, the delayed-release coating may be degraded by a bacteria present in the small intestine. In another embodiment, the delayed-release coating may be degraded by a bacteria present in the large intestine.

[096] The present invention provides for modified-release formulations that release multiple doses of the anti-methanogenic lovastatin analog or derivative along the gastrointestinal tract. The overall release profile of such a formulation may be adjusted by utilizing, for example, multiple particle types or multiple layers. In one embodiment, the first dose of the anti-methanogenic lovastatin analog or derivative may be formulated for release in, for example, the duodenum, whereas the second dose is formulated for delayed release in, for example, the ileum. In another embodiment, the first dose of the anti-methanogenic lovastatin analog or derivative may be formulated for release in, for example, the small intestines, whereas the second dose is formulated for delayed release in, for example, the large intestines. Alternatively, multiple doses are released at different locations alone the intestine.

[097] In one embodiment, one or more doses of the anti-methanogenic lovastatin analog or derivative may be encapsulated in a core particle, for example, in the form of a microbead. For example, the first dose of the anti-methanogenic lovastatin analog or derivative may be encapsulated in a core particle coated with a modified-release coating designed for release at a first location along the intestinal tract, and the second dose of the anti-methanogenic lovastatin analog or derivative may be encapsulated in a core particle coated with a modified-release coating designed for release at a second location along the intestinal tract. The formulation may comprise a plurality of such modified-release particles. For example, the formulation is in the form of capsules comprising multiple microbeads. In such an embodiment, a combination of microbeads may be utilized in which each microbead is designed to release at a specific time point or location. In an alternative embodiment, the formulation is formulated as a capsule within a capsule, with each capsule having different time- or pH- dependent release properties.

[098] In another embodiment, one or more doses of the anti-methanogenic lovastatin analog or derivative may be encapsulated in a layer. For example, the first dose of the anti-methanogenic lovastatin analog or derivative may be encapsulated in a layer coated with a modified-release coating designed for release at a first location along the intestinal tract, and the second dose of the anti-methanogenic lovastatin analog or derivative may be encapsulated in a layer coated with a modified-release coating designed for release at a second location along the intestinal tract. The formulation may comprise a plurality of such modified-release layers. For example, the formulation is in the form of multi-layered tablet or a multi-layered capsule. Each layer may have different time- or pH-dependent release properties.

[099] In the above embodiments, the coated particles or layers with the delayed-release coating may be further covered with an overcoat layer. The overcoat layer can be applied as described for the other coating compositions. The overcoat materials are pharmaceutically acceptable compounds such as sugar, polyethylene glycol, polyvinylpyrrolidone, polyvinyl alcohol, polyvinyl acetate, hydroxypropyl cellulose, methylcellulose, ethylcellulose, hydroxypropyl methylcellulose, carboxymethylcellulose sodium and others, used alone or in mixtures. The overcoat materials can prevent potential agglomeration of particles coated with the delayed- release coating, protect the delayed-release coating from cracking during the compaction process or enhance the tableting process.

[0100] Furthermore, in various embodiments, the agents described herein may be in the form of a pharmaceutically acceptable salt, namely those salts which are suitable for use in contact with the tissues of humans and other animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. The salts can be prepared in situ during the final isolation and purification of the therapeutic agents, or separately by reacting the free base function with a suitable acid or a free acid functionality with an appropriate alkaline moiety. Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphersulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2- hydroxyethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate, valerate salts, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. [0101] In various embodiments, the formulation comprises at least one microbead or mini-tablet. In some embodiments, each microbead or mini-tablet comprises about 5-20% by weight the lovastatin analog or derivative (including without limitation lovastatin diol lactone). For example, the lovastatin analog or derivative (including without limitation lovastatin diol lactone) may be present at about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, or about 20% by weight. In some embodiments, each microbead or mini-tablet may further comprise about 50-70% by weight tablet diluent (e.g., about 50%, about 51%, about 52%, about 53%, about 54%, about 55%, about 56%, about 57%, about 58%, about 59%, about 60%, about 61%, about 62%, about 63%, about 64%, or about 65%, or about 66%, about 67%, or about 68%, or about 69%, or about 70%). In some embodiments, each microbead or mini-tablet may further comprise about 1-10% by weight tablet binder (e.g., about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10%). In some embodiments, each microbead or mini-tablet may further comprise about 0.1-3.0% by weight viscosity and dispersion agent (e.g., about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.1%, about 1.2%, about 1.3%, about 1.4%, about 1.5%, about 1.6%, about 1.7%, about 1.8%, about 1.9%, about 2%, about 2.1%, about 2.2%, about 2.3%, about 2.4%, about 2.5%, about 2.6%, about 2.7%, about 2.8%, about 2.9%, or about 3.0%). In some embodiments, each microbead or mini-tablet may further comprise about 0.1-3.0% by weight lubricant, for example, to facilitate tableting (e.g., about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.1%, about 1.2%, about 1.3%, about 1.4%, about 1.5%, about 1.6%, about 1.7%, about 1.8%, about 1.9%, about 2%, about 2.1%, about 2.2%, about 2.3%, about 2.4%, about 2.5%, about 2.6%, about 2.7%, about 2.8%, about 2.9%, or about 3.0%). In some embodiments, each microbead or mini-tablet may further comprise about 1-10% by weight tablet disintegrant (e.g., about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10%). In some embodiments, each microbead or mini-tablet may further comprise about 10-20% by weight an enteric polymer that dissolves at a pH of either about 5.5 or about 7.0 (e.g., about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, or about 20%). [0102] In various embodiments, the formulation comprises one or more of, or two or more of, or three or more of, or four or more of, or five or more of, or all of an lovastatin analog or derivative (including without limitation lovastatin diol lactone), the lovastatin analog or derivative (including without limitation lovastatin diol lactone) optionally being in two doses; microcrystalline cellulose (e.g. Avicel PH102); copovidone (e.g. Kollidon VA64 Fine); silicon dioxide (e.g. Aerosil 200); magnesium stearate; crospovidone (e.g. Kollidon CL or Kollidon CL-F); where the first dose of at least one lovastatin analog or derivative (including without limitation lovastatin diol lactone) is encapsulated by an enteric polymer that dissolves at a pH of about 5.5 (e.g. EUDRAGIT L 30 D- 55 + PlasACRYL HTP20); and the second dose of at least one lovastatin analog or derivative (including without limitation lovastatin diol lactone) is encapsulated by an enteric polymer that dissolves a at pH of about 7.0 (e.g. EUDRAGIT FS 30 D + PlasACRYL T20 and/or EUDRAGIT® S 100). [0103] In various embodiments, the formulation comprises at least one microbead or mini-tablet. Each microbead or mini-tablet comprises about 5-20% by weight of the lovastatin analog or derivative (including without limitation lovastatin diol lactone). For example, the lovastatin analog or derivative (including without limitation lovastatin diol lactone) may be present at about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, or about 20% by weight. In some embodiments, each microbead or mini-tablet may further comprise about 50-70% by weight microcrystalline cellulose (e.g. Avicel PH102). For example, the microcrystalline cellulose may be present at about 50%, about 51%, about 52%, about 53%, about 54%, about 55%, about 56%, about 57%, about 58%, about 59%, about 60%, about 61%, about 62%, about 63%, about 64%, or about 65%, or about 66%, about 67%, or about 68%, or about 69%, or about 70% by weight. In some embodiments, each microbead or mini-tablet may further comprise about 1-10% by weight copovidone (e.g. Kollidon VA64 Fine). For example, the copovidone may be present at about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10% by weight. In some embodiments, each microbead or mini-tablet may further comprise about 0.1-3.0% by weight silicon dioxide (e.g. Aerosil 200). For example, the silicon dioxide may be present at about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.1%, about 1.2%, about 1.3%, about 1.4%, about 1.5%, about 1.6%, about 1.7%, about 1.8%, about 1.9%, about 2%, about 2.1%, about 2.2%, about 2.3%, about 2.4%, about 2.5%, about 2.6%, about 2.7%, about 2.8%, about 2.9%, or about 3.0% by weight. In some embodiments, each microbead or mini- tablet may further comprise about 0.1-3.0% by weight magnesium stearate (for example, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.1%, about 1.2%, about 1.3%, about 1.4%, about 1.5%, about 1.6%, about 1.7%, about 1.8%, about 1.9%, about 2%, about 2.1%, about 2.2%, about 2.3%, about 2.4%, about 2.5%, about 2.6%, about 2.7%, about 2.8%, about 2.9%, or about 3.0%). In some embodiments, each microbead or mini-tablet may further comprise about 1- 10% by weight crospovidone (e.g. Kollidon CL or Kollidon CL-F). For example, the crospovidone may be present at about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10% by weight. In some embodiments, each microbead or mini-tablet may further comprise about 10-20% by weight an enteric polymer that dissolves at a pH of about 5.5 (e.g. EUDRAGIT L 30 D-55 + PlasACRYL HTP20) or about 7.0 (e.g. EUDRAGIT FS 30 D + PlasACRYL T20 and/or EUDRAGIT® S 100). For example, the enteric polymer may be about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, or about 20% by weight. [0104] In some embodiments, the formulation comprises at least one microbead or mini-tablet with each microbead or mini-tablet comprising about 12% by weight the lovastatin analog or derivative (including without limitation lovastatin diol lactone); about 60% by weight microcrystalline cellulose (e.g. Avicel PH102); about 6% by weight copovidone (e.g. Kollidon VA64 Fine); about 2% by weight silicon dioxide (e.g. Aerosil 200); about 1% by weight magnesium stearate; about 5% by weight crospovidone (e.g. Kollidon CL or Kollidon CL-F); and about 15% by weight an enteric polymer that dissolves at a pH of about 5.5 (e.g. EUDRAGIT L 30 D-55 + PlasACRYL HTP20) or about 7.0 (e.g. EUDRAGIT FS 30 D + PlasACRYL T20 and/or EUDRAGIT® S 100). [0105] In some embodiments, the formulation comprises at least one microbead or mini-tablet with each microbead or mini-tablet comprising about 12.2% by weight lovastatin diol lactone; about 60.9% by weight microcrystalline cellulose (Avicel PH102); about 6.1% by weight copovidone (Kollidon VA64 Fine); about 1.7% by weight silicon dioxide (Aerosil 200); about 0.9% by weight magnesium stearate; about 5.2% by weight crospovidone (Kollidon CL-F); and either about 13.0% by weight of EUDRAGIT L 30 D-55 + PlasACRYL HTP20 coating (which dissolves at a pH of about 5.5) or 13% by weight of EUDRAGIT FS 30 D + PlasACRYL T20 coating (which dissolves at a pH of about 7.0). [0106] In various embodiments, the present formulation comprise a mini-tablet enteric coating thickness, e.g. EUDRAGIT, e.g. EUDRAGIT L 30 D-55 or EUDRAGIT FS 30 D, of greater than about 10%, about 13%, about 15%, or about 17%, or about 20%, or about 25%. [0107] In various embodiments, the formulation of the present invention may comprise at least one mini- tablet that releases at a first pH (e.g. pH of about 5.5) and at least one mini-tablet that releases at a second pH (e.g., pH of about 7.0) at a ratio of 1:2. In such embodiments, the formulation may comprise about 5-20% by weight the lovastatin analog or derivative (including without limitation lovastatin diol lactone). For example, the lovastatin analog or derivative (including without limitation lovastatin diol lactone) may be present at about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, or about 20% by weight of the entire formulation. In some embodiments, the formulation may further comprise about 30-60% by weight tablet diluent (e.g., about 30%, about 31%, about 32%, about 33%, about 34%, about 35%, about 36%, about 37%, about 38%, about 39%, about 40%, about 41%, about 42%, about 43%, about 44%, about 45%, about 46%, about 47%, about 48%, about 49%, about 50%, about 51%, about 52%, about 53%, about 54%, about 55%, about 56%, about 57%, about 58%, about 59%, or about 60%). In some embodiments, the formulation may further comprise about 1- 10% by weight tablet binder (e.g., about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10%). In some embodiments, the formulation may further comprise about 0.1- 3.0% by weight viscosity and dispersion agent (e.g., about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.1%, about 1.2%, about 1.3%, about 1.4%, about 1.5%, about 1.6%, about 1.7%, about 1.8%, about 1.9%, about 2%, about 2.1%, about 2.2%, about 2.3%, about 2.4%, about 2.5%, about 2.6%, about 2.7%, about 2.8%, about 2.9%, or about 3.0%). In some embodiments, the formulation may further comprise about 0.1-3.0% by weight lubricant, for example, to facilitate tableting (e.g., about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.1%, about 1.2%, about 1.3%, about 1.4%, about 1.5%, about 1.6%, about 1.7%, about 1.8%, about 1.9%, about 2%, about 2.1%, about 2.2%, about 2.3%, about 2.4%, about 2.5%, about 2.6%, about 2.7%, about 2.8%, about 2.9%, or about 3.0%). In some embodiments, the formulation may further comprise about 1-10% by weight tablet disintegrant (e.g., about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10%). In some embodiments, the formulation may further comprise about 0.5-10% by weight an enteric polymer that dissolves at a pH of about 5.5 (e.g. EUDRAGIT L 30 D-55 + PlasACRYL HTP20). For example, the enteric polymer that dissolves at a pH of about 5.5 may be present in the formulation at about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10% by weight. In some embodiments, the formulation may further comprise about 1-15% by weight an enteric polymer that dissolves at a pH of about 7.0. (e.g. EUDRAGIT FS 30 D + PlasACRYL T20 and/or EUDRAGIT® S 100). For example, the enteric polymer that dissolves at a pH of about 7.0 may be present in the formulation at about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, or about 15% by weight. In such embodiments, the lovastatin analog or derivative (including without limitation lovastatin diol lactone) may be released in two doses. The first dose of lovastatin analog or derivative (including without limitation lovastatin diol lactone) is encapsulated by the enteric polymer that dissolves at a pH of about 5.5; and the second dose of lovastatin analog or derivative (including without limitation lovastatin diol lactone) is encapsulated by the enteric polymer that dissolves a at pH of about 7.0. [0108] For example, the formulation may comprise at least one mini-tablet that releases at a first pH (e.g. pH of about 5.5) and at least one mini-tablet that releases at a second pH (e.g., pH of about 7.0) at a ratio of 1:2. The formulation may comprise about 9% by weight the lovastatin analog or derivative (including without limitation lovastatin diol lactone); about 42% by weight microcrystalline cellulose (e.g. Avicel PH102); about 4% by weight copovidone (e.g. Kollidon VA64 Fine); about 1% by weight silicon dioxide (e.g. Aerosil 200); about 0.5% by weight magnesium stearate; about 4% by weight crospovidone (e.g. Kollidon CL or Kollidon CL-F); about 3% by weight an enteric polymer that dissolves at a pH of about 5.5 (e.g. EUDRAGIT L 30 D-55 + PlasACRYL HTP20); and about 6% by weight an enteric polymer that dissolves at a pH of about 7.0 (e.g. EUDRAGIT FS 30 D + PlasACRYL T20 and/or EUDRAGIT® S 100). [0109] In another example, the formulation may comprise at least one mini-tablet that releases at a first pH (e.g. pH of about 5.5) and at least one mini-tablet that releases at a second pH (e.g., pH of about 7.0) at a ratio of 1:2. The formulation may comprise about 8.5% by weight the lovastatin analog or derivative (including without limitation lovastatin diol lactone); about 42.4% by weight microcrystalline cellulose (e.g. Avicel PH102); about 4.2% by weight copovidone (e.g. Kollidon VA64 Fine); about 1.2% by weight silicon dioxide (e.g. Aerosil 200); about 0.6% by weight magnesium stearate; about 3.6% by weight crospovidone (e.g. Kollidon CL or Kollidon CL- F); about 3% by weight an enteric polymer that dissolves at a pH of about 5.5 (e.g. EUDRAGIT L 30 D-55 + PlasACRYL HTP20); and about 6.1% by weight an enteric polymer that dissolves at a pH of about 7.0 (e.g. EUDRAGIT FS 30 D + PlasACRYL T20 and/or EUDRAGIT® S 100). [0110] In another embodiment, the formulation of the present invention may at least one mini-tablet that releases at a first pH (e.g. pH of about 5.5) and at least one mini-tablet that releases at a second pH (e.g., pH of about 7.0) at a ratio of 1:5. In such embodiments, the formulation may comprise about 5-20% by weight the lovastatin analog or derivative (including without limitation lovastatin diol lactone). For example, the lovastatin analog or derivative (including without limitation lovastatin diol lactone) may be present at about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, or about 20% by weight of the entire formulation. In some embodiments, the formulation may further comprise about 30-60% by weight tablet diluent (e.g., about 30%, about 31%, about 32%, about 33%, about 34%, about 35%, about 36%, about 37%, about 38%, about 39%, about 40%, about 41%, about 42%, about 43%, about 44%, about 45%, about 46%, about 47%, about 48%, about 49%, about 50%, about 51%, about 52%, about 53%, about 54%, about 55%, about 56%, about 57%, about 58%, about 59%, or about 60%). In some embodiments, the formulation may further comprise about 1- 10% by weight tablet binder (e.g., about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10%). In some embodiments, the formulation may further comprise about 0.1- 3.0% by weight viscosity and dispersion agent (e.g., about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.1%, about 1.2%, about 1.3%, about 1.4%, about 1.5%, about 1.6%, about 1.7%, about 1.8%, about 1.9%, about 2%, about 2.1%, about 2.2%, about 2.3%, about 2.4%, about 2.5%, about 2.6%, about 2.7%, about 2.8%, about 2.9%, or about 3.0%). In some embodiments, the formulation may further comprise about 0.1-3.0% by weight lubricant, for example, to facilitate tableting (e.g., about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.1%, about 1.2%, about 1.3%, about 1.4%, about 1.5%, about 1.6%, about 1.7%, about 1.8%, about 1.9%, about 2%, about 2.1%, about 2.2%, about 2.3%, about 2.4%, about 2.5%, about 2.6%, about 2.7%, about 2.8%, about 2.9%, or about 3.0%). In some embodiments, the formulation may further comprise about 1-10% by weight tablet disintegrant (e.g., about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10%). In some embodiments, the formulation may further comprise about 0.5-10% by weight an enteric polymer that dissolves at a pH of about 5.5 (e.g. EUDRAGIT L 30 D-55 + PlasACRYL HTP20. For example, the enteric polymer that dissolves at a pH of about 5.5 may be present in the formulation at about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10% by weight. In some embodiments, the formulation may further comprise about 1-15% by weight an enteric polymer that dissolves at a pH of about 7.0 (e.g. EUDRAGIT FS 30 D + PlasACRYL T20 and/or EUDRAGIT® S 100. For example, the enteric polymer that dissolves at a pH of about 7.0 may be present in the formulation at about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, or about 15% by weight. In such embodiments, the lovastatin analog or derivative (including without limitation lovastatin diol lactone) may be released in two doses. The first dose of lovastatin analog or derivative (including without limitation lovastatin diol lactone) is encapsulated by the enteric polymer that dissolves at a pH of about 5.5; and the second dose of lovastatin analog or derivative (including without limitation lovastatin diol lactone) is encapsulated by the enteric polymer that dissolves a at pH of about 7.0. [0111] For example, the formulation may comprise at least one mini-tablet that releases at a first pH (e.g. pH of about 5.5) and at least one mini-tablets that release at a second pH (e.g., pH of about 7.0) at a ratio of 1:5. The formulation may comprise about 10% by weight the lovastatin analog or derivative (including without limitation lovastatin diol lactone); about 50% by weight microcrystalline cellulose (e.g. Avicel PH102); about 5% by weight copovidone (e.g. Kollidon VA64 Fine); about 1% by weight silicon dioxide (e.g. Aerosil 200); about 0.5% by weight magnesium stearate; about 4% by weight crospovidone (e.g. Kollidon CL or Kollidon CL-F); about 2% by weight an enteric polymer that dissolves at a pH of about 5.5 (e.g. EUDRAGIT L 30 D-55 + PlasACRYL HTP20); and about 9% by weight an enteric polymer that dissolves at a pH of about 7.0. (e.g. EUDRAGIT FS 30 D + PlasACRYL T20 and/or EUDRAGIT® S 100). [0112] In another example, the formulation may comprise at least one mini-tablet that releases at a first pH (e.g. pH of about 5.5) and at least one mini-tablet that releases at a second pH (e.g., pH of about 7.0) at a ratio of 1:5. The formulation may comprise about 10% by weight the lovastatin analog or derivative (including without limitation lovastatin diol lactone); about 50% by weight microcrystalline cellulose (e.g. Avicel PH102); about 5% by weight copovidone (e.g. Kollidon VA64 Fine); about 1.4% by weight silicon dioxide (e.g. Aerosil 200); about 0.7% by weight magnesium stearate; about 4.3% by weight crospovidone (e.g. Kollidon CL or Kollidon CL-F); about 1.8% by weight an enteric polymer that dissolves at a pH of about 5.5 (e.g. EUDRAGIT L 30 D-55 + PlasACRYL HTP20); and about 8.9% by weight an enteric polymer that dissolves at a pH of about 7.0. (e.g. EUDRAGIT FS 30 D + PlasACRYL T20 and/or EUDRAGIT® S 100). [0113] The therapeutic agents or their pharmaceutically acceptable salts which are used in accordance with the present invention may exhibit stereoisomerism by virtue of the presence of one or more asymmetric or chiral centers in the compounds. The present invention contemplates the various stereoisomers and mixtures thereof. Desired enantiomers can be obtained by chiral synthesis from commercially available chiral starting materials by methods well known in the art, or may be obtained from mixtures of the enantiomers by resolution using known techniques. [0114] Solvate as used herein refers to a pharmaceutically acceptable solvate form of a specified therapeutic agent that retains the biological effectiveness of such agent. Examples of solvates include therapeutic agents of the invention in combination with, for example, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, or ethanolamine. [0115] Prodrug, as used herein refers to a therapeutic agent that is converted under physiological conditions or by solvolysis or metabolically (e.g., in vivo) to a specified agent that is pharmaceutically active. [0116] Active metabolite, as used herein refers to a pharmacologically active product produced through metabolism in the body of a specified therapeutic agent. [0117] Co-crystal as used herein refers to a physical association of two or more molecules which owe their stability through non-covalent interaction. One or more components of this molecular complex provide a stable framework in the crystalline lattice. In certain instances, the guest molecules are incorporated in the crystalline lattice as anhydrates or solvates.

Administration and Dosage [0118] It will be appreciated that the actual dose of the anti-methanogenic lovastatin analog or derivative to be administered according to the present invention will vary according to the particular compound, the particular dosage form, and the mode of administration. Many factors that may modify the action of the anti-methanogenic lovastatin analog or derivative (e.g., body weight, gender, diet, time of administration, route of administration, rate of excretion, condition of the subject, drug combinations, genetic disposition and reaction sensitivities) can be taken into account by those skilled in the art. Administration can be carried out continuously or in one or more discrete doses within the maximum tolerated dose. Optimal administration rates for a given set of conditions can be ascertained by those skilled in the art using conventional dosage administration tests. [0119] Individual doses of the anti-methanogenic lovastatin analog or derivative can be administered in unit dosage forms (e.g., tablets or capsules) containing, for example, from about 0.01 mg to about 100 mg, from about 0.1 mg to about 100 mg, from about 0.1 mg to about 90 mg, from about 0.1 mg to about 80 mg, from about 0.1 mg to about 70 mg, from about 0.1 mg to about 60 mg, from about 0.1 mg to about 50 mg, from about 0.1 mg to about 40 mg active ingredient, from about 0.1 mg to about 30 mg, from about 0.1 mg to about 20 mg, from about 0.1 mg to about 10 mg, from about 0.1 mg to about 5 mg, from about 0.1 mg to about 3 mg, from about 0.1 mg to about 1 mg per unit dosage form, or from about 5 mg to about 80 mg per unit dosage form. For example, a unit dosage form can be about 0.01 mg, about 0.02 mg, about 0.03 mg, about 0.04 mg, about 0.05 mg, about 0.06 mg, about 0.07 mg, about 0.08 mg, about 0.09 mg, about 0.1 mg, about 0.2 mg, about 0.3 mg, about 0.4 mg, about 0.5 mg, about 0.6 mg, about 0.7 mg, about 0.8 mg, about 0.9 mg, about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 6 mg, about 7 mg, about 8 mg, about 9 mg about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, about 80 mg, about 85 mg, about 90 mg, about 95 mg, or about 100 mg, inclusive of all values and ranges therebetween.

[0120] In one embodiment, the anti-methanogenic lovastatin analog or derivative is administered at an amount of from about 0.01 mg to about 100 mg daily, an amount of from about 0.1 mg to about 100 mg daily, from about 0.1 mg to about 95 mg daily, from about 0.1 mg to about 90 mg daily, from about 0.1 mg to about 85 mg daily, from about 0.1 mg to about 80 mg daily, from about 0.1 mg to about 75 mg daily, from about 0.1 mg to about 70 mg daily, from about 0.1 mg to about 65 mg daily, from about 0.1 mg to about 60 mg daily, from about 0.1 mg to about 55 mg daily, from about 0.1 mg to about 50 mg daily, from about 0.1 mg to about 45 mg daily, from about 0.1 mg to about 40 mg daily, from about 0.1 mg to about 35 mg daily, from about 0.1 mg to about 30 mg daily, from about 0.1 mg to about 25 mg daily, from about 0.1 mg to about 20 mg daily, from about 0.1 mg to about 15 mg daily, from about 0.1 mg to about 10 mg daily, from about 0.1 mg to about 5 mg daily, from about 0.1 mg to about 3 mg daily, from about 0.1 mg to about 1 mg daily, or from about 5 mg to about 80 mg daily. In various embodiments, the anti-methanogenic lovastatin analog or derivative is administered at a daily dose of about 0.01 mg, about 0.02 mg, about 0.03 mg, about 0.04 mg, about 0.05mg, about 0.06 mg, about 0.07 mg, about 0.08 mg, about 0.09 mg, about 0.1 mg, about 0.2 mg, about 0.3 mg, about 0.4 mg, about 0.5 mg, about 0.6 mg, about 0.7 mg, about 0.8 mg, about 0.9 mg, about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 6 mg, about 7 mg, about 8 mg, about 9 mg about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, about 80 mg, about 85 mg, about 90 mg, about 95 mg, or about 100 mg, inclusive of all values and ranges therebetween.

[0121] In some embodiments, a suitable dosage of the anti-methanogenic lovastatin analog or derivative is in a range of about 0.01 mg/kg to about 10 mg/kg of body weight of the subject, for example, about 0.01 mg/kg, about 0.02 mg/kg, about 0.03 mg/kg, about 0.04 mg/kg, about 0.05 mg/kg, about 0.06 mg/kg, about 0.07 mg/kg, about 0.08 mg/kg, about 0.09 mg/kg, about 0.1 mg/kg, about 0.2 mg/kg, about 0.3 mg/kg, about 0.4 mg/kg, about 0.5 mg/kg, about 0.6 mg/kg, about 0.7 mg/kg, about 0.8 mg/kg, about 0.9 mg/kg, about 1 mg/kg, about 1.1 mg/kg, about 1.2 mg/kg, about 1.3 mg/kg, about 1.4 mg/kg, about 1.5 mg/kg, about 1.6 mg/kg, about 1.7 mg/kg, about 1.8 mg/kg, 1.9 mg/kg, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg, about 10 mg/kg body weight, inclusive of all values and ranges therebetween. In other embodiments, a suitable dosage of the anti-methanogenic lovastatin analog or derivative is in a range of about 0.01 mg/kg to about 10 mg/kg of body weight, in a range of about 0.01 mg/kg to about 9 mg/kg of body weight, in a range of about 0.01 mg/kg to about 8 mg/kg of body weight, in a range of about 0.01 mg/kg to about 7 mg/kg of body weight, in a range of 0.01 mg/kg to about 6 mg/kg of body weight, in a range of about 0.05 mg/kg to about 5 mg/kg of body weight, in a range of about 0.05 mg/kg to about 4 mg/kg of body weight, in a range of about 0.05 mg/kg to about 3 mg/kg of body weight, in a range of about 0.05 mg/kg to about 2 mg/kg of body weight, in a range of about 0.05 mg/kg to about 1.5 mg/kg of body weight, or in a range of about 0.05 mg/kg to about 1 mg/kg of body weight. [0122] In accordance with certain embodiments of the invention, the anti-methanogenic lovastatin analog or derivative may be administered, for example, more than once daily, about once per day, about every other day, about every third day, about once a week, about once every two weeks, about once every month, about once every two months, about once every three months, about once every six months, or about once every year.

[0123] In various embodiments, the anti-methanogenic lovastatin analog or derivative may be administered in a patient that is fasting. In various embodiments, the anti-methanogenic lovastatin analog or derivative may be administered in a patient with a meal. In various embodiments, the anti-methanogenic lovastatin analog or derivative may be administered in a patient that is postprandial. In various embodiments, patient is on an elemental diet. A comestible total enteral nutrition (TEN) formulation, which is also called an “elemental diet” are commercially available, for example, VIVONEX T.E.N. (Nestle) and its variants, or the like. A useful total enteral nutrition formulation satisfies all the subject’s nutritional requirements, containing free amino acids, carbohydrates, lipids, and all essential vitamins and minerals, but is in a form that is readily absorbable in the upper gastrointestinal tract, thus depriving or“starving” the methanogen syntrophic microorganism of nutrients of at least some of the nutrients they use for proliferating. Thus, methanogen syntrophic microorganism growth is inhibited.

Additional Agents and Combination Therapy or Co-Formulation / Patient Selection

[0124] Administration of the present formulations may be combined with additional therapeutic agents. Co- administration of the additional therapeutic agent and the present formulations may be simultaneous or sequential. Further the present formulations may comprise an additional therapeutic agent (e.g. via co- formulation).

[0125] In some embodiments, the modified-release formulations of the present invention are administered in combination with an additional therapeutic agent. In an embodiment, the additional therapeutic agent and the anti-methanogenic lovastatin analog or derivative are combined into a single modified-release formulation. In some embodiments, the methods of treatment and/or prevention comprise administering the modified-release formulations of the present invention to a subject that is undergoing treatment with an additional therapeutic agent.

[0126] In one embodiment, the additional therapeutic agent and the anti-methanogenic lovastatin analog or derivative are administered to a subject simultaneously. The term“simultaneously” as used herein, means that the additional therapeutic agent and the anti-methanogenic lovastatin analog or derivative are administered with a time separation of no more than about 60 minutes, such as no more than about 30 minutes, no more than about 20 minutes, no more than about 10 minutes, no more than about 5 minutes, or no more than about 1 minute. Administration of the additional therapeutic agent and the anti-methanogenic lovastatin analog or derivative can be by simultaneous administration of a single formulation (e.g., a formulation comprising the additional therapeutic agent and the anti-methanogenic lovastatin analog or derivative) or of separate formulations (e.g., a first formulation including the additional therapeutic agent and a second formulation including the anti-methanogenic lovastatin analog or derivative).

[0127] Co-administration does not require the additional therapeutic agents to be administered simultaneously, if the timing of their administration is such that the pharmacological activities of the additional therapeutic agent and the anti-methanogenic lovastatin analog or derivative overlap in time, thereby exerting a combined therapeutic effect. For example, the additional therapeutic agent and the anti-methanogenic lovastatin analog or derivative can be administered sequentially. The term“sequentially” as used herein means that the additional therapeutic agent and the anti-methanogenic lovastatin analog or derivative are administered with a time separation of more than about 60 minutes. For example, the time between the sequential administration of the additional therapeutic agent and the anti-methanogenic lovastatin analog or derivative can be more than about 60 minutes, more than about 2 hours, more than about 5 hours, more than about 10 hours, more than about 1 day, more than about 2 days, more than about 3 days, or more than about 1 week apart. The optimal administration times will depend on the rates of metabolism, excretion, and/or the pharmacodynamic activity of the additional therapeutic agent and the anti-methanogenic lovastatin analog or derivative being administered. Either the additional therapeutic agent or the anti-methanogenic lovastatin analog or derivative may be administered first.

[0128] Co-administration also does not require the additional therapeutic agents to be administered to the subject by the same route of administration. Rather, each therapeutic agent can be administered by any appropriate route, for example, parenterally or non-parenterally.

[0129] The formulations of the present invention may comprise a pharmaceutically acceptable excipient. In some embodiments, the formulation may further include agent which prevents or reduces lactone ring-opening, such as an esterase inhibitor (e.g. grapefruit juice or components naringenin, kaempferol) and/or a paraoxonase inhibitor (e.g. PON1 or PON3 inhibitor). In some embodiments, the esterase inhibitor and/or a paraoxonase inhibitor is one or more of amiodarone, anastrozole, azithromyzcin, cannabinoids, cimetidine, clarithromycin, clotrimazolem, cyclosporine, danazol, delavirdine, dexamethasone, diethyldithiocarbamate, diltiazem, dirithyromycin, disulfiram, entacapone, erythromycin, ethinyl estradiol, fluconazole, fluoxetine, fluvoaxamine, gestodene, grapefruit juice, indinavir, isoniazid, ketoconazole, metronidazole, mibefradil, miconazole, nefazodone, nelfinavir, nevirapine, norfloxacin, norfluoxetine, omeprazole, oxiconazole, paroxetine, propoxyphene, quinidine, quinine, quinupristine and dalfopristin, ranitidine, ritonavir, saquinavir, sertindole, sertraline, troglitazone, troleandomycin, valproic acid and/or a lactam agent selected from oxindole, isatin, δ- valerolactam, ε-caprolactam, 2-hydroxyquinoline, and 3,4-dihydro-2(1H)-quinoline and N-bromo-ε-caprolactam.

[0130] In various embodiments, the modified-release formulation of the present invention is administered in combination with an inhibitor of the organic anion transporting polypeptide (OATP) transporter. In an embodiment, the OATP inhibitor and the anti-methanogenic lovastatin analog or derivative are combined into a single modified-release formulation. Without wishing to be bound by theory, it is believed that inclusion of the OATP inhibitor minimizes absorption of the anti-methanogenic lovastatin analog or derivative from the intestine and/or reduces the enterohepatic recirculation of the anti-methanogenic lovastatin analog or derivative, thereby maximizing retention of the anti-methanogenic lovastatin analog or derivative in the intestine and minimizing any potential systemic side effects of the anti-methanogenic lovastatin analog or derivative. Illustrative OATP inhibitors include, but are not limited to, grapefruit juice or grapefruit juice constituents such as naringin and hesperidin, orange juice and orange juice constituents, apple juice and apple juice constituents, and green tea and green tea extracts such as epicatechin gallate (ECG), epigallocatechin gallate (EGCG). In an embodiment, the OATP inhibitor is released in the intestine prior to release of the anti-methanogenic lovastatin analog or derivative. [0131] In one embodiment, the additional therapeutic agent is a prokinetic agent that facilitates movement of a mass through the intestinal tract. Illustrative prokinetic agents include, but are not limited to, prucalopride (e.g. RESOLOR) or a macrolide antibiotic such as erythromycin. In another embodiment, the additional therapeutic agent is a natural product such as peppermint oil, which alleviates abdominal pain.

[0132] The present invention also contemplates the use of additional therapeutic agent that are useful for treating constipation such as, for example, laxatives, guanylate cyclase C agonist (e.g., linaclotide), a serotonin agonist (e.g., prucalorpride, tegaserod), a chloride channel agonist (e.g., lubiprostone), and combinations thereof.

[0133] In some embodiments, the additional therapeutic agent is an agent useful for treating IBS (including IBS-C). In some embodiments, the additional therapeutic agent is a selective chloride channel activator, including, for example, molecules derived from prostaglandins such as lubiprostone (e.g. AMITIZA) and those compounds described in US Patent Nos.5,284,858, 6,414,016 and 6,583,174, the contents of which are hereby incorporated by reference in their entireties. In some embodiments, the additional therapeutic agent is an agent, including a peptide agent, that increases the secretion of chloride and/or water in the intestines and/or soften stools and/or stimulate bowel movements, such as, for example, linaclotide (e.g. LINZESS) and those compounds described in US Patent No.7,304,036, the contents of which are hereby incorporated by reference in their entirety. In some embodiments, the additional therapeutic agent is an agent that relaxes the colon and/or slows the movement of waste through the lower bowel. In some embodiments the additional therapeutic agent is a 5-HT 3 antagonist, including, but not limited to, alosetron (e.g. LOTRONEX).

[0134] In some embodiments, the additional therapeutic agent is a small molecule that acts as a peripherally selective κ-opioid agonist, such as, for example, EMD-61753 ((N-methyl-N-[(1S)-1-phenyl-2-((3S)-3- hydroxypyrrolidin-1-yl)-ethyl]- 2,2-diphenyl-acetamide hydrochloride, ASMADOLINE) and those compounds described in US Patent No. US 6,344,566, the contents of which are hereby incorporated by reference in their entirety. In some embodiments, the additional therapeutic agent is a cholecystokinin antagonist, e.g. one selective for the CCK A subtype and/or inhibits gastrointestinal motility and reduces gastric secretions, such as, for example, Dexloxiglumide ((4R)-4-[(3,4-dichlorobenzoyl)amino]-5-(3-methoxypropylpenty lamino)-5- oxopentanoic acid) and those compounds described in US Patent No. US 5,602,179, the contents of which are hereby incorporated by reference in their entirety. In some embodiments, the additional therapeutic agent is tapentadol (1R,2R)-3-(3-dimethylamino-1-ethyl-2-methyl-propyl)-phenol), as described in US Patent Publication No.2013/0116334, the contents of which are hereby incorporated by reference in their entirety

[0135] In some embodiments, the additional therapeutic agent is a laxative, including but not limited to osmotic laxatives (such as, for example, magnesium carbonate, magnesium hydroxide (e.g. Milk of Magnesia), magnesium oxide, magnesium peroxide, magnesium sulfate, lactulose, lactitol, sodium sulfate, pentaerythritol, macrogol, mannitol, sodium phosphate, sorbitol, magnesium citrate, sodium tartrate, laminarid, and polyethylene glycol (e.g., macrogol-containing products, such as MOVICOL and polyethylene glycol 3350, or SOFTLAX, MIRALAX, DULCOLAX BALANCE, CLEARLAX, OSMOLAX OR GLYCOLAX, GOLYTELY, GAVILYTE C, NULYTELY, GLYCOLAX, FORTRANS, TRILYTE, COLYTE, HALFLYTELY, SOFTLAX, LAX-A-DAY, CLEARLAX AND MOVIPREP). In some embodiments, the additional therapeutic agent is a laxative, including but not limited to stimulant laxatives (such as, for example, SENOKOT). Also provided are contact laxatives (e.g. oxyphenisatine, bisacodyl, dantron, phenolphthalein, castor oil, senna glycosides, cascara, sodium picosulfate, and bisoxatin) and bulk-forming laxatives (e.g. ispaghula, ethulose, sterculia, linseed, methylcellulose, triticum, and polycarbophil calcium). In some embodiments, the additional therapeutic agent is an enema, such as, for example, sodium laurilsulfate, sodium phosphate, bisacodyl, dantron, glycerol, oil, and sorbitol. Peripheral opioid antagonists such as, for example, Alvimopan and Methylnaltrexone, as well as Prostaglandins such as, for example, Lubiprostone are also additional therapeutic agents in some embodiments. Also, Linaclotide, Prucalopride, and Tegaserod may be additional therapeutics.

[0136] In some embodiments, the additional therapeutic agent is an agent used for long-term pain and cramping, including but not limited to anticholinergics (antispasmodics), such as, for example, dicyclomine (BENTYL) and or antidepressants, including, for example, desipramine (such as, for example, NORPRAMIN), imipramine (TOFRANIL) or nortriptyline (PAMELOR), which are optionally administered at low doses. In low doses, they can help with pain caused by IBS.

[0137] In some embodiments, the additional therapeutic agent is fiber supplement, such as, for example, psyllium (METAMUCIL) or methylcellulose (CITRUCEL).

[0138] In some embodiments, the additional therapeutic agent is an agent useful for treating obesity. Illustrative agents include, but are not limited to, orlistat, loracaserin, phentermine-topiramate, sibutramine, rimonabant, exenatide, pramlintide, phentermine, benzphetamine, diethylpropion, phendimetrazine, bupropion, and metformin. In various embodiments, the additional agent is an agent that that interfere with the body’s ability to absorb specific nutrients in food, such as orlistat, glucomannan, and guar gum. Agents that suppress appetite are also among the additional agents, e.g. catecholamines and their derivatives (such as phentermine and other amphetamine-based drugs), various anti-depressants and mood stabilizers (e.g. bupropion and topiramate), anorectics (e.g. dexedrine, digoxin). Agents that increase the body’s metabolism are also among the additional therapeutic agents. In some embodiments, additional therapeutic agents may be selected from among appetite suppressants, neurotransmitter reuptake inhibitors, dopaminergic agonists, serotonergic agonists, modulators of GABAergic signaling, anticonvulsants, antidepressants, monoamine oxidase inhibitors, substance P (NKl) receptor antagonists, melanocortin receptor agonists and antagonists, lipase inhibitors, inhibitors of fat absorption, regulators of energy intake or metabolism, cannabinoid receptor modulators, agents for treating addiction, agents for treating metabolic syndrome, peroxisome proliferator-activated receptor (PPAR) modulators; and dipeptidyl peptidase 4 (DPP-4) antagonists. In some embodiments, additional therapeutic agents may be selected from among amphetamines, benzodiazepines, sulfonyl ureas, meglitinides, thiazolidinediones, biguanides, beta-blockers, ACE inhibitors, diuretics, nitrates, calcium channel blockers, phenlermine, sibutramine, lorcaserin, cetilistat, rimonabant, taranabant, topiramate, gabapentin, valproate, vigabatrin, bupropion, tiagabine, sertraline, fluoxetine, trazodone, zonisamide, methylphenidate, varenicline, naltrexone, diethylpropion, phendimetrazine, repaglinide, nateglinide, glimepiride, pioglitazone, rosiglilazone, and sitagliptin.

[0139] In an embodiment, the additional therapeutic agent is an agent for treating pre-diabetes, diabetes, type II diabetes, insulin resistance, glucose intolerance, or hyperglycemia. Examples of drugs include, but are not limited to, alpha-glucosidase inhibitors, amylin analogs, dipeptidyl peptidase-4 inhibitors, GLP1 agonists, meglitinides, sulfonylureas, biguanides, thiazolidinediones (TZD), and insulin. Additional examples of such agents include bromocriptine and Welchol. Examples of alpha-glucosidase inhibitors include but are not limited to acarbose and miglitol. An example of an amylin analog is pramlintide. Examples of dipeptidyl peptidase-4 inhibitors include but are not limited to saxagliptin, sitagliptin, vildagliptin, linagliptin, and alogliptin. Examples of GLP1 agonist include but are not limited to liraglutide, exenatide, exenatide extended release. Examples of meglitinides include but are not limited to nateglinide, and repaglinide. Examples of sulfonylureas include but are not limited to chlorpropamide, glimepiride, glipizide, glyburide, tolazamide, and tolbutamide. Examples of biguanides include but are not limited to metformin, Riomet, Glucophage, Glucophage XR, Glumetza. Examples of thiazolidinedione include but are not limited to rosiglitazone and pioglitazone. Examples of insulin include but are not limited to Aspart, Detemir, Glargine, Glulisine, and Lispro. Examples of combination drugs include but are not limited to glipizide/metformin, glyburide/metformin, pioglitazone/glimepiride, pioglitazone/metformin, repaglinide/metformin, rosiglitazone/glimepiride, rosiglitazone/metformin, saxagliptin/metformin, sitagliptin/simvastatin, sitagliptin/metformin, linagliptin/metformin, alogliptin/metformin, and alogliptin/pioglitazone.

[0140] In another embodiment, the additional therapeutic agent is a probiotic. In some embodiments, enteric dietary formulations containing low residual material, such as pre-digested or basic amino acid formulations and other methods and products as described in U.S. Patent 8,110,177 (the contents of which are incorporated herein by reference) may be employed. In a further embodiment, such low residual enteric dietary formulations may be formulated in low carbohydrate and low fat forms either with or without lovastatin analogs or derivatives or red yeast rice which may be particularly useful for weight loss and diabetes. In various embodiments, the probiotic may comprise the following illustrative cells: E. coli Nissle 1917, a lactobacillus (e.g. acidophilus, Lactobacillus brevis, L. bulgaricus, L. plantarum, L. rhamnosus, Rhamnosus L. fermentum, L. caucasicus, L. helveticus, L. lactis, L. reuteri and L. casei) or a bifidobacteria (Bifidobacterium bifidum, B. infantis) Streptococcus thermophiles, and Enterococcus faecium. Other suitable probiotics and prebiotics are disclosed for example in R. Spiller, Aliment Pharmacol Ther 28, 385-396, the contents of which are hereby incorporated by reference in their entirety.

[0141] In some embodiments, a probiotic agent that optionally inhibits the growth of methanogens, for example, Bifidobacterium spp. or Lactobacillus species or strains, e.g., L. acidophilus, L. rhamnosus, L. plantarum, L. reuteri, L. paracasei subsp. paracasei, or L. casei Shirota, or probiotic Saccharomyces species, e.g., S. cerevisiae, is selected and/or administered. The probiotic agent that inhibits methanogenesis may be administered in a pharmaceutically acceptable ingestible formulation, such as in a capsule, or for some subjects, consuming a food supplemented with the inoculum is effective, for example milk, yogurt, cheese, meat or other fermentable food preparation. Probiotic agents can inhibit the growth of methanogens, for example, by competing against methanogens for growth and thus reduce or inhibit the growth of methanogens.

Methods of Treatment

[0142] In one aspect, the present invention provides methods of treating or preventing a methanogen- associated disorder by administering an anti-methanogenic lovastatin analog or derivative to the intestine (i.e., small and/or large intestine) in a subject in need thereof. In another aspect, the present invention provides methods of treating or preventing a methanogen-associated disorder by administering a modified-release formulation comprising at least one anti-methanogenic lovastatin analog or derivative to the intestine (i.e., small and/or large intestine) in a subject in need thereof.

[0143] In some embodiments, the methanogen-associated disorder is a disease or disorder or condition caused by, resulted from, or related to one or more of the abnormal presence or absence of methanogens, abnormal levels of methanogens, overgrowth of methanogens, elevated levels of methanogenesis, elevated enteric methane levels, excessive hydrogen scavenging by hydrogen-consuming methanogens or colonization of methanogens in an abnormal location (e.g., in the small bowel rather than large bowel), either alone or in combination with non-methanogen syntrophic organisms.

[0144] Illustrative methanogen-associated disorders include, but are not limited to, enteric methanogen colonization, IBS, IBS-C, IBS-M, constipation, diabetes, type 2 diabetes, metabolic syndrome, insulin resistance, metabolic syndrome, obesity, constipation, chronic constipation, chronic intestinal pseudo-obstruction, systemic sclerosis, systemic lupus, erythematosus, dermatomysitis/polymyositis, periartiytis nodosa, mixed connective tissue disorder, rheumatoid arthritis, spinal cord injury, Parkinson’s disease, hypothyroidism/hypoparathyroidism, Hirschsprung’s disease, Chagas’ disease, intestinal hypoganglionosis, and Ehlers-Danlos Syndrome.

[0145] In one aspect, the present invention provides methods of reducing or eliminating the production and/or accumulation of methane in the GI tract by administering an anti-methanogenic lovastatin analog or derivative or a modified-release formulation comprising at least one anti-methanogenic lovastatin analog or derivative to the intestine (e.g. the small and/or large intestine) of a subject in need thereof. In another aspect, the present invention provides methods of reducing or eliminating methane, for example as produced by a methanogen in the GI tract by administering an anti-methanogenic lovastatin analog or derivative or a modified- release formulation comprising at least one anti-methanogenic lovastatin analog or derivative to the intestine (i.e., small and/or large intestine) of a subject in need thereof.

[0146] In various embodiments, the methanogen is a microorganism that produces methane as a metabolic byproduct. Methanogens are classified as archaea. Examples of methanogens include but are not limited to Methanobacterium bryantii, Methanobacterium formicum, Methanobrevibacter arboriphilicus, Methanobrevibacter gottschalkii, Methanobrevibacter ruminantium, Methanobrevibacter smithii, Methanocalculus chunghsingensis, Methanococcoides burtonii, Methanococcus aeolicus, Methanococcus deltae, Methanococcus jannaschii, Methanococcus maripaludis, Methanococcus vannielii, Methanocorpusculum labreanum, Methanoculleus bourgensis (Methanogenium olentangyi, Methanogenium bourgense), Methanoculleus marisnigri, Methanofollis liminatans, Methanogenium cariaci, Methanogenium frigidum, Methanogenium organophilum, Methanogenium wolfei, Methanomicrobium mobile, Methanopyrus kandleri, Methanoregula boonei, Methanosaeta concilii, Methanosaeta thermophile, Methanosarcina acetivorans, Methanosarcina barkeri, Methanosarcina mazei, Methanosphaera stadtmanae, Methanospirillium hungatei, Methanothermobacter defluvii (Methanobacterium defluvii), Methanothermobacter thermautotrophicus (Methanobacterium thermoautotrophicum), Methanothermobacter thermoflexus (Methanobacterium thermoflexum), Methanothermobacter wolfei (Methanobacterium wolfei), and Methanothrix sochngenii.

[0147] In one aspect, the present invention provides methods of reducing or eliminating the methane derived from Methanobrevibacter smithii in the GI tract. In another aspect, the present invention provides methods of reducing or eliminating methane, for example as produced by Methanobrevibacter smithii, in the GI tract by administering an anti-methanogenic lovastatin analog or derivative or a modified-release formulation comprising at least one anti-methanogenic lovastatin analog or derivative to the intestine (i.e., small and/or large intestine) in a subject in need thereof.

[0148] In various embodiments, the present invention relates to the substantial reduction of methane gas in a subjects GI tract (e.g. eradication of intestinal methane). In some embodiments the present formulations and methods prevent the increase in levels of methane gas in a subject’s GI tract. In some embodiments, the patient’s GI methane levels (as assessed by methods described herein and methods known in the art) are reduced to about 1 ppm, or about 2 ppm, or about 3 ppm, or about 4 ppm, or about 5 ppm, or about 10 ppm, or about 15 ppm, or about 20 ppm, or about 25 ppm, or about 30 ppm, or about 35 ppm, or about 40 ppm, or about 45 ppm, or about 50 ppm, or about 55 ppm, or about 60 ppm, or about 65 ppm, or about 70 ppm, or about 75 ppm, or about 80 ppm, or about 85 ppm, or about 90 ppm, or about 100 ppm. In various embodiments, the present formulations and methods reduce the patient’s GI methane levels to less than about 250 ppm, or less than about 225 ppm, or less than about 200 ppm, or less than about 175 ppm, or less than about 150 ppm, or less than about 125 ppm, or less than about 100 ppm, or less than about 50 ppm. In various embodiments, substantial reduction of methane gas is not accompanied by a substantial reduction in hydrogen gas.

[0149] In various embodiments, the present invention relates to the treatment of IBS, including IBS-C as described by ICD-10 (International Statistical Classification of Diseases and Related Health Problems, WHO edition). In various embodiments, the present invention relates to the treatment of irritable colon, as classified in ICD-10 as [K58]. IBS may include irritable bowel syndrome without diarrhea, as classified in ICD-10 as [K58.9]. Irritable bowel syndrome without diarrhea may also include irritable bowel syndrome not otherwise specified (NOS). Further, the diseases as classified in ICD-10 as K59 are also included (e.g. constipation; K59.1 Functional diarrhea; K59.2 Neurogenic bowel, not elsewhere classified; K59.3 Megacolon, not elsewhere classified (including dialatation of colon, toxic megacolon, megacolon in Chagas disease (B57.3), congenital (aganglionic) (Q43.1), and Hirschsprung disease (Q43.1)); K59.4 Anal spasm (including Proctalgia fugax); K59.8 Other specified functional intestinal disorders (including atony of colon) and K59.9 Functional intestinal disorder, unspecified).

[0150] In various embodiments, the present invention relates to the treatment of spastic colon, nervous colitis, mucous colitis, functional colitis or colonic neurosis. In various embodiments, the present invention relates to the treatment of diseases that have been described as sigma elongatum mobile, cecum mobile, chronic colitis, splanchnoptosia and the like. Typological classification of the disease generally include convulsive large bowel, diarrhea nervosa and colica mucosa, and the disease may also be classified in convulsive constipation type, atonic constipation type, intestinal gas syndrome, or chronic celiopathy.

[0151] Furthermore, IBS may also include cholangiodyskinesia, gastric emptying hypofunction, hysteric globus, non-specific esophagus functional abnormalities, nervous vomiting, recurrent abdominal pain, simple constipation, chronic idiopathic constipation and the like. As diagnostic criteria of IBS those of NIH, Manning, Cook et al. and the like are suitable (see Asakura, Clinical Digestive Internal Medicine, 8 (8): 1373-1381 (1993), the contents of which are hereby incorporated by reference in their entirety).

[0152] In various embodiments, the present invention relates to the treatment of IBS, including IBS-C of varying stages or severity. In one embodiment, stages or severity of the IBS may be evaluated with a health- related quality of life (HRQoL) evaluation. In some embodiments, the stage or severity of the disease in the patient to be treated is assessed by an evaluation of one or more of patient pain, distension, bowel dysfunction and quality of life/global well-being.

[0153] In some embodiments, the stage or severity of the disease in the patient to be treated is assessed by the Rome Scale (for the last 3 months with symptom onset at least 6 months prior to diagnosis: recurrent abdominal pain or discomfort (e.g. uncomfortable sensation not described as pain.) at least 3 days/month in the last 3 months associated with two or more of improvement with defecation, onset associated with a change in frequency of stool, and onset associated with a change in the form (appearance) of stool.

[0154] In some embodiments, the stage or severity of the disease in the patient to be treated is assessed by the Kruis scale (Gastroenterology 87: 1-7, the contents of which are hereby incorporated by reference). This scale incorporates both the“cardinal” symptoms (pain, bloating, altered bowel function) and“red flag” signs of potential underlying organic disease that would thus exclude an IBS diagnosis. IBS is diagnosed if the sum of scores >44.

[0155] In some embodiments , the patient is evaluated with the ass essment desc ribed in Fran cis, et al Aliment P harmacol Th er 1997; 11: 395-402, the c ontents of w hich are here by incorporate d by referenc e in their entirety. For instance, a scoring sy stem based o n patient ran king of pain, distension, b owel dysfun ction and quality o f life/global w ell-being on a scale of up t o 500 is used . Mild, moder ate and sever e cases were indicated by scores of 75 to 175 , 175 to 300 and >300. In some embod iments, the p atient of the p resent invent ion has a score of 75 to 175. In s ome embodim ents, the pat ient of the pre sent inventio n has a score of 175 to 300. In some embodim ents, the pat ient of the p resent invent ion has a sc ore of >300. In some em bodiments t he scales described in Wong an d Drossman ( Expert Rev. Gastroenterol . Hepatol. 4(3 ), (2010), the contents of which are hereby in corporated b y reference in their entire ty). For exam ple, in some embodimen ts, the patien ts of the present i nvention are e valuated for the paramete rs of dysphor ia, activity inte rference, bod y image, hea lth worry, food avo idance, social reaction, and sexual relat ionships and optionally sco red on a 0-100 as describ ed on the Patrick s cale; and/or t he patients o f the present invention are evaluated fo r the parame ters of daily activities, emotiona l impact, fam ily relations, food, sleep and fatigue, social impa ct, sexual re lations symp toms and optionally scored on a 0-216 as des cribed on the Groll scale; t he patients of the present i invention are evaluated for the pa rameters of a ctivities, anxi ety, diet, slee p, discomfort, health perce ption, disease coping and s tress and optionally scored on a 0-100 as de scribed on t he Chassany scale; the p atients of the present inve ntion are evaluated for the para meters of em otional health , mental healt h, sleep, ene rgy, physical functioning, d iet, social role, phys ical role, and sexual relatio ns and option ally scored o n a 0-100 as described on the Hahn sca le; and/or the patie nts of the pr esent inventi on are evalu ated for the parameters o f bowel sym ptoms, fatigu e, activity impairme nt, emotional dysfunction a nd optionally scored as d omain averag e scores (cal lculated by di viding the domain s um score by t he number of items: range 1–7) as desc ribed on the W ong scale.

[0156] In some embodiments , patients may be stratified based on one or more of m ethane dete ction (e.g. via breat h test) and m ethanogen d etection (e.g . via PCR, e .g. qPCR). In some embo diments, the patient is considered methane breath test positive if the subject presents with greater than about 3 ppm methane. In some embodiments, the patient of the present invention has greater than about 10 4 , or about 10 5 , or about 10 6 copies of M. smithii per grams of wet stool. In some embodiments, the patient of the present invention is defined by a measurement of the fractional methanogen contribution to the total microbial content of the feces. In some embodiments, the patient has greater than about 0.5%, or about 0.6%, or about 0.7%, or about 0.8%, or about 0.9%, or about 1.0%, or about 1.1%, or about 1.2%, or about 1.3%, or about 1.4%, or about 1.5%, or about 2.5% M. smithii fraction of the total microbial content of the feces.

[0157] In some embodiments, methods of the present invention treat or prevent constipation. Constipation may be associated with, for example, chemotherapy, vinca alkaloids, oxaliplatins, taxanes, thalidomide, opioids, sedatives, anticholinergics, gastrointestinal antispasmodics, antiparkinsonism agents, antidepressants, phenothiazines, calcium- and aluminum-based antacids, diuretics, tranquilizers, sleeping medications, general anesthesia, pudendal blocks, inadequate fluid intake, excessive use of laxatives and/or enemas, prolonged immobility, inadequate exercise. spinal cord injury or compression, fractures, fatigue, weakness, inactivity, bedrest, cardiac problems, diverticulitis, neurological lesions, cerebral tumors, spinal cord injury, spinal cord compression, paraplegia, cerebrovascular accident with paresis, weak abdominal muscles, hypothyroidism, lead poisoning, uremia, dehydration, hypercalcemia, hypokalemia, hyponatremia, anorexia, immobility, antidepressants, inability to increase intra-abdominal pressure, emphysema, neuromuscular impairment of the diaphragm, neuromuscular impairment of abdominal muscles, abdominal hernias, malnutrition, cachexia, anemia, carcinoma, and senility.

[0158] In various embodiments, the constipation is associated with IBS. But, the present invention, in some embodiments, can also relate to chronic functional constipation.

[0159] In various embodiments, the present invention relates to the treatment of increased visceral hypersensitivity. In various embodiments, the present invention relates to the treatment of one or more of stomachaches, pain, nausea, straining, and bloating and/or gas. The present formulations and methods also treat one or more of as hard stools, infrequent stools, difficulty or straining at stools, feeling of being unable to completely empty during a bowel movement, and the sensation of wanting to go but not being able to.

[0160] In various embodiments, the present invention relates to the treatment for diabetes (type 1 or type 2) and/or glucose intolerance. In some embodiments, the present invention relates to a method for treating patient at risk of diabetes, one or more of insulin resistance, prediabetes, impaired fasting glucose (IFG), impaired glucose tolerance (IGT), and acanthosis nigricans.

[0161] In some embodiments, methods for inducing weight loss or preventing weight gain (or treating or preventing obesity or inducing weight loss or preventing weight gain in a patient that does not substantially change caloric intake), comprising administering an anti-methanogenic lovastatin analog or derivative or a modified-release formulation of the present invention are provided. Patients may have undertaken or will undertake a surgery of the digestive system; be greater than about 80-100 pounds overweight; have a BMI of greater than about 35 kg/m 2 ; or have a health problem related to obesity

[0162] In some embodiments, administration of the anti-methanogenic lovastatin analog or derivative or the modified-release formulation of the present invention does not confer cholesterol-lowering cardiovascular effects associated with systemic administration. For example, the present formulations and methods may avoid or reduce a subject’s systemic exposure to the lovastatin analog or derivative. For example, the present formulations and methods may provide an average reduction of less than about 20%, about 19%, about 18%, about 17%, about 16%, about 15%, about 14%, about 13%, about 12%, about 11%, about 10%, about 9%, about 8%, about 7%, about 6%, about 5%, about 4%, about 3%, or about 2% in serum LDL-C levels after treatment.

[0163] In some embodiments, the patient is one who does not require statins or statin analogs or derivatives for their cardiovascular therapeutic uses. In some embodiments, the patient is one who does not require statins or statin analogs or derivatives for their cardiovascular therapeutic uses and is methane-positive (e.g. as assessed by the methods described herein such as the methane breath test and qPCR).

[0164] By maximizing retention of the anti-methanogenic lovastatin analog or derivative to the intestines, the methods of the invention also minimize the side effects associated with systemic release. For example, the present method prevents and/or minimizes various adverse effects associated with lovastatin or lovastatin analog or derivative usage including, muscle-associated adverse effects, such as myositis, myalgia, rhabdomyolysis, drug-drug-interactions, cognitive effects, increased cancer risk, increases in liver enzymes, hemorrhagic stroke, increase in blood glucose levels, sleep disorders, peripheral neuropathy, sexual dysfunction, thyroid dysfunction, renal toxicity, irritability, shortness of breath, hyperkalemia, weight gain, neurodegenerative disease, pancreatitis, liver pathology, mitochondrial syndromes, dermatologic conditions, dry mouth, cataracts, olfaction, hematalogic and bone marrow adverse effects, hypotension, gastrointestinal adverse effects, including, ulcerative colitis and gastric ulceration, fatigue and headache. In some embodiments, the methods of the invention also minimizes the following side effects associated with systemic release: muscle pain, tenderness, or weakness, lack of energy, weakness, fever, dark colored urine, jaundice, pain in the stomach, including the upper right part of the stomach, nausea, unusual bleeding or bruising, loss of appetite, flu-like symptoms, rash, hives, itching, difficulty breathing or swallowing, and swelling of the face, throat, tongue, lips, eyes, hands, feet, ankles, or lower legs, hoarseness.

[0165] Accordingly, the anti-methanogenic lovastatin analog or derivative or the modified-release formulation of the present invention may be used to target subjects where systemic statin or statin analog or derivative levels are undesirable. In one embodiment, the subject may be women and children who are otherwise healthy and have no need for a cardiovascular medicine (as characterized, for example, as having low or zero myocardial event risk factors as per the ATP III Guideline). In another embodiment, the subject may be a child with IBS-C who has no need for a cholesterol-lowering agent. In such embodiments, administration of the anti- methanogenic lovastatin analog or derivative or the modified-release formulation of the present invention results in an average reduction of less than about 20%, about 19%, about 18%, about 17%, about 16%, about 15%, about 14%, about 13%, about 12%, about 11%, about 10%, about 9%, about 8%, about 7%, about 6%, about 5%, about 4%, about 3%, or about 2% in serum LDL-C levels after treatment.

[0166] The anti-methanogenic lovastatin analog or derivative or the modified-release formulation of the present invention may also be utilized as part of a treatment regimen wherein a subject is provided with an initial anti-methanogenic therapy followed by a chronic anti-methanogenic or methane-reducing and/or eliminating maintenance therapy.

[0167] The initial anti-methanogenic therapy may employ agents other than statins or statin analogs or derivatives such as, for example, antibiotics which eradicate the methanogens. For example nitroimidazoles such as metronidazole, metronidazole esters and/or isomers or hydrophobic imidazole derivatives or rifaximin or neomycin sufficient to eradicate, substantially reduce, or reduce the enteric methanogen colonization may be used. Such initial therapy may be for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 14, 28, 42, 56, 60, 90, 120 or 180 days or more. Examples of antibiotics include but are not limited to aminoglycosides (e.g., amikacin, gentamicin, kanamycin, neomycin, netilmicin, streptomycin, tobramycin, paromomycin), ansamycins (e.g., geldanamycin, herbimycin), carbacephems (e.g., loracarbef), carbapenems (e.g., ertapenem, doripenem, imipenem, cilastatin, meropenem), cephalosporins (e.g., first generation: cefadroxil, cefazolin, cefalotin or cefalothin, cefalexin; second generation: cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime; third generation: cefixime, cefdinir, cefditoren, cefoperazone, cefotaxime, cefpodoxime, ceftazidime, ceftibuten, ceftizoxime, ceftriaxone; fourth generation: cefepime; fifth generation: ceftobiprole), glycopeptides (e.g., teicoplanin, vancomycin), macrolides (e.g., azithromycin, clarithromycin, dirithromycin, erythromycin, roxithromycin, troleandomycin, telithromycin, spectinomycin), monobactams (e.g., aztreonam), penicillins (e.g., amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, meticillin, nafcillin, oxacillin, penicillin, piperacillin, ticarcillin), antibiotic polypeptides (e.g., bacitracin, colistin, polymyxin b), quinolones (e.g., ciprofloxacin, enoxacin, gatifloxacin, levofloxacin, lomefloxacin, moxifloxacin, norfloxacin, ofloxacin, trovafloxacin), rifamycins (e.g., rifampicin or rifampin, rifabutin, rifapentine, rifaximin), sulfonamides (e.g., mafenide, prontosil, sulfacetamide, sulfamethizole, sulfanilamide, sulfasalazine, sulfisoxazole, trimethoprim, trimethoprim-sulfamethoxazole (co- trimoxazole,“tmp-smx”), and tetracyclines (e.g., demeclocycline, doxycycline, minocycline, oxytetracycline, tetracycline) as well as arsphenamine, chloramphenicol, clindamycin, lincomycin, ethambutol, fosfomycin, fusidic acid, furazolidone, isoniazid, linezolid, metronidazole, mupirocin, nitrofurantoin, platensimycin, pyrazinamide, quinupristin/dalfopristin combination, and tinidazole.

[0168] Following the initial therapy, a subject may be placed on maintenance therapy in order to maintain reduced methanogen and/or methane levels. In some embodiments, the maintenance therapy utilizes a lovastatin analog or derivative or a modified-release formulation of the present invention. In an embodiment, the initial therapy includes an antibiotic followed by a chronic maintenance regimen of low dose lovastatin analog or derivative formulations. In various embodiments, the maintenance regiment may be administered for at least 1 week, at least 2 weeks, at least 3 weeks, at least 4 weeks, at least one month, at least two months, at least three months, at least four months, at least five months, at least six months, at least seven months, at least eight months, at least nine months, at least ten months, at least eleven months, at least 1 year, at least 2 years, at least 3 years, at least 4 years, at least 5 years, at least 10 years, or indefinitely.

[0169] The anti-methanogenic lovastatin analog or derivative or the modified-release formulation of the present invention may be utilized solely for chronic maintenance therapy. In various embodiments, the present invention provides a method of treating previously methane positive patients who do not have one or more of cardiovascular disease, an LDL level of 190 mg/dL or higher, Type 2 diabetes who are between 40 and 75 years of age, an estimated 10-year risk of cardiovascular disease of 7.5 percent or higher who are between 40 and 75 years of age with an anti-methanogenic lovastatin analog or derivative or a modified-release formulation herein in order to maintain their methane negative status. Accordingly, in some embodiments, the anti-methanogenic lovastatin analog or derivative or the modified-release formulation of the present invention finds use as a prevention measure in a high risk patient.

[0170] In various embodiments, methods of the invention are useful in treatment a human subject. In some embodiments, the human is a pediatric human. In other embodiments, the human is an adult human. In other embodiments, the human is a geriatric human. In other embodiments, the human may be referred to as a patient. In some embodiments, the human is a female. In some embodiments, the human is a male.

[0171] In certain embodiments, the human has an age in a range of from about 1 to about 18 months old, from about 18 to about 36 months old, from about 1 to about 5 years old, from about 5 to about 10 years old, from about 10 to about 15 years old, from about 15 to about 20 years old, from about 20 to about 25 years old, from about 25 to about 30 years old, from about 30 to about 35 years old, from about 35 to about 40 years old, from about 40 to about 45 years old, from about 45 to about 50 years old, from about 50 to about 55 years old, from about 55 to about 60 years old, from about 60 to about 65 years old, from about 65 to about 70 years old, from about 70 to about 75 years old, from about 75 to about 80 years old, from about 80 to about 85 years old, from about 85 to about 90 years old, from about 90 to about 95 years old or from about 95 to about 100 years old. In one embodiment, the human is a child. In one embodiment, the human is a female.

Methods to Determine Methanogen Levels / Diagnostic and Patient Selections

[0172] Intestinal methanogen and/or methane levels can be determined by breath tests that measure breath methane levels. Breath testing may be utilized to identify subjects who are“methane-positive” and who can potentially benefit from methods of the present invention. Further, breath testing can also be used to monitor the efficacy of treatment. Breath testing analysis methods and equipment are known in the art (see, for example, International Patent Publication WO/2014/152754, the entire contents of which are incorporated by reference herein). Examples of such equipment include, for example, the QuinTron BreathTracker gas chromatographic (GC) analyzer or the QuinTron BreathTracker device (QuinTron Instrument Company, Inc., Milwaukee, WI).

[0173] Further, abnormal lactulose breath test results are common in subjects with IBS and therefore the present invention provides for the use of lactulose breath tests in evaluating patients. In some embodiments, a patient is evaluated with a lactulose breath test before and/or after administration with the anti-methanogenic lovastatin analog or derivative or the modified release formulations described herein.

[0174] In general, individuals having a breath methane level of at least about 3 ppm are generally associated with methanogen-associated disorders and are likely to benefit from methods of the present invention. Alternatively, methods of the invention may be practiced on subjects having a breath methane level of at least 1 ppm, at least 1.5 ppm, at least 2 ppm, at least 2.5 ppm, at least 3 ppm, at least 3.5 ppm, at least 4 ppm, at least 5 ppm, at least 6 ppm, at least 7 ppm, at least 8 ppm, at least 9 ppm, at least 10 ppm.

[0175] One method for measuring methanogen levels involves calculation of a subject’s breath methane area under the curve (BM-AUC). This method involves obtaining multiple breath samples averaging about 15 minutes apart for a period of about 90 minutes, or about 120 minutes, or for up to 4 hours or more at potentially less frequent intervals. The time period results are used to calculate a person’s BM-AUC. For example, a subject may undergo a such as lactulose, xylose, lactose, or glucose breath test after a 12 hour fast. The breadth test may comprise a baseline breath measurement after which the subject ingests about 10 g of such as lactulose, xylose, lactose, or glucose. Following lactulose ingestion, the subject is then asked to provide a breath sample about every 15 minutes for about 90 to about 120 minutes to determine methane production. BM-AUC may be utilized for more precisely determining and monitoring, for example, the efficacy of the anti-methanogenic therapy. BM-AUC measurements could also be utilized to segregate“methane positive” from“methane negative” subjects for improved clinical decision making. BM-AUC may be compared to or utilized with measurement of methanogen levels in stool samples via PCR, e.g. qPCR. Alternatively, measurement of methanogen levels in stool samples via PCR, e.g. qPCR may supplant the use of a breath test. More precise techniques may also involve measurement of breath methane taking into account and subtracting ambient methane levels.

[0176] Spot breath methane analysis via commercially available equipment such as BreathTracker may be used in discriminating“methane-positive” from“methane-negative” individuals, and monitoring the success, failure, dose titration, dosing schedule (daily or non-daily, for example) of the anti-methanogenic lovastatin analog or derivative or the modified-release formulation described herein. For example, the lowest minimum effective dose may be identified as such. Additional instruments and techniques for measuring methane levels include, but are not limited to, cavity enhanced absorption techniques such as a LGR-FMR methane measurement instrument having a range as low as 0.01 ppm (Los Gatos Research, Inc., Mountain View, CA), wavelength-scanned cavity down-ring spectroscopy, carbon isotope analysis (G2132-i13C, Picarro, Inc, Santa Clara, CA), gas chromatography, mass spectroscopy, membrane extracted carbon isotope analysis (Pollock, 2012 GSA Annual Meeting,“Membrane Extracted Carbon Isotope Analysis Of Dissolved Methane"), headspace gas chromatography with FID detector and GC combustion with IRMS instruments, for example. Other instruments having the ability to measure low concentration breath methane levels at higher precision than the clinical validated instrument marketed as the QuinTron BreathTracker include high precision breath methane analysis (HPBMA). Use of HPBMA may be used to test spot breath methane levels or in BM-AUC form. [0177] In some embodiments, detection of hydrogen quantity and methane quantity is by gas chromatography with mass spectrometry and/or radiation detection to measure breath emissions of isotope- labeled carbon dioxide, methane, or hydrogen, after administering an isotope-labeled substrate that is metabolizable by gastrointestinal bacteria but poorly digestible by the human host, such as lactulose, xylose, mannitol, or urea (e.g., G. R. Swart and J. W. van den Berg, 13 C breath test in gastrointestinal practice, Scand. J. Gastroenterol. [Suppl.] 225:13-18 [1998]; S. F. Dellert et al., The 13 C-xylose breath test for the diagnosis of small bowel bacterial overgrowth in children, J. Pediatr. Gastroenterol. Nutr.25(2):153-58 [1997]; C. E. King and P. P. Toskes, Breath tests in the diagnosis of small intestinal bacterial overgrowth, Crit. Rev. Lab. Sci. 21(3):269-81 [1984]). A poorly digestible substrate is one for which there is a relative or absolute lack of capacity in a human for absorption thereof or for enzymatic degradation or catabolism thereof.

[0178] Suitable isotopic labels include 13 C or 14 C. For measuring methane suitable isotopic labels can also include 2 H and 3 H or 17 O and 18 O, as long as the substrate is synthesized with the isotopic label placed in a metabolically suitable location in the structure of the substrate, i.e., a location where enzymatic biodegradation by intestinal microflora results in the isotopic label being sequestered in the gaseous product. If the isotopic label selected is a radioisotope, such as 14 C, 3 H, or 15 O, breath samples can be analyzed by gas chromatography with suitable radiation detection means (e.g., C. S. Chang et al., Increased accuracy of the carbon-14 D-xylose breath test in detecting small-intestinal bacterial overgrowth by correction with the gastric emptying rate, Eur. J. Nucl. Med. 22(10):1118-22 [1995]; C. E. King and P. P. Toskes, Comparison of the 1-gram [ 14 C]xylose, 10-gram lactulose-H 2 , and 80-gram glucose-H 2 breath tests in patients with small intestine bacterial overgrowth, Gastroenterol.91(6):1447-51 [1986]; A. Schneider et al., Value of the 14 C -D-xylose breath test in patients with intestinal bacterial overgrowth, Digestion 32(2):86-91 [1985]).

[0179] In various embodiments, treatments using the anti-methanogenic lovastatin analog or derivative or the modified-release formulation of the invention result in a reduction of breath methane level of at least about 1 ppm, at least about 2 ppm, at least about 3 ppm, at least about 4 ppm, at least about 5 ppm, at least about 6 ppm, at least about 7 ppm, at least about 8 ppm, at least about 9 ppm, at least about 10 ppm, at least about 20 ppm, at least about 30 ppm, at least about 40 ppm, at least about 50 ppm, at least about 60 ppm, at least about 70 ppm, at least about 80 ppm, at least about 90 ppm, at least about 100 ppm, at least about 110 ppm, at least about 120 ppm, at least about 130 ppm, at least about 140 ppm, at least about 150 ppm, at least about 160 ppm, at least about 170 ppm, at least about 180 ppm, at least about 190 ppm, at least about 200 ppm, at least about 210 ppm, at least about 220 ppm, at least about 230 ppm, at least about 240 ppm, and at least about 250 ppm.

[0180] The samples used for the present invention include a patient’s breath. In various embodiments, measurement of methanogen levels in stool samples via PCR, e.g. qPCR or other molecular biology approaches, for example, is also provided. Further, aspirates of the fluid in the GI tract may be analyzed for methanogen and/or methane levels. Also mucosal biopsies from a site in the gastrointestinal tract may be analyzed for methanogen and/or methane levels. [0181] Methods of “quantitative” amplification are well known to those of skill in the art. For example, quantitative PCR involves simultaneously co-amplifying a known quantity of a control sequence using the same primers. This provides an internal standard that may be used to calibrate the PCR reaction. Detailed protocols for quantitative PCR are provided in, for example, Innis, et al. (1990) PCR Protocols, A Guide to Methods and Applications, Academic Press, Inc. N.Y.). Measurement of DNA copy number at microsatellite loci using quantitative PCR anlaysis is described in, for example, Ginzonger, et al. (2000) Cancer Research 60:5405-5409. The known nucleic acid sequence for the genes is sufficient to enable one of skill in the art to routinely select primers to amplify any portion of the gene. Fluorogenic quantitative PCR may also be used in the methods of the invention. In fluorogenic quantitative PCR, quantitation is based on amount of fluorescence signals, e.g., TaqMan and Sybr green.

[0182] Other suitable amplification methods include, but are not limited to, ligase chain reaction (LCR) (see, for example, Wu and Wallace (1989) Genomics 4: 560, Landegren, et al. (1988) Science 241:1077, and Barringer et al. (1990) Gene 89: 117), transcription amplification (Kwoh, et al. (1989) Proc. Natl. Acad. Sci. USA 86: 1173), self-sustained sequence replication (Guatelli, et al. (1990) Proc. Nat. Acad. Sci. USA 87: 1874), dot PCR, and linker adapter PCR, etc.

[0183] In still other embodiments of the methods provided herein, sequencing of individual nucleic molecules (or their amplification products) is performed. In one embodiment, a high throughput parallel sequencing technique that isolates single nucleic acid molecules of a population of nucleic acid molecules prior to sequencing may be used. Such strategies may use so-called“next generation sequencing systems” including, without limitation, sequencing machines and/or strategies well known in the art, such as those developed by Illumina/Solexa (the Genome Analyzer; Bennett et al. (2005) Pharmacogenomics, 6:373-20 382), by Applied Biosystems, Inc. (the SOLiD Sequencer; solid.appliedbiosystems.com), by Roche (e.g., the 454 GS FLX sequencer; Margulies et al. (2005) Nature, 437:376-380; U.S. Pat. Nos. 6,274,320; 6,258,568; 6,210,891) and others. Other sequencing strategies such as stochastic sequencing (e.g., as developed by Oxford Nanopore) may also be used, e.g., as described in International Patent Publication No. WO/2010/004273.

[0184] In still other embodiments of the methods provided herein, deep sequencing can be used to identify and quantify the methanogen or methanogen syntrophic microorganism. These techniques are known in the art. Kits

[0185] The present invention is also directed to a kit for the treatment of a methanogen-associated disorder. The kit is an assemblage of materials or components, including at least one of the anti-methanogenic lovastatin analog or derivative or the modified-release formulations described herein. The kit may further include materials and components for the quantification of methanogens. The exact nature of the components configured in the kit depends on its intended purpose. In one embodiment, the kit is configured for the purpose of treating human subjects. [0186] Instructions for use may be included in the kit. Instructions for use typically include a tangible expression describing the technique to be employed in using the components of the kit to affect a desired outcome, such as to treat a disorder associated with methanogens. Optionally, the kit also contains other useful components, such as, diluents, buffers, pharmaceutically acceptable carriers, syringes, catheters, applicators, pipetting or measuring tools, bandaging materials or other useful paraphernalia as will be readily recognized by those of skill in the art.

[0187] The materials and components assembled in the kit can be provided to the practitioner store in any convenience and suitable ways that preserve their operability and utility. For example, the components can be provided at room, refrigerated or frozen temperatures. The components are typically contained in suitable packaging materials. In various embodiments, the packaging material is constructed by well-known methods, preferably to provide a sterile, contaminant-free environment. The packaging material may have an external label which indicates the contents and/or purpose of the kit and/or its components. EXAMPLES

Example 1: Evaluation of Anti-Methanogenic Lovastatin Analogs or Derivatives in Stool Samples of Human IBS- C Patients.

[0188] Fresh stool samples were provided by two methane-producing human IBS-C patients (breath methane > 10 ppm) who had provided an informed consent. Different concentrations of stool from each patient were homogenized in deoxygenated PBS (3 ml PBS/2 g stool) inside an anaerobic incubation chamber. Head gas was extracted through a stopcock at various timepoints (0, 90, 180 and 270 minutes, and following overnight incubation) and methane levels in the head gas at each timepoint were analyzed by gas chromatography using a Quintron SC gas chromatograph (Quintron Instrument Company, Milwaukee, WI). 4 g of stool appeared adequate to capture both methane-producing subjects within an acceptable concentration range.

[0189] The effects of lovastatin lactone, lovastatin ß-hydroxyacid and lovastatin diol on methane production were evaluated by incubating various concentrations of these molecules (0 mg, 0.5 mg, 1 mg, 5 mg and 10 mg of statin per g of human stool) with human stool homogenates in an anaerobic chamber as described above.. Head gas samples were drawn from each flask for methane analysis at baseline and at 90 minute intervals for 3 hours, followed by a final sample after overnight incubation (~720 min).

[0190] Illustrative methane vs. time profiles for human stool samples treated with the different lovastatin species are presented in Figure 4. Lovastatin lactone and lovastatin diol lactone were both effective inhibitors of methane production in human stool; however, lovastatin ß-hydroxyacid was ineffective as an inhibitor of methane production in this system.

[0191] Further studies using this experimental design are used to evaluate the compounds of Formulae Ia-e, IIa-e, IIIa-e, IVa-e, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XVI, XVII, XVIII, XVIX, XXX, XXXI, XXXII, XXXIII, XXXIV, XXXV and XXXVI. Example 2: Animal Studies of Anti-Methanogenic Lovastatin Analogs or Derivatives

[0192] The effects of anti-methanogenic lovastatin analogs or derivatives in a rat model of diet-induced constipation and M. smithii proliferation are evaluated. Adult, male Sprague-Dawley rats are placed on a high fat diet (e.g. about 60% kcal from fat, Teklad high fat diet TD.06414, Harlan Laboratories Inc, Madison, WI) for about 7 weeks. The rats are assessed for increased M. smithii by qPCR before and after the diet and then divided into 3 groups. Group 1 is given anti-methanogenic lovastatin analogs or derivatives (e.g. compounds of Formulae Ia- e, IIa-e, IIIa-e, IVa-e, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XVI, XVII, XVIII, XVIX, XXX, XXXI, XXXII, XXXIII, XXXIV, XXXV, and XXXVI), optionally in the lactone form (about 1.5 mg/rat), group 2 is given anti-methanogenic lovastatin analogs or derivatives (e.g. compounds of Formulae Ia-e, IIa-e, IIIa-e, IVa-e, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XVI, XVII, XVIII, XVIX, XXX, XXXI, XXXII, XXXIV, XXXV, and XXXVI) hydroxy acid (about 1.5 mg/rat), and the Group 3 was gavaged with a placebo. Each group is gavaged daily for about 10 days. Three day stool collections are performed to assess average stool wet-weight and daily variability prior to commencing the high fat diet, after about 7 weeks of high fat diet, and the finals days of the lovastatin gavage (still on high fat diet). On day 10 of the gavage, rats are euthanized and DNA is extracted from contents of ligated bowel segments (ileum, jejunum, duodenum, cecum and left colon). qPCR is performed using primers for total luminal bacteria and M. smithii.

[0193] The studies of Example 2 are optionally carried out using lovastatin diol lactone.

Example 3: Clinical Evaluation of Different Release Profiles

[0194] The present formulations are evaluated clinically as shown in Figure 5. Duodenal and ileocecal release profiles are compared separately and in combination to evaluate any benefit of one over the other or synergy in the combination. Further, Figure 6 shows a clinical study design with the present formulations. Further an evaluation of the pharmacokinetics and breath methane effects of different doses and dosing profiles in methane positive subjects may be undertaken. Such evaluations are optionally of lovastatin diol lactone. Definitions

[0195] As used herein,“a,”“an,” or“the” can mean one or more than one.

[0196] Further, the term“about” when used in connection with a referenced numeric indication means the referenced numeric indication plus or minus up to 10% of that referenced numeric indication. For example, the language“about 50%” covers the range of 45% to 55%.

[0197] An“effective amount,” when used in connection with medical uses is an amount that is effective for providing a measurable treatment, prevention, or reduction in the rate of pathogenesis of a disorder of interest.

[0198] As used herein, something is“decreased” if a read-out of activity and/or effect is reduced by a significant amount, such as by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 98%, or more, up to and including at least about 100%, in the presence of an agent or stimulus relative to the absence of such modulation. As will be understood by one of ordinary skill in the art, in some embodiments, activity is decreased and some downstream read-outs will decrease but others can increase.

[0199] Conversely, activity is“increased” if a read-out of activity and/or effect is increased by a significant amount, for example by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 98%, or more, up to and including at least about 100% or more, at least about 2-fold, at least about 3-fold, at least about 4-fold, at least about 5-fold, at least about 6-fold, at least about 7-fold, at least about 8-fold, at least about 9-fold, at least about 10-fold, at least about 50-fold, at least about 100-fold, in the presence of an agent or stimulus, relative to the absence of such agent or stimulus.

[0200] As referred to herein, all compositional percentages are by weight of the total composition, unless otherwise specified. As used herein, the word“include,” and its variants, is intended to be non-limiting, such that recitation of items in a list is not to the exclusion of other like items that may also be useful in the compositions and methods of this technology. Similarly, the terms“can” and“may” and their variants are intended to be non- limiting, such that recitation that an embodiment can or may comprise certain elements or features does not exclude other embodiments of the present technology that do not contain those elements or features.

[0201] Although the open-ended term“comprising,” as a synonym of terms such as including, containing, or having, is used herein to describe and claim the invention, the present invention, or embodiments thereof, may alternatively be described using alternative terms such as“consisting of” or“consisting essentially of.”

[0202] As used herein, the words“preferred” and“preferably” refer to embodiments of the technology that afford certain benefits, under certain circumstances. However, other embodiments may also be preferred, under the same or other circumstances. Furthermore, the recitation of one or more preferred embodiments does not imply that other embodiments are not useful, and is not intended to exclude other embodiments from the scope of the technology.

[0203] The amount of compositions described herein needed for achieving a therapeutic effect may be determined empirically in accordance with conventional procedures for the particular purpose. Generally, for administering therapeutic agents (e.g., anti-methanogenic lovastatin analog or derivative and/or additional therapeutic agents described herein) for therapeutic purposes, the therapeutic agents are given at a pharmacologically effective dose. A“pharmacologically effective amount,”“pharmacologically effective dose,” “therapeutically effective amount,” or“effective amount” refers to an amount sufficient to produce the desired physiological effect or amount capable of achieving the desired result, particularly for treating the disorder or disease. An effective amount as used herein would include an amount sufficient to, for example, delay the development of a symptom of the disorder or disease, alter the course of a symptom of the disorder or disease (e.g., slow the progression of a symptom of the disease), reduce or eliminate one or more symptoms or manifestations of the disorder or disease, and reverse a symptom of a disorder or disease. Therapeutic benefit also includes halting or slowing the progression of the underlying disease or disorder, regardless of whether improvement is realized.

[0204] Effective amounts, toxicity, and therapeutic efficacy can be determined by standard pharmaceutical procedures in cell cultures, tissue samples, tissue homogenates or experimental animals, e.g., for determining the LD50 (the dose lethal to about 50% of the population) and the ED50 (the dose therapeutically effective in about 50% of the population). The dosage can vary depending upon the dosage form employed and the route of administration utilized. The dose ratio between toxic and therapeutic effects is the therapeutic index and can be expressed as the ratio LD50/ED50. In some embodiments, compositions and methods that exhibit large therapeutic indices are preferred. A therapeutically effective dose can be estimated initially from in vitro assays, including, for example, cell culture assays or measurements or methane production in stool samples. Also, a dose can be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 as determined in cell culture, or in an appropriate animal model. Levels of the described compositions in plasma can be measured, for example, by high performance liquid chromatography. The effects of any particular dosage can be monitored by a suitable bioassay. The dosage can be determined by a physician and adjusted, as necessary, to suit observed effects of the treatment.

[0205] In certain embodiments, the effect will result in a quantifiable change of at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 70%, or at least about 90%. In some embodiments, the effect will result in a quantifiable change of about 10%, about 20%, about 30%, about 50%, about 70%, or even about 90% or more. Therapeutic benefit also includes halting or slowing the progression of the underlying disease or disorder, regardless of whether improvement is realized.

[0206] As used herein,“methods of treatment” are equally applicable to use of a composition for treating the diseases or disorders described herein and/or compositions for use and/or uses in the manufacture of a medicaments for treating the diseases or disorders described herein. EQUIVALENTS

[0207] While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure as come within known or customary practice within the art to which the invention pertains and as may be applied to the essential features hereinbefore set forth and as follows in the scope of the appended claims.

[0208] Those skilled in the art will recognize, or be able to ascertain, using no more than routine experimentation, numerous equivalents to the specific embodiments described specifically herein. Such equivalents are intended to be encompassed in the scope of the following claims. INCORPORATION BY REFERENCE

[0209] All patents and publications referenced herein are hereby incorporated by reference in their entireties.

[0210] The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention.

[0211] As used herein, all headings are simply for organization and are not intended to limit the disclosure in any manner. The content of any individual section may be equally applicable to all sections.