Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS AND DEVICES FOR USING ISOPERILLYL ALCOHOL
Document Type and Number:
WIPO Patent Application WO/2012/083178
Kind Code:
A1
Abstract:
The present invention provides for a method of treating a disease such as cancer, comprising the step of administering to a patient a therapeutically effective amount of an isomer or analog of monoterpene or sesquiterpene (or its derivative), such as an isoperillyl alcohol. The present invention also provides for a method of treating a disease comprising the step of administering to a patient a therapeutically effective amount of a derivative of an isomer or analog of monoterpene or sesquiterpene, such as an isoperillyl alcohol carbamate. The derivative may be an isoperillyl alcohol conjugated with a therapeutic agent such as a chemotherapeutic agent. The route of administration may vary, including inhalation, intranasal, oral, transdermal, intravenous, subcutaneous or intramuscular injection.

Inventors:
CHEN THOMAS (US)
LEVIN DANIEL (US)
PUPPALI SATISH (US)
DICKMAN DANIEL A (US)
Application Number:
PCT/US2011/065513
Publication Date:
June 21, 2012
Filing Date:
December 16, 2011
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
NEO ONCOLOGY INC (US)
CHEN THOMAS (US)
LEVIN DANIEL (US)
PUPPALI SATISH (US)
DICKMAN DANIEL A (US)
International Classes:
C07C35/18
Foreign References:
US20100226913A12010-09-09
US7056491B22006-06-06
US20100226913A12010-09-09
Other References:
EL HOUSSAMEA ET AL.: "Palladium-catalyzed alkoxycarbonylation of allylic natural terpenic functionalized olefins", LABORATOIRE DE CATALYSE DE LILLE, UPRESA CNRS 8010, vol. 168, 4 August 2000 (2000-08-04), ENSC LILLE BP 108, 59652 VILLENEUVE D?ASCQ, FRANCE, pages 15 - 23, XP055108760
WEN ET AL.: "Malignant gliomas in adults", NEW ENGLAND J MED, vol. 359, 2008, pages 492 - 507
STUPP ET AL.: "Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma", NEW ENGLAND J MED, vol. 352, 2005, pages 987 - 996, XP002439490, DOI: doi:10.1056/NEJMoa043330
SCHOBERT ET AL.: "Monoterpenes as Drug Shuttles: Cytotoxic (6-minomethylnicotinate) dichloridoplatinum(II) Complexes with Potential To Overcome Cisplatin Resistance", J. MED. CHEM., vol. 50, 2007, pages 1288 - 1293
GOULD, M.: "Cancer chemoprevention and therapy by monoterpenes", ENVIRON HEALTH PERSPECT, vol. 105, no. 4, June 1997 (1997-06-01), pages 977 - 979
DAS ET AL.: "Design and synthesis of potential new apoptosis agents: hybrid compounds containing perillyl alcohol and new constrained retinoids", TETRAHEDRON LETTERS, vol. 51, 2010, pages 1462 - 1466, XP026894676, DOI: doi:10.1016/j.tetlet.2010.01.003
See also references of EP 2651864A4
Attorney, Agent or Firm:
DAVITZ, Michael, A. (Veltrop & Harkrider LLP114 West 47th Stree, New York NY, US)
Download PDF:
Claims:
What is claimed is:

1. A method for treating a disease in a mammal, comprising the step coadministering to the mammal a therapeutical iy effective amount of an isoperillyl alcohol,

2. The method of claim I, wherein the isoperillyl alcohol is selected from the group

consisting of (4-isopropylidene cyclohex-1-enyl^ethanol, (4-isopropyl cyctohexa- l ,3-dienyl)methanol, (4-isopropyl cyclohexa-1,4-dienyl)methanoi, (4- isopropylphenyl)methano1 and (4-isoproperjyiphenyi)methanol.

3. 'Ihe method of claim 1, wherein the disease is cancer.

4. The method of claim 3 , wherei the cancer is a tumor of the ner vous sy stem,

5. The method of claim 4, wherein the tumor is a gl ioblastoma.

6. The method of claim 1 , further comprising the step of treating the mammal with

radiation,

7. The method of claim 6, wherein the isoperillyl alcohol is administered before, during or after radiation.

8. The method of claim 1 , further comprisin the step of administering to the rnainmal a chemodierapeutic agent

9. The method of claim 8, wherein the isoperillyl alcohol is administered before, during or after the administration of the chemotherapeutic agent.

10. The method of claim 1, wherein the isoperillyl alcohol is administered by inhalation, intranasally, orally, intravenously, subcutaneously or intramuscularly. U. A method for treating a disease in a mammal, comprising the step of administering to the mammal a therapeutically effective amount of an isoperillyl alcohol carbamate.

12. The method of claim 11, wherein the disease is cancer.

13. The meihod of claim 12, wherein the cancer is a rumor of the nervous system.

14. The method of claim 13, wherein the tumor is a glioblastoma. 15. The method of claim 1 1, further comprising the step of treating the mammal with

radiation.

16. The method of claim 1 1 , further comprising the step of administering to the mammal a chemotherapeutic agent.

17. The method of claim 11 , wherein the isoperillyl alcohol carbamate is administered by inhalation, mtranasally, orally, intravenously, subcntaneously or intramuscularly.

15. The method of claim 11, wherein the isoperillyl alcohol carbamate is an isoperillyl alcohol conj ugated with a therapeutic agent.

19. The method of claim 18, wherein the therapeutic agent is a chemotherapeutic agent.

20. The method of claim , wherein the chemoiherapeutic ageni is selected from the group consisting of a DNA alkylating agent, a topoisomerase inhibitor, an endoplasmic reticulum stress inducing agent, a platinum compound, an antimetabolite, an enzyme inhibitor, and a receptor antagonist.

21. The method of claim 18, wherein the therapeutic agent is selected from the group consisting of dimethyl celocoxib (DMC), temo olomide (TMZ) and rolipram.

22. A method for treating a disease in a mammal, comprising the step of administering to the mammal a therapeutically effective amount of an isoperillyl alcohol or an isoperillyl alcohol carbamate using a nasal delivery device.

23. The method of claim 22, wherein the nasal delivery device is selected from the group consisting of an intranasal inhaler, an intranasal spray device, an atomizer, a nebulizer, a metered dose inhaler (MDI), a pressurized dose inhaler, an insufflator, a unit dose container, a pump, a dropper, a squeeze bottle and a bi-directional device.

24. A pharmaceutical composition comprising an isoperillyl alcohol carbamate. 25. The pharmaceutical composition of claim 24, wherein the isoperillyl alcohol carbamate is an isoperillyl alcohol conjugated wiih a therapeutic agent

26. The pharmaceutical composition of claim 25, wherein the therapeutic agent is a

chemotherapeutic agent.

27. The pharmaceutical composition of claim 25, wherein the therapeutic agent is selected froxii the group consisting of dimethyl celocoxib (DMC), temozolomide (TMZ) and rolipram. 28. The pharmaceutical composition of claim 25, wherein the chemotherapeutic agent is selected from the group consisting of a DNA alkylating agent, a topoisomerase inhibitor, an endoplasmic reticulum stress inducing agent, a platinum compound, an antimetabolite, an enzyme inhibitor, and a receptor antagonist.

29. 'Ihe pharmaceutical composition of claim 24, wherein the pharmaceutical composition is administered by inhalation, iiitranasally, orally, intravenously, subcutaneously or intramuscularly, 30. The pharmaceutical composition of claim 24, wherein the pharmaceutical composition is admixed or coformulated with a therapeutic agent.

31. A pharmaceutical composition compri sing an isoperillyl alcohol admixed or

coformulated with a therapeutic agent.

32. A process for making an isoperillyl alcohol carbamate, comprising the step of reacting a first reactant of isoperillyl chJoro formate with a second reactant selected from the group consisting of dimethyl eelocoxib (DMC), temozolomide (TMZ) and rolipram. 33. The process of claim 32, wherein the second reactant is dimethyl eelocoxib.

34. lite process of claim 32, wherein the reaction is carried out in the presence of acetone and a catalyst of potassium carbonate. 35. 'Ihe process of claim 32, wherein the second reactant is rolipram.

36. The process of claim 35, wherein the reaction is carried out in the presence of

tetrahydrofuran and a catalyst of n-butyl lithium. 37. The process of claim 32, wherein .isoperillyl chloroformate is prepared by reacting

isoperillyl alcohol with phosgene.

Description:
METHODS AND DEVICES FOR USING ISOPERILLYL ALCOHOL

Cross Reference to Related Application

This application claims priority to U.S. Provisional Application Nos. 61/424,332 (filed on December 17, 2010) and 61/436,365 (filed on January 26, 2011). These applications are incorporated herein by reference in their entirety.

Field of the Invention

'I¾e present invention relates to isoperiUyl alcohol (iso-POH) and isoperillyl alcohol derivatives. IsoperiUyl alcohot includes any isomers or analogs of periUyl alcohol. The present invention further relates to methods of using isoperiUyl alcohol and isoperiUyl alcohot derivatives such as isoperiUyl alcohol carbamates to treat cancer.

Backgrou nd of the Invention

Malignant gliomas, the most common form of central nervous system (CNS) cancers, are currently considered essentially incurable. Among the various malignant gliomas, anaplastic astrocytomas (Grade III) and glioblastoma multiforme (GBM; Grade IV) have an especially poor prognosis due io their aggressive growth and resistance to currently available therapies. The present standard of care tor malignant gliomas consists of surgery, ionizing radiation, and chemotherapy. Despite recent advances in medicine, the past 50 years have not seen any significant improvement in prognosis for malignant gliomas. Wen et al. Malignant gliomas in adults. New England i Med. 359: 492-507, 2008. Stupp et aL Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. New England J Med. 352: 987-996, 2005.

The poor response of tumors, including malignant gliomas, to various types of chemotherapeutic agents are often due to intrinsic drug resistance. Additionally, acquired resistance of initially well-responding tumors and unwanted side effects are other problems that frequently thwart long-term treatment using chemotherapeutic agents. Hence, various analogues of chemotherapeutic agents have been prepared in an effort to overcome these problems. The analogues include novel therapeutic agents which are hybrid molecules of at least two existing therapeutic agents. For example, cisptai in has been conjugated with cytotoxic codrugs, or conjugated with bioactive shuttle components such as porphyrins, bile acids, hormones, or modulators that expedite the transmembrane transport or the drug accumulati n within the cell. i&-Ammometh} nicotinate) dichloridoplatinum (II) complexes esterirled with terpene alcohols were tested on a panel of human tumor cell lines. The terpenyl moieties in these complexes appeared to fulfill a transmembrane shuttle function and increased the rate and extent of the uptake of these conjugates into various tumor cell lines. Schobert et al. Monoterpenes as Drug Shuttles: Cytotoxic (6-niinomethylnicotinate) dichloridop!atinum(II) Complexes with Potential To Overcome Cisplatin Resistance. J. Med. Chem. 2007, 50, 1288-1293.

Perillyi alcohol (POH), a naturally occurring monoterpene, has been suggested to be an effective agent against a variety of cancers, including CNS cancer, breast cancer, pancreatic cancer, lung cancer, melanomas and colon cancer. Gould, M. Cancer chemoprevention and therapy by monoterpenes. Environ Health Perspect, 97 June; 105 (Suppl 4): 977-979, Hybrid molecules containing both perillyi alcohol and retinoids were prepared to increase apoptosis- inducing activity. Das et al. Design and synthesis of potential new apoptosis agents: hybrid compounds containing perillyi alcohol and new constrained retinoids. Tetrahedron Letters 2010, 51, 1462-1466.

in order to improve performance over perillyi alcohol and its derivatives, there is a need to prepare isomers or analogs including isoperillyl alcohol conjugated with other therapeutic agents, and use this material in the treatment of cancers such as malignant gliomas, as well as other brain disorders such as Parkinson's and Alzheimer's disease. These compounds may be administered alone or in combination with other treatment methods including radiation, standard chemotherapy, and surgery. "The administration can also be through various routes iuchiding intranasal, oral, oral-tracheal for pulmonary delivery, and transdermal-

Summary of the Invention

The invention provides for a method for treating a disease in a mammal, comprising the step of delivering to the mammal a therapeutically effective amount of an isoperillyl alcohol, The invention also pro ides for a method for treating a disease in a mammal, comprising the step of delivering to the mammal a therapeutically effective amount of an isoperillyl alcohol carbamate. The method may further comprise the step of treating the mammal with radiation, and/or further comprise the step of delivering to the mammal a chemotherapeutic agent. The diseases treated may be cancer, including a tumor of the nervous system, such as a glioblastoma. The routes of administration include inhalation, intranasal, oral, intravenous, subcutaneous or intramuscular administration.

The present invention further provides for a pharmaceutical composition comprising an isoperillyl alcohol carbamate. The isoperillyl alcohol carbamate may be isoperillyl alcohol conjugated with a therapeutic agent, such as a chemotherapeutic agent. The chemotherapeutic agents that may be used in the present invention include a DNA alkylating agent, a

topoisomerase inhibitor, an endoplasmic reticulum stress inducing agent, a platinum compound, an antimetabolite, an enzyme inhibitor, and a receptor antagonist. In certain embodiments, the therapeutic agents are dimethyl celocoxib (DMC), temozolomide (TMZ) or rolipram. The present invention provides for a pharmaceutical composition comprising an isoperillyl alcohol admixed or c formulated with a therapeutic agent. The pharmaceutical compositions of die present invention may be administered before, during or after radiation. The pharmaceutical compositions may be administered before, during or after the administration of a

chemotherapeutic agent.

The present invention also provides for a process for making an isoperillyl alcohol carbamate, comprising the step of reacting a first reactant of isoperillyl chloroformate with a second reactant, which may be dimethyl celocoxib (DMC), temozolomide (TMZ) or rolipram. When the second reactant is dimethyl celocoxib, the reaction may be carried out in the presence of acetone and a catalyst of potassium carbonate. When the second reactant is rolipram, the reaction may be carried out in the presence of tetrahydrofuran with n-butyl lithium. The isoperillyl chloroformate may also be prepared by reacting isoperillyl alcohol with phosgene. Brief Description of the Drawings

Figure 1 shows die results of the MTT cytotoxicity assays demonstrating the efficacy of different types of POH and iso-POH in killing LN229 human glioma cells.

Figure 2 shows the results of the MTT cytotoxicity assays demonstrating the efficacy of different types of POH and iso-POH in killing U251 human glioma celts.

Figure 3 shows the results of the MTT cytotoxicity assays demonstrating the efficacy of different types of POH and iso-POH in killing A 172 human glioma cells.

Figures 4A and 4B show the results of the MTT cytotoxicity assays demonstrating the efficacy of different types of POH and iso-POH in killing Al 72 human glioma cells (temozolomide- sensitive) (Figure 4A) and Al 72 temozoiomide-resistant ceils (Figure 4B). SGP-527-155 is the POH purified to the GLP quality (with a GC relative area purity (area under the curve) of about 98.7%). SGP-561-79P and SGP-561-65P are two different batches of Iso-POH. SGP-561-79P is more purified than SGP-561 -65P and the details are as follows.

Note: Due to the decomposition of the sample by the GC analysis, we have analyzed the Iso- POH by HPLC and compared it with Sigma-Aldrich POH as a standard for wt% assay. Sigma POH is the POH purchased from Sigma Chemicals. Figures 5A and 5B show the results of the MTT cytotoxicity assays demonstrating the efficacy of different types of POH and iso-POH in killing U251 human glioma cells (temoaioiomide- senshive) (Figure 5 A) and U2 1 temozolomide-resisiant cells (Figure 5B). Figures 6A and 6B show the results of the MTT cytotoxicity assays demonstrating the efficacy of different types of POH arid iso-POH in killing LN229 human glioma cells (temozolomide- sensitive) (Figure 6A) and L 229 te ozolomide-resistant celts (Figure 6B).

Figures ?A and 7B show the results of the. MTT c totoxicity assays demonstrating the efficacy of different types of POH and iso-POH in killing U87 human glioma cells (temozolomide- sensitive) (Figure 7 A) and U87 teniozoiomide-resistant ceils (Figure 7B).

Figures 8 A , 8B and 8C show the results of the MTT cytotoxicity assays demonstrating the efficacy of different types of POH and iso-POH in killing U251 human glioma cells

(iemozoiomide-sertsitive) (Figure 8A) and U251 temozolomide-resistant cells (Figures 8B and 8C). U251-TR1 and U25t -TRl refer to two temozolomide-resistant U251 cell lines. Sigma POH is the POH purchased from Sigma Chemicals. GLP POH is the POH purified to the GLP quality (with a GC relative area purity (area under the curve) of about 98.7%). _so~POH65 and Iso-POH79 are different batches of iso-POH (see details above for Figures 4 A. and 4B).

Figure 9 shows the results of the MTT cytotoxicity assays demonstrating the efficacy of POH and iso-POH in killing USC04 glioblastoma cancer stem cell line.

Figures I OA and J OB show Western blot performed after an 18-hour treatment of U25.1 glioma cells with Sigma POH (1 .5mM, having a purity of about 95% (AUC» or ultrapure POH ("CLP- POH" ( 1.5 raM; having a purity of about 98.7% (AUC)) in both U251 T Z-sensitive and TMZ- resistant (U251-TR1, U251-TR2) ceils demonstrating increased expression of glucose-regulatory protein 78 (GRP-78) and the apoptosis marker CHOP, showing increased endoplasmic reticulum (ER) stress after treatment (Figure 10A). Under the same conditions, iso-POH (iso-POH65, iso- POH79) also increased ER stress (Figure lOB). Figure 1 1 shows Western blot performed after a 24-hour treatment of U251 glioma cells with. 500 μΜ Sigma POH, GLP POH or iso-POH (isoPOH65, isaPOH79) demonstrating that alt treatments tlecreasetl Kras expression.

Detailed Description of the Invention

The present invention provides for methods of treating a disease such as cancer using an isoperillyl alcohol or a derivative of an isoperillyl alcohol. Routes of administration include inhalation, intranasal, oral, transdermal, intravenous, subcutaneous and inttamascular injection. in the present methods, a patient is administered a therapeutically effective amount of an isomer or analog of monoterpene or sesquiterpene, such as an isoperillyl alcohol. The present invention also provides for a method of treating a disease comprising the step of administering to a patient a therapeutically effective amount of a derivative of an isomer or analog of

monoterpene or sesquiterpene, such as an isoperillyl alcohol carbamate. The derivative may be a isoperillyl alcohol conjugated with a therapeutic agent such as a che otherapeutic agent.

For example, the i somer or analog of monoterpene or sesquiterpene can be an isoperillyl alcohol (iso-POH). Isoperillyl alcohols include any isomers or analogs of perillyl alcohol, In one embodiment, the isoperillyl alcohol is (4-isopropyiidene cyclohex-1 -enyl)methanol. Other examples of isoperillyl alcohol include, but are not limited to, (4-isopropyi cyclohexa-1 ,3- dienyi)methanol, (4-isopropyt cyclohexa-l ^-dieny methanol, (4-isopropylphenyi)methanol and (4-isopropenylphenyl)methanol.

A exemplary isoperillyl alcohol, (4-isopropyltderte cyclohex- 1 -enyl)methanoI, is shown below:

The compounds of the present invention may be used for the treatment of nervous system cancers, such as a malignant glioma (e.g., astrocytoma, anaplastic astrocytoma, glioblastom multiforme), retinoblastoma, pilocytic astrocytomas (grade I), meningiomas, metastatic brain tumors, neuroblastoma, pituitary adenomas, skull base meningiomas, and skull base cancer. The present invention also provides methods of treating CNS (central nervous system) disorders, including, without limitation, primary degenerative neurological disorders such, as Alzheimer's, Parkinson's, psychological disorders, psychosis and depression.

Also encompassed by the present invention is a derivative of an isomer or analog of monoterpene or sesquiterpene, such as an isoperiHyl alcohol derivative, For example, the isoperillyt alcohol derivative may be an isoperiHyl alcohol carbamate, ester, or ether. The derivative of an isomer or analog of monoterpene or sesquiterpene may be an isomer or analog of monoterpene or sesquiterpene conjugated with a therapeutic agent such as a chemotherapeutic agent. The isoperiHyl alcohol derivative may be isoperiHyl alcohol conjugated with a therapeutic agent such as a chemotherapeutic agent.

The compounds of the present invention thus include both isomers or analogs of monoterpene or sesquiterpene, and derivatives of an isomer or analog of monoterpene or sesquiterpene. The isomer or analog of monoterpene or sesquiterpene (or the derivative of an isomer or analog of monoterpene or sesquiterpene), may be formulated into a pharmaceutical composition, where the isomer or analog of monoterpene or sesquiterpene (or the derivative of an isomer or analog of monoterpene or sesquiterpene), is present in amounts ranging from about 0.01% (w/w) to about 100% (w/w), from about 0.1% (w/w) to about 80% (w/w), from about 1% (w/w) to about 70% (w/w), from about 10% (w/w) to about 60% (w/w), or from about 0.1 % (w/w) to about 20% (w/w). The present compositions can be administered alone, or may be coadministered together with radiation or another agent (e.g., a chemotherapeutic agent), to treat a disease such as cancer. Treatments may be sequential, with isomer or analog of monoterpene or sesquiterpene (or the derivative of an isomer or analog of monoterpene or sesquiterpene) bein administered before or after the administration of other agents. For example, an isoperiHyl alcohol (or an isoperiHyl alcohol carbamate, ester, or ether) may be used to sensitize a cancer patient to radiation or chemotherapy. Alternatively, agents may be administered concurrently. The unite of administration may vary, and can include, inhalation, intranasal, oral, transdermal, intravenous, subcutaneous or intramuscular injection.

The compositions of the present in ention may contain one or more types of isomers or analogs of monoterpene or sesquiterpene (or the derivatives of isomers or analogs of

monoterpene or sesquiterpene), Monoterpenes include terpenes that consist of two isoprene units. Monoterpenes may be linear (acyclic) or contain rings. Derivatives of monoterpenoids are also encompassed by the present invention. Monoterpenoids may be produced by biochemical modifications such as oxidation or rearrangement of monoterpenes. Examples of moiioierpenes and monoterpenoids include, perillyl alcohol (S(-)) and (R(+)) » ocimene, myrcene, geraniol, citrai, citroneiloi, citroneilal, tinalool, pinene, terpineol, terpinen, limonene, terpmenes, phellandrenes, terpinoJene, terpinen-4-ol (or tea tree oil), pinene, terpineol, terpinen; tlie terpenoids such as p-cy nene which is derived from monocyclic terpenes such as menthol, thymol and carvacrot; bicyctic monoterpenoids such as camphor, borneol and eucalyptol.

Monoterpenes may be distinguished by the structure of a carbon skeleton and may be grouped into acyclic monoterpenes (e.g., myrcene, (Z> and (E)-ocimene, Imalooi, geranioi, nerol, citroneltol, myrcenol, geraniat, cirral a, neral, citral b, cifronellal, etc.), monocyclic monoterpenes (e.g., Hmonene, terpinene, phellandrene, terpinolene, menthol, carveol, etc.), bicyclie monoterpenes (e.g., pinene, myrtenol, myrtenal, verbanol, verbanon, pinocarveol, carene, sabinene, camphene, thujene, etc.) and tricyclic monoterpenes (e.g. tricyclene). See Encyclopedia of Chemical Technology. Fourth Edition, Volume 23, page 834-835.

Sesquiterpenes of the present invention include terpenes that consist of three isoprene units. Sesquiterpenes may be linear (acyclic) or contain rings. Derivatives of sesquiterpenoids are also encompassed by the present invention. Sesquirerpenoids may be produced by biochemical modifications such as oxidation or rearrangement of sesquiterpenes. Examples of sesquiterpenes include farnesol, farnesal, farnesylic acid and nerolidoi. U.S. Provisional Application Nos. 61/310,231 (filed on March 3, 2010), 61/377,747 (filed on August 27, 2010), 61/471,402 (filed on April 4, 201 1) and 61/562,105 (filed on November 21 , 201 1). PCT Application Nos. PCT US20H/027051 (filed on March 3, 201 1) and PCT US2011/049392 (filed on August 26, 201 1 ). U.S. Application No. 13/040,059 (filed on March 3, 201 1 ). All these applications ate incorporated herein by reference in their entirety,

The derivatives of isomers or analogs of monoterpene or sesquiterpene include, but are not limited to, carbamates, esters, ethers, alcohols and aldehydes of the monoterpene (or sesquiterpene). Alcohols may be derivatked to carbamates, esters, ethers, aldehydes or acids.

Carbamate refers to a class of chemical compounds sharing the functional group

ased on a carbonyi group flanked by an oxygen and a nitrogen. R L . 2 and "' can be a group such as a!kyl, aryl, etc., which can e substituted. The . groups on the nitrogen an the oxygen may form a ring. R'-GH may be a monoterpene. e.g., POH. The R^- - ' moiety may be a therapeutic agent.

Carbamates may be synthesized by reacting isocyanate and alcohol, or by reacting chloroformate with amine. Carbamates may be synthesized by reactions making use of phosgene or phosgene equivalents. For example, carbamates may be synthesized by reacting phosgene gas, diphosgene or a solid phosgene precursor such as triphosgene with two amines or an amine and an alcohol. Carbamates (also known as ureihanes) can also lie made from reaction of ure intermediate with an alcohol. Dimethyl carbonate and di phenyl carbonate are also used for making carbamates. Alternatively, carbamates may be synthesized through the reaction of alcohol and/or amine precursors with an ester -substituted diary! carbonate, such as

bismethylsaiicyicai' mate (BMSC). U.S. Patent Publication No. 201001 1.381.9.

Carbamates may be synthesized by the following approach:

Suitable reaction solvents include, but are not limited to, tetrahydK>furan 5 dicb.loromethane, dichloroethane, acetone, and diisopropyS etiier. The reaction may be performed at a temperature ranging from about -7Q"C to about 80"C, or from about -65"C to about 50"C. The molar ratio of isoperillyi chloroformate to the substrate R ~ Ntな may range from about 1 ; 1 to about 2: 1, from about 1 : 1 to about 1.5 : 1 5 from about 2 : 1 to about 1:1 , or from about 1.05 : 1 to about 1 ,1 : 1.

Suitable bases include, but are not limited to, organic bases, such as triethylamine, potassium carbonate, iN^ '-diisopiOpyletliylamine, butyl lithium, and potassium- t-butoxide.

Alternatively, carbamates may be synthesized by the following approach:

Suitable reaction solvents include,, but are not limited to, dichloro et ane, dichloroethane, toluene, diisopropyl ether, and tetrahydrofwan, The reaction may be performed at a temperature ranging from about 25X to about 1 lt C, or from about 30°C to about 8( C, or about 50な. The molar ratio of isoper.ill.yi alcohol to the substrate -N^C-Q may range from, about 1 : 1 to about 2:1 , from about 1 :1 to about 1 .5: 1, from about 2: 1 to about 1 :1 , or from about 1 .05: 1. to about, 1.1 :1.

Esters of the alcohols of the isomers or analogs of monoterpen or sesquiterpen can be derived from an inorganic acid or an organic acid, inorganic acids include, but are not limited to, phosphoric acid, sulfuric acid, and nitric acid. Organic acids include, but are not limited to, carboxylic acid such as benzoic acid, fatty acid, acetic acid and propionic acid, and any therapeuti agent, bearing at least one carboxylic acid unctional group. Examples of the esters of alcohols include, but are not limi ted to, carboxylic acid esters (such as bei oate esters, fatty acid esters (e.g., pal nutate ester, 1 trioleate ester, stearate ester, butyry) ester and oleate ester), acetates, propionates (or propanoates), and formates), phosphates, sulfates, and carbamates (e.g., N,N- dimethylaminocarbotryl). Wikipedia - Ester. Retrieved from URL:

The derivatives of isoperillyi alcohol include isoperillyi alcohol carbamates, isoperillyi alcohol esters, isoperiilic aldehydes, isoperiilic acid, isoperiilic aldehyde derivatives, and isoperiilic acid esters. The derivatives of isoperillyi alcohol may also include its oxidative and nucleophilic/electrophilic addition derivatives. Few examples of derivatives of isoperillyi alcohol are '.reported in the chemistry literature, See U.S. Patent No. 5,994,598 and Japanese Paten No. 07048264A. in certain embodiments, an iso-POH carbamate is synthesized by a process comprising the step of reacting a first reactant of isoperillyi chloroformate wi th a second reactant such as dimethyl celocoxib (DMC), temozoiomide (TMZ) and rolipram. The reaction may be carried out in the presence of tetrahydrofuran and a base such as n-butyl lithium. Isoperillyl chloroformate may be made by reacting iso-POH with phosgene. For example, iso-POH conjugated with temozoiomide through a carbamate bond may be synthesized by reacting temozoiomide with oxalyl chloride followed by reaction with isoperillyl alcohol. The reaction may be carried out in the presence of ,2-dich.oroethane.

Iso-POH carbamates encompassed by the present invention include, but are not limited to, (3-Methyl 4^xo-3,4-dihydroimidazo[5,l iHl A3,5]te^ acid -4- isopropylidene cyclohex-1-e imethyl ester, 4-(3-Cyciopenryloxy-4-methoxyphenyl)-2-oxo- pyrrolidine- 1-carboxylic acid 4-isopropylidene cyclohex-1-enylmethyl ester, 4-(Bis-N.,N'-4- isopropylidene cyclohex -enylmethyloxy carbonyi [5-{2 ? 5-di.methyl phenyl)-3-ttifluoromethyl pyrazol-1-yl] benzenesuifonamide. The details of the chemical reactions generating these compounds are described in the Examples below, In certain embodiments, iso-peri!iyl alcohol derivatives may be isoperillyl alcohol fatty acid esters, such as palmitoyl ester of iso-POH and linoieoyl ester of iso-POH.

The derivative of an isomer or analog of monoterpene or sesquiterpene may be an isomer or analog of monoterpene or sesquiterpene conj gated with a therapeutic agent. A conjugate encompassed by the present invention is a molecule having an isomer or analog of monoterpene or sesquiterpene covaJently bound via a chemic l linking group to a therapeutic agent. The molar ratio of the isomer or analog of monoterpene or sesquiterpene to the therapeutic agent in the conjugate may be 1 : 1 , 1 :2, 1 :3, 1 :4, 2: 1.„ 3; 1 , 4: 1 , or any other suitable molar ratios. The isomer or analog of monoterpene or sesquiterpene and the therapeutic agent may be covaiently linked through carbamate, ester, ether bonds, or any other suitable chemical functional groups. When the isomer or ana log of monoterpene or sesquiterpene and the therapeutic agent are conjugated through a carbamate bond, the therapeutic agent may be any agent bearing at least one carboxylie acid functional group, or any agent bearing at least one amine functional group. In a specific example, an isoperillyl alcohol conjugate is isoperillyl alcohol covaiently bound via a chemical linking group to a chemotherapeutic agent. According to the present invention, the therapeutic agents that may be conjugated with an isomer or analog of monoterpene or sesquiterpene include, but are not limited to,

chemotherapeutic agents, therapeutic agents for treatment of CNS disorders (including, without limitation, primary degenerative neurological disorders such as Alzheimer's, Parkinson' s, multiple sclerosis, Attention-Deficit Hyperactivity Disorder or ADHD, psychological disorders, psychosis and depression), immunotberapeutic agents, angiogenesis inhibitors, and antihypertensive agents. Anti-cancer agents that may be conjugated with monoterpene or sesquiterpene can have one or more of the following effects on cancer cells or the subject: celi death; decreased celi proliferation; decreased numbers of cells; inhibition of cell growth;

apoptosis; necrosis; mitotic catastrophe; cell cycle arrest; decreased cell size; decreased cell division; decreased cell survival; decreased celi metabolism; markers of cell damage or cytotoxicity; indirect indicators of cell damage or cytotoxicity such as rumor shrinkage;

improved survival of a subject; or disappearance of markers associated with undesirable, unwanted, or aberrant cell proliferation, US. Patent Publication No. 20080275057.

Also encompassed by the present invention are admixtures and/or coformulations of an isomer or analog of monoterpene or sesquiterpene and at least one therapeutic agent.

Chemotherapeutic agents include, but are not limited to, DNA alkylating agents, topoisomerase inhibitors, endoplasmic reticulum stress inducing agents, a platinum compound, an antimetabolite, vinealkaloids, taxanes, epothilones, enzyme inhibitors, receptor antagonists, tyrosine kinase inhibitors, boro radiosensitizers (i.e. velcade), and chemotherapeutic combination therapies.

Non-limiting examples of DNA alkylating agents are nitrogen mustards, such as Cyclophosphamide (Ifosfamide, Trofosfamide), Chlorambucil (Melphalan, Prednimustine), Bendamustine, Uramustine and Estramustine; nitrosoureas, such as Carmustine (BCNU), Lomustine (Semustine), Fotemustine, Nimustine, Ranimustine and Streptozocin; alky] sulfonates, such as Busuifan (Mannosul an, Treosuifan); Aziridines, such as Carboquone, Triaziquone, TriethyJenemelamine; Hydrazines (Procarbazine); Triazenes such as Dacarbazine and Temoaolomide; Altretamine and itobronitol.

Non-limiting examples of Topoisomerase I inhibitors include Campothecin derivatives including SN-38, APC, NPC, campothecin, topotecan, exatecan mesylate, 9-mtiOcamptothecm, 9-aminocamptotl.iecin, lurtotecan, rubitecan, silatecan, gimatecan, diflomotecan, extatecan, BN- 80927, DX-8951f, and MAG-CPT as decribed in Pommier Y. (2006) Nat Rev. Cancer

6(10):789~802 and U.S. Patent Publication No. 200510250854; Pratobetberine alkaloids and derivatives thereof including berberrubine and coralyne as described in Li et al. (2000)

Biochemistry 39(24): 7107-7.1 6 and Gatto et al. (1996) Cancer Res. 15(12):2795-2800;

Phenanthroline derivatives including Benzo[i]phenanmridme, Nittdiae, and fagaronine as described in Makhey et al, (2003) Bioora, Med. Chem. 11 (8); 1809-1820; Terbenzimidazole and derivatives thereof as described in Xu (1998) Biochemistry 37( 10):3558-3566; and

AnthracyeUne derivatives including Doxorubicin, Daunorubicin, and Mitoxantrone as described in Foglesong et al. (1992) Cancer Chemother. Pharmacol. 30{2):123-]25, Crow et al, (1994) L Med. Chem. 37(19):31913194, and Crespt et al. (1986) Biochem. Biophvs. Res. Commun.

136(2):521 -8. Topoisomerase Π inhibitors include, but are not limited to Etoposide and

Teniposide. Dual topoisomerase ί and 11 inhibitors include, but are not limited to, Saintopm and other Naphthecenediones, DACA and other Acridine^ arboxamindes s Intoplicme and other Benzopyridoindoles, TAS-I03 and other 7H-indeno[2 J l-c]Quinoline-7-ones, Pyrazotoacridine, XR 11576 and other Benzophenazines, XR 5944 and other Dimeric compounds, 7-oxo-7H- dibenz{f,ij]l$oqumoiines and 7 >xo-7H-berizo[e]pyrimi iines, and Anihracenyl-amtno Acid Conjugates as described in Denny and Baguley (2003) Curr. Top. Med, Chem. 3(31:339-353. Some agents inhibit Topoisomerase 11 and have DNA intercalation activit such as, but not limited to, Anthracyclin.es (Aciarubicm, Daunorubicin, Doxorubicin, Epirubicin, idarabiein, Axnrubicitt, Pirarub cin, Valrubiciii, Zorubiciii) and Antracenediones (Mitoxantrone and

Pixantrone).

Examples of endoplasmic reticulum stress inducing agents include, but are not limited to, dimethyl-celecoxib (D C), nelfmavir, celeooxib, and boron radiosensitizers (i.e. velcade (Bortezomib)).

Platinum based compounds are a subclass of DNA alkylating agents. Non-limiting examples of such agents include Cisplatin, Nedap latin, Oxalipiatin, Triplatin teiranitrate, Satraplatin, Aroplatin, Lobaplatin, and JM-216. (see c eage et al. (1997) J. Citn. Oncol 201 :1232-1237 and i general, CHEMOTHERAPY FOR GYNECOLOGICAL NEOPLASM, CURRENT THERAPY AND NOVEL APPROACHES, in the Series Basic and Clinical Oncology, Angioli et al. Eds., 2004). "FOLFQX" is an abbreviation for a type of combination therapy that is used to treat colorectal cancer. It includes 5-FU, oxaiipiatin and leucovorin. Information regarding this treatment is available on the Nati nal Cancer Institute's web site, cancer.gov, last accessed on January 1.6, 2008.

"FOLFQX/B V" is an abbreviation for a type of combination therapy that is used to treat colorectal cancer, This therapy includes 5-FU, oxaiipiatin, leucovorin and Bevacizumab.

Furthermore, "XELOX/BV* is another combination therapy used to treat colorectal cancer, which includes the prodrug to 5-FU, known as Capecitabine (Xeloda) in combination with oxaiipiatin and bevacizumab. Information regarding these treatments are available on the National Cancer institute's web site, cancer.gov or from 23 the National Comprehensive Cancer Network's web site, nccn.org, last accessed on May 27,2008.

Non-limiting examples of antimetabolite agents include Folic acid based, i.e.

dihydrofolate reductase inhibitors, such as Aminopterin, Methotrexate and Pemetrexed;

thymtdylate synthase inhibitors, such as Raltitrexed, Pemetrexed; Purine based, i.e. an adenosine deaminase inhibitor, such as Pentostatm, a thiopurine, such as Thioguanine and Mercaptopurine, a haiogenated/ribonucieotide reductase inhibitor, such as Cladribine, Clofarabtne, Fludarabine, or a guanine/guanosine: th opurine, such as Thioguanine; or Pyrimidine based, i.e.

cytosine/cytidine: hypomethylating agent, such as Azacitidine and Decitabine, a DNA polymerase inhibitor, such as Cytarabine, a ribonucleotide reductase inhibitor, such as

Gemcitabine, or a thymine/thymidine: thymidylate synthase inhibitor, such as a Fluorouracil (5- FU). Equivalents to 5-FU include prodrugs, analogs and derivative thereof such as 5' -deoxy-5- fluorouridine (doxifluroidine), i-tetrahydrofurauyi-5-.fluorouracil (ftorafur), Capecitabine (Xeloda), S-I (MBMS-24761.6, consisting of tegafur and two modulators, a 5-chIoro-2,4- dihydroxypyridine and potassium oxonate), mliriuexed (to udex), nolatrexed (Thymitaq, AG337), LY231514 and ZD 33i , as described for example in Papamieheal (1999) The

Oncologist 4:478-487.

Examples of vincaikaloids, include, but are not limited to Vinblastine, Vincfistine, Vinflunine, Vindesine and Vmorelbine.

Examples of taxanes include, but are not limited to docetaxel, Larotaxet, Ortataxel, Paclitaxel and Tesetaxel. An example of an epothilone is tabepilone. Examples of enzyme inhibitors include, but are not limited to farnes ltransferase inhibitors (Tipifarnib); CDK inhibitor (Alvocidib, Seliciciib); proteasome inhibitor

(Bort&comib); phosp diesterase inhibitor (Anagrelide; rolipram); IMP dehydrogenase inhibitor (Tiazofurine); and lipoxygenase inhibitor (Masoprocol). Examples of receptor antagonists include, but are not limited to ERA (Atrasentan); retinoid X receptor (Bexarotene); and a sex steroid (Testolactone).

Examples of tyrosine kinase inhibitors include, but are not limited to inhibitors to £rbB: HERl EGFR (Erlotinib, Gefttinib, Lapatinib, Vandetanib, Sunitinib, Neratinib); HER2/neu (Lapatinib, Neratinib); RT class 111: C-kit (Axitinib, Sunitinib, Sorafenib), FLT3 (Lestaurtinib), PDGFR (Axitinib, Sunitinib. Sorafenib); and VEGFR (Vandetanib, Semaxanib, Cediranib,

Axitinib, Sorafenib); bcr-abl (Imatinib, Nilotinib, Dasatinib); Src (Bosutinib) and Janus kinase 2 (Lestaurtinib).

"Lapatinib" (TykerbD) is a dual EGFR and erbB-2 inhibitor, Lapatinib has been investigated as an anticancer monotherapy, as well as in combination with trastuzumab, capecitabine, tetrozole, paclitaxel and FOLFIRI (irinotecan, 5-fluorouracil and leucovorin), in a number of clinical trials, it is currently in phase ΪΠ testing for the oral treatment of metastatic breast, head and neck, lung, gastric, renal and bladder cancer.

A chemical equivalent of lapatinib is a small molecule or compound that is a tyrosine kinase inhibitor (ΤΚΪ) or alternatively a HER-1 inhibitor or a HER-2 inhibitor. Several TKIs have been found to have effective antitumor activity and have been approved or are in clinical trials. Examples of such include, but are not limited to, Zactima (ZD6474), Iressa (gefttinib), imatinib mesylate (ST157 ; Gieevec), erlotinib (OSI- 774; Tarceva), cauertinib (CI 1033), semaxinib (SU5 1.6), vatalanib (FTK787/ZK222584), sorafenib (BAY 43- 9006), sutent (SUI 1248) and lefitmomide (SUl OI).

PT /ZK is a tyrosine kinase inhibitor with broad specificity that targets ail VEGF receptors (VEGFR), the platelet-derived growth factor (PDGF) receptor, c-ΚΠ and c-Fms. Drevs (2003) Idrugs 6(8):787-794. PTK Z is a targeted drug that blocks angiogenesis and lymphangiogenesis by inhibiting the activi ty of all known receptors that bind VEGF including VEGFR-I (Fit- 1), VEGFR-2 (KDR/Flfc-1) and VEGFR-3 (KM). The chemical names of P K/ZK are l~[4-Chioroanilino]-4-[4-pyridyImetiiyl] phthalazine Succinate or 1 -

Phthalazinamine, N-(4-chlorophenyl)-4^4-pyTiditrylmediyl)-buiat5edioate (1: 1). Synonyms and analogs of PT T are known as Vatalanib, CGP7 787D, PT 787/Z 222584, CGP-79787, DE-00268, PTK-787, PTK787A, VEGFR-TK inhibitor, ZK 222584 and ZK,

Chemotherapeutic agents that can be used in admixtures and/or cofoTmulatio s and/or conjugated with an isomer or analog of monoterpene or sesquiterpene may also include amsacrine, Trabectedin, retinoids (Atitretinoin, Tretinoin), Arsenic trioxide, asparagine depleter Asparaginase Pegaspargase), Celecoxib, Demecolcine, Elesclomol, Elsamitrucin, Etogiucid, Lonidamine, Lucanthone, Mitoguazone, Mitotane, Oblimersen, Temsirotimus, and Vorinostat.

An isomer or analog of monoterpene or sesquiterpene may be conjugated and/or use in admixtures and'Or coformulations with angiogenesis inhibitors. Examples of angiogenesis inhibitors include, but are not limited to, angiostarin, angiozyme, antithrombin HI, AG3340, VEGF inhibitors, batimastat, bevacizumab (avastin), BMS -2752 1, CAI, 2C3, HuMV833 Canstatin, Captopril, carboxyamidotriazole, cartilage derive inhibitor (CD1), CC-5013, 6-0- (chloroacetyl-carbonyl^tknagillol, COL-3, combretastarin, combreiastatin A4 Phosphate, Dalteparin, EMD 121974 (Cttengitide), endostatin, erlotinib, gefitinib (Iressa), genistein, halofuginone hydrobtximide, Idl, Id3, 1M862, imatinib mesylate, IMC-iCl 1 inducible protein 10, interferon-alpha, interieukin 12, lavendustin A, LY317615 or AE-941, marimastat, mspin, medroxpregesterone acetate, eth-1 , Meth-2, 2-methoxyestradiot (2 -ME), neovastat, oteopontin cleaved product, PEX, pigment epithelium growth factor (PEGF), platelet factor 4, prolactin n^gment, proliterin-related protein (PRP), PTK787/Z 222584, ZD6474, recombinant human platelet factor 4 (rPF4), restin, squalamine, SU5416, SU666S, SU11248 suramin, Taxol,

Tecogaian, thalidomide, thrombospondin, TNP-470, iroponin-l, vasostatin, VEGl, VEGF-Trap, and ZD6474.

Non-limiting examples of angiogenesis inhibitors also include, tyrosine kinase inhibitors, such as inhibitors of the tyrosine kinase receptors Flt-1 (VEGFRl) and Flk-l/KDR (VEGFR2), inhibitors of epidermal-derived, fibroblast-derived, or platelet derived growth factors, MMP (matrix metal loprotease) inhibitors, i tegrin blockers, pentosan polysulfate, angiotensin II antagonists, cyclooxygenase inhibitors (including non-sterotdai anti-intlammatory drugs (NSAIDs) such as aspirin and ibuprofen, as well as selecti e cyclooxyge.nase-2 inhibitors such as celecoxib and rofecoxib), and steroidal anti-mflammatories (such as corticosteroids,

mineralocoriicoids, dexamethasone, prednisone, prednisolone, methylpred, betamethasone). Other therapeutic agents that modulate or inhibit angiogenesis and may also be conjugated and/or admixed and/or coformulated with an isomer or analog of moiioterpene or sesquiterpene include agents that modulate or inhibit the coagulation and fibrinolysis systems, including, but not limited to, heparin, low molecular weight heparins and carboxypeptidase U inhibitors (also known as inhibitors of active thrombin activatabte fibrinolysis inhibitor

[TAFIa]), U.S. Patent Publication No. 20090328239. U.S. Patent No. 7,638,549,

Non-Hmiting examples of the anti-hypertensive agents include angiotensin converting enzyme inhibitors (e.g., captopril, enalapri!, delapril etc.), angiotensin 11 antagonists (e.g., candesartan cilexetil, candesartan, losartan (or Cozaar), losartan potassium, eprosartan, valsartan (or Diovan), termisartan, irbesartan, tasosartan, olmesartan, olmesartan medoxomtl etc.), calcium antagonists (e.g., manidipine, nifedipine, amlodipine (or Amlodin), efonidipine, nicardipine etc), diuretics, renin inhibitor (e.g., atiskiren etc.), aldosterone antagonists (e.g., spironolactone, eplerenone etc.), beta-blockers (e.g., metoprolol (or Toporol), atenolol, propranolol, carvedilol, pindolol etc.), vasodilators (e.g., nitrate, soluble guanylate cyclase stimulator or activator, prostacycHne etc.), angiotensin vaccine, cionidine and the like. U.S. Patent Publication No. 201001 13780.

Other therapeutic agents that may be conjugated and/or admixed and/or coformulated with an isomer or analog of monoterpene or sesquiterpene include, but are not limited to, Sertraline (Zoloft), Topiramate (Topamax), Duioxeime(Cyrnbalta), Sumatriptan (Imitrex).

Pregabalin (Lyrica), Lamotrigine (Lamictai), Valaciclovir (Valtrex), Tamsulosin (Fiomax), Zidovudine (Combivir), Larnivudine (Combivir), Efavirenz (Sustiva), Abacavir (Epzicom), Lopinavir ( aletra), Pioglitazone (Actos), Desloratidine (Ciarinex), Cetirizine (Zyrtec),

Pentoprazole (Protouix). lansoprazole (Prevacid), Rebepra o!e (Aeiphex), Moxifloxaciu (Avelox), Meloxicam ( obic), Dorzolamide (Truspot), Diclofenac (Voltaren), Enlapril

(Vasotec), ontelukast (Singulair), Sildenafil (Viagra), Carvedilol (Coreg), Ramipril (Delix).

Table 1 lists pharmaceutical agents that can be conjugated vvitl a isomer or analog monoterpene or sesquiterpene, including the structure of the pharmaceutical agent and the preferred derivative for conjugation.

By way of example, an L-DOPA iso-POH conjugate is shown below;

The purity of an isomer or analog of monoterp ne or sesquiterpene, or its derivatives, may be assayed by gas chromatography (GC) or high pressure liquid chromatography (HPLC). Other techniques for assaying the purity of the compounds of the present invention and for determining the presence of impurities include, but are not limited to, nuclear magnetic resonance (N R) spectroscopy, mass spectrometry (MS), GC-MS, infrared spectroscopy (IR), and thin layer chromatography (TIC), Chirai purity can be assessed by chiral GC or measurement of optical rotation.

The isomer or analog of monoterpene or sesquiterpene (or its deri vatives), may be purified by methods such as crystallization, or by separating the isomer or analo of monoterpene or sesquiterpene (or its derivative), from impurities according to the unique physicochemic-al properties (e.g., solubility or polarity) of the isomer or analog of monoterpene or sesquiterpene (or its derivative). Accordingly, the isomer or analog of monoterpene or sesquiterpene (or its derivative) can be separated by suitable separation techniques known in the art, such as preparative chromatography, (fractional) distillation, or (fractional) crystallization.

The invention also provides for methods of using an isomer or analog of monoterpene or sesquiterpene, as well as using a derivative of an isomer or analog of monoterpene or sesquiterpene, to treat a disease, such as cancer or other nervous system disorders. The compounds of the present invention may be administered alone, or in combination with radiation, surgery or chemotherapeutic agents. An isomer or analog of monoterpene or sesquiterpene, or its derivative, may also be co-administered with antiviral agents, anti- inflammatory agents or antibiotics. The agents may be administered concurrently or sequentially. The compounds of the present invention can be administered before, during or after the administration of the other acti e agent(s).

The compounds and methods of the present invention may used to inhibit the Ras protein. The Ras family is a protein family of smal t GTPases that are involved in cellular signal transduction. Activation of Ras signaling causes cell growth, differentiation and survival.

Mtations in ras genes can permanently activate it and cause inappropriate transmission inside the ceil even in the absence of extracellular signals. Because these signals result in cell growth and division, dysregulated Ras signaling can ultimately lead to oncogenesis and cancer. Activating mutations in Ras are found in 20-25% of all human tumors and up to 90% in specific tumor types. Goodsell DS ( 1999). Downward J., "The molecular perspective: the ras oncogene".

Oncologist 4 (3): 263-4. (Januar 2003). "Targeting RAS signalling pathways in cancer therapy". Nat, Rev, Cancer 3 (1): 1 1-22. Ras family members include, but are not limited to, HRAS; RAS; NRAS; DIRAS1; D1RAS2; DIRAS3; ERAS; GEM; MRAS; NKIRAS1 ;

NKIRAS2; NRAS; RALA; RALB; RAPiA; RAP I B; RAP2A RAP2B; RAP2C; RASD1 ;

RASD2; RASLIOA; RASLIOB; RASL11 A; RASLl IB; RASL12; REM I; R.EM2; RERG;

RERGL; RRAD; RRAS: and RRAS. Wennerberg K, Ross an L. Der CJ (March 2005). "The Ras superfamity at a glance". J. Cell. Sci. 1 18 (Pt 5): 843-6.

The isomer or analog of monoterpene or sesquiterpene, or a derivative of the isomer or analog of monoterpene or sesq iterpene, may be used in combination with radiation therapy. In one embodiment, the present invention provides for a method of treating tumor cells, such as malignant glioma cells, with radiation, where the cells are treated with an effective amount of an isomer or analog of monoterpene or sesquiterpene (or its derivative), such as isoperillyl alcohol, and then exposed to radiation. Treatment by the compounds of the present invention may be before, during and/or after radiation . For example, the compounds of the presen t in vention may be administered continuously beginning one week prior to the initiation of radiotherapy and continued for two weeks after the completion of radiotherapy. U.S. Patent os. 5,587,402 and 5,602,184.

in one embodiment, the present invention provides for a method of treating tumor cells, such as malignant glioma ceils, with chemotherapy, where the cells are treated with an effective amount of an isomer or analog of monoterpene or sesquiterpene (or a derivative of the isomer or analog of monoteipene or sesquiterpene), such as isoperillyl alcohol, and then exposed to chemotherapy. Treatment by the compounds of the present invention may be before, during and/or after chemotherapy. The compounds of the present invention may be used for the treatment of nervous system cancers, such as a malignant glioma {e.g., astrocytoma, anaplastic astrocytoma, glioblastoma multiforme), retinoblastoma, pilocytic astrocytomas (grade I), meningiomas, metastatic brain tumors, neuroblastoma, pituitary adenomas, skull base meningiomas, and skull base cancer. As used herein, the term "nervous system tumors" refers to a condition in which a subject has a malignant proliferation of nervous system cells.

Cancers that can be treated by the present compounds include, but are not limited to, lung cancer, ear, nose and throat cancer, leukemia, colon cancer, melanoma, pancreatic cancer, mammar cancer, prostate cancer, breast cancer, hematopoietic cancer, ovarian cancer, basal cell carcinoma, biliary tract cancer; bladder cancer; bone cancer; breast cancer; cervical cancer; choriocarcinoma; colon and rectum cancer; connective tissue cancer; cancer of the digestive system; endometrial cancer, esophageal cancer; eye cancer; cancer of the head and neck; gastric cancer; intra-epithelial neoplasm; kidney cancer; larynx cancer; leukemia including acute myeloid leukemia, acute lymphoid leukemia, chronic myeloid leukemia, chronic lymphoid leukemia; liver cancer; lymphoma includin Hodgkin's and Non-Hodgkin's lymphoma;

myeloma; fibroma, neuroblastoma; oral cavity cancer (e.g., lip, tongue, mouth, and pharynx); ovarian cancer; pancreatic cancer; prostate cancer; retinoblastoma; rhabdomyosarcoma; rectal cancer; renal cancer; cancer of the respiratory system; sarcoma; skin cancer; stomach cancer; testicular cancer; thyroid cancer; uterine cancer; cancer of the urinary system, as well as other carcinomas and sarcomas. U.S. Patent: No. 7,601 ,355.

The present invention also provides methods of treating CNS disorders, including, without limitation, primary degenerative neurological disorders such as Alzheimer's . ,

Parkinson's, psychological disorders, psychosis and depression. Autism may also be treated by the present compositions and methods. Treatment may consist of the use of a compound of the present invention alone or in combination with current medications used in the treatment of Parkinson's, Alzheimer's, or psychological disorders. The present invention also provides a method of improving immunomodulatory therapy responses comprising the steps of exposing cells to an effective amount of a compound of the present invention, such as isoperiilyl alcohol, before or during immunomodulatory treatment Preferred immunomodulatory agents are cytokines, such interleukins,, lymphokines, monokines, interfereons and chemofcines.

The present composition may be administered by any method known in the art, including, without limitation, intranasal, oral, transdermal, ocular, intraperitoneal, inhalation, intravenous, ICV, intracisternal injection or infusion, subcutaneous, implant, vaginal, sublingual, urethral (e.g., urethral suppository), subcutaneous, intramuscular, intravenous, rectal, sub-tinguat, mucosal, ophthalmic, spinal, intrathecal, intra-articular, intra-arterial, sub-arachinoid, bronchial and lymphatic administration. Topical formulation may be in the form of gel, ointment, cream, aerosol, etc; intranasal formulation can be delivered as a spray or in a drop; transdermal formulation may be administered via a transdermal patch or iontorphoresis; inhalation formulation can be delivered using a nebulizer or similar device. Compositions can also take the form of tablets, pills, capsules, semisolids, powders, sustained release formulations, solutions, suspensions, elixirs, aerosols, or any other appropriate compositions.

To prepare such pharmaceutical compositions, one or more of compound of the present invention may be mixed with a pharmaceutical acceptable carrier, adjuvant and/or excipient, according to conventional pharmaceutical compounding techniques. Pharmaceutically acceptable carriers that can be used in the present compositions encompass any of the standard pharmaceutical carriers, such as a phosphate buffered saline sortition, water, and emulsions, such as an oil water or water/oil emulsion, and various types of wetting agents. The wmpos.itio.us can additionally contain solid pharmaceutical excipients such as starch, cellulose, taic, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, magnesium stearate, sodium stearate, glycerol monostearate, sodium chloride, dried skim milk and die like. Liquid and semisolid excipients may be selected from glycerol, propylene glycol, water, ethanol and various oils, including those of petroleum, animal, vegetable or synthetic origin, e.g., peanut oil, soybean oil, mineral oil, sesame oil, etc. Liquid carriers, particularly for injectable solutions, include water, sal ine, aqueous dextrose, and glycols. For examples of carriers, stabilizers and adjuvants, see Remington's Pharmaceutical Sciences, edited by E. W . Martin (Mack Publishing Company, 18th ed., 1 90), The compositions also can include stabilizers and preservatives.

As used herein, the terra "therapeutically effective amount" is an amount sufficient to treat a specified disorder or disease or alternatively to obtain a pharmacological response treating a disorder or disease . Methods of determining die most effecti ve means and dosage of administration can vary with the composition used for therapy, the purpose of the therapy, the target cell being treated, and the subject being treated. Treatment dosages generally may be titrated to optimize safety and efficacy. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician. Suitable dosage formulations and methods of administering the agents can be readily determined by those of skill in the art. For example, me composition are administered at about 0.01 mg/kg to about 200 mg/kg, about 0.1 rag kg to about 100 mg kg, or about 0.5 mg/kg to about 50 mg kg. When the compounds described herein are co-administered with another agent or therapy, the effective amount may be less than when the agent is used alone,

Transdermal formulations may be prepared by incorporating the active agent in a thixotropic or gelatinous carrier such as a celtulosic medium, e.g.„ methyl cellulose or hydroxyethyl cellulose, with the resulting formulation then being packed in a transdermal device adapted to be secured in dermal contact with the skin of a wearer. If the composition is in the form of a gel, the composition may be rubbed onto a membrane of the patient, for example, the skin, preferabl intact, clean, and dry skin, of the shoulder or upper ami and or die upper torso, and maintained thereon for a period of time sufficient for delivery of the present compound to the blood serum of the patient. The composition of the present invention in gel form may be contained in a tube, a sachet, or a metered pump. Such a tube or sachet may contain one unit dose, or more than one unit dose, of the composition. A metered pump ma b capable of dispe sing one metered dose of the compositi n.

This invention also provides the compositions as described above for intranasal administration. As such, the compositions can further comprise a permeation enhancer. Sout ali e al. Developments in Nasal Dru g Delivery. 2000. The present compound may be administered intranasal^ in a liquid form such as a solution, an emulsion, a suspension, drops, or in a solid form such as a powder, gel, or ointment. Devices to deliver intranasal medications are well know in the art. Nasal drug delivery can be carried out using devices including, but not limited to, intranasal inhalers, intranasal spray devices, atomizers, nasal spray bottles, unit dose containers, pumps, droppers, squeeze bottles, nebulizers, metered dose inhalers (MI ) .), pressurized dose inhalers, insufflators, and bi- directional devices. The nasal delivery device can be metered to administer an accurate effective dosage amount to the nasal cavity, The nasal delivery device can be for single unit delivery or multiple unit delivery, in a specific example, the ViaNase Electronic Atomizer from Karve Technology (Bethell, Washington) can be used in this invention (http://w-ww.kurvetech.com). The compounds of the present invention may also be delivered through a tube, a catheter, a syringe, a packtail, a pledget, a nasal tampon or by submucosal infusion, U.S. Patent Publication Nos. 20090326275, 20090291894, 20090281522 and 20090317377.

The present compound can be formulated as aerosols using standard procedures. The compound may be formulated with or without solvents, and formulated with or without carriers. The formulation may be a solution, or may be an aqueous emulsion with one or more surfactants. For example, an aerosol spray may be generated from pressurized container with a suitable propeilant such as, dichlorodif uoromethane, trichlorofluoromerhane. dichtorotetrafliioroethane, hydrocarbons, compressed air, nitrogen, carbon dioxide, or other suitable gas. The dosage unit can be determined by providing a valve to deliver a metered amount Pump spray dispensers can dispense a metered dose or a dose having a specific particle or droplet size. As used herein, the term "aerosol" refers to a suspension of fine sol id particles or liquid solution droplets in a gas. Specific lly, aerosol includes a gas-borne suspension of droplets of a monoterpene (or sesquiterpene), as may be produced in any suitable device, such as an MD1, a nebulizer, or a mist sprayer. Aerosol a lso includes a dry powder composi tion of the composi tion of the instant invention suspended in air or other carrier gas. Gonda ( 1 90) Critical Reviews in Therapeutic Drug Carrier Systems 6:273-31 . Raeburn et al., (1 92) Pharmacol. Toxicol. Methods 27: 143- 159.

Hie present compound may be delivered to the nasal cavity as a powder in a form such as microspheres delivered by a nasal insufflator. The present compound may be absorbed to a solid surface, for example, a carrier. The powder or microspheres may be administered in a dry, air- dispensable form. The powder or microspheres may be stored in a container of the insufflator. Alternatively the powder or microspheres may be filled into a capsule, such as a gelatin capsule, or other single dose unit adapted for nasal adirtinistraiion.

The pharmaceutical composition can be delivered to the nasal cavity by direct placement of the composition in the nasal ca vity, for example, in the form of a gel, an ointment, a nasal emulsion, a lotion, a cream, a nasal tampon, a dropper, or a bioadfeesive strip, in certain embodiments, it can be desirable to prolong the residence time of the pharmaceutical composition in the nasal cavity, for example, to enhance absorption. Thus, the pharmaceutical composition can optionally be formulated with a bioadhesive polymer, a gum (e.g., xanthan gum), chitosan (e.g., highly purified cationic polysaccharide), pectin (or any carbohydrate that thickens like a gel or emulsifies when applied to nasal mucosa), a microsphere (e.g., starch, albumin, dextran, cyclodextrin), gelatin, a liposome, carbamer, polyvinyl alcohol,, alginate, acacia, chitosans and/or cellulose (e.g., methyl or propyl; hydroxyl or carboxy; carboxymethyt or hydroxy-propyl),

The composition containing the present compound can be administered by oral inhalation into the respiratory tract, i .e., the lungs.

Typical delivery systems for inhalable agents include nebulizer inhalers, dry powder inhalers (DPI), and metered-dose inhalers (MD1).

Nebulizer devices produce a stream of high velocity air that causes a therapeutic agent in the form of liquid to spray as a mist. The therapeutic agent is formulated in a l iquid form such as a solution or a suspension of particles of suitable size, in one embodiment, the particles are micronized. The term "micronized" is defined as having about 90% or more of the particles with a diameter of less than about 10 μτη, Suitable nebulizer devices are provided commercially, for example, by PARI. GmbH (Sternberg, Germany). Other nebulizer devices include Respimat (Boehringer Ingelheim) and those disclosed in, for example, U.S. Patent Nos. 7,568,480 and 6,123,068, and WO 97/12687, The present compound can be formulated for use in a nebulizer device as an aqueous solution or as a liquid suspension.

DPI devices typically administer a therapeutic agent in the form of a free flowing powder that can be dispersed in a patient's air-stream during inspiration. DPI devices which use an external energy source may also be used in die present invention, hi order to achieve a free flowing powder, the present compound can be formulated with a suitable excipient (e.g., lactose), A dry powder formulation can be made, for example, by combining dry lactose having a particle size between about 1 um and 100 μ η α with micronized particles of the present compound and dry blending. Alternatively, the compound can be formulated without excipients. The formulation is loaded into a dry powder dispenser, or into inhalation cartridges or capsules for use with a dry powder delivery device. Examples of DPI devices provided commercially include Di&khaler (GlaxoSmith iine, Research Triangle Park, N.C.) (see, e.g., U.S. Patent No.

5,035,237); Diskus (GlaxoSmithKHne) (see, e.g., U.S. Patent No. 6,378,519; Turbuhaler (AstraZeneca, Wilmington, Del.) (see, e.g., U.S. Patent No. 4,524,769); and Rotahaler

(GlaxoSmithKiine) (see, e.g., U.S. Patent No. 4,353,365). Further examples of suitable DPI devices are described in U.S. Patent Nos. 5, 15,162, 5,239,993, and 5,71 ,810 and references therein.

MDI devices typically discharge a measured amount of the stored composition using compressed propellant gas. Formulations for MDI administration include a solution or suspension of an active ingredient in a liquefied propellant. Examples of propellaxits include hydrofluoroalklanes (UFA), such as l ,U ,2 euafIuoroedmtie (HFA 134a) and 1 ,1, 1 ,2,3,3,3- heptafluoro-n^propane (HFA 227), and chlorofluorocarbons, such as CCI3F. Additional components of HFA formulations for MDI administration include co-solvents, such as ethanol, pentane, water, and surfactants, such as sorbitan trioleate, oleic acid, lecithin, and glycerin. (See, for example, U.S. Patent No. 5,225,183, EP 0717987, and WO 92/22286). The formulation is loaded into an aerosol canister, which forms a portion of an MDI device. Examples of MDI devices developed specifically for use with HFA propellams are provided in U.S. Patent Nos. 6,006,745 and 6,143,227. For examples of processes of preparing suitable formulations and devices suitable for inhalation dosing see U.S. Patent Nos. 6,268,533, 5,983,956, 5,874,063, and 6,221,398, and WO 99/53901 , WO 00/61 108, WO 99/55319 and WO 00/30614.

The present compound may be encapsulated in liposomes or microcapsules for delivery via inhalation, A liposome is a vesicle composed of a lipid bilayer membrane and an aqueous interior. The lipid membrane may be made of phospholipids, examples of which include phosphatidylcholine such as lecithin and lysotecithm; acidic phospholipids such as

phosphatidylserine and phosphatidylglycerol; and sphingophospholipids such as

phosphatidy!ethanolamine and sphingomyelin. Alternatively, cholesterol may be added. A microcapsule is a particle coated with coating material. For example, the coating material may consist of a mixture of a film-forming polymer, a hydrophobic plasticize , a surface activating agent or/and a lubricant nitrogen-containing polymer. U.S. Patent N ' os. 6,313,176 and 7,563,768.

The present compound may also be used alone or in combination with other

chemotherapeutic agents via topical application for the treatment of localized cancers such as breast cancer or melanomas. The present compound may also be used in combination with narcotics or anaigesics for transdermal delivery of pain medication,

'Iriis invention also provides the compositions as described above for ocular

administration. As such, the compositions can further comprise a permeation enhancer, For ocular administration, the compositions described herein can be formulated as a solution, emulsion, suspension, etc. A variety of vehicles suitable for administering compounds to the eye are known in the art. Specific non-limiting examples are described in U.S. Patent Nos.

6,261,547; 6, 97,934; 6,056,950; 5,800,807; 5,776,445; 5,698.219; 5.521 ,222; 5,403,841;

5,077,033; 4,882,150; and 4,738,851.

The present compound can be given alone or in combination with other drugs for the treatmen t of the above diseases for a short or prolonged period of time. The present

compositions can be administered to a mammal, preferably a human. Mammals include, but are not limited to, murines, rats, rabbit, simians, bo vines, ovine, porcine, canines, feline, farm animals, sport animals, pets, equine, and primates. The inventio also provides a method for inhibiting the growth of a cell in vitro, ex vivo or in vivo, where a cell, such as a cancer cell, is contacted with, an effective amount of the present compound as described herein.

Pathological cells or tissue such as hyperproli erati ve cells or tissue may be treated by contacting the cel ts or tissue with an effective amount of a composition of this in vention. The cells, such as cancer cells, can be primary cancer cells or can be cultured cells available from tissue banks such as the American Type Culture Collection (ATCC). The pathological cells can be cells of a systemic cancer, gliomas, meningiomas, pituitary adenomas, or a CNS metastasis from a systemic cancer, lung cancer, prostate cancer, breast cancer, hematopoietic cancer or ovaria cancer. The cells ca be from a vertebrate, preferably a mammal, more preferably a human. U.S. Patent Publication No. 2004/008765 . Balassiano et at. (2002) Intern. J. ol. Med. 10:785-788. Thorne, et al. (2004) Neuroscience 127:481 -496. Fernandes, et al. (2005) Oncology Reports 13:943-947. Da Fotiseca. et al. (2008) Surgical Neurology 70:259267. Da Foi teca, et al. (2008) Arch, Immunol. Thar. EXP, 56:267-276, Hashizume, et al. (2008)

Neuroncoiogy 10: 1 12-120.

In vitro efficacy of the present composition can be determined using methods well known in the art. For example, the cytoxicity of the present compound may be studied by MTT [3-(4 f 5- dimetb.yU ' hi.azoi-2-yl)-2,5-<3iphenyl tetrazolium bromide] cytotoxicity assay. MTT assay is based on the pr inciple of uptake of MTT. a tetrazolium salt, by metabolically acti ve cells where it is metabolized into a blue colored for azon product; which, ca be read spectrometricaUy. J. of Immunological Methods 65: 55 63, 1983. The cytoxicity of the present compound may be studied, by colony formation assay , Functional assays for inhibition of VEGF secretion and l.L-8 secretion may be performed via EL!SA, Cell cycle block by the present compound may be studied by standard propid sm iodide (Pi) staining and flow cytometry. Invasion inhibition may be studied by Boyden chambers. In this assay a layer of reconstituted basement membrane, Matrigel, is coated onto ch.emoiax.is filters and acts as a barrier to the migration of cells in the Boyden chambers. Only cells with invasive capacity can cross the Matrigel barrier, Other assays include, but are not limited to, cell viability assays, apoptosis assays, and morphological assays.

The following are examples of the present invention and are not to be construed as limiting.

Examples

Exam le 1 : Synthesis of Jso-PGH

The reaction scheme is the following:

Preparation of 44wp pyHdet -l, 4^tima-tipim{ 4, SJ eca (2) : isopropyltriphenylphosphonium iodide (83.02 g, 1 2 lnmol) was added to NaH (60%, in mineral oil, 8.38 g, 1 2 mmol) in dry dimethyl sulfoxide (120 mL) at room temperature under a nitrogen atmosphere. The reaction mixture was slowly heated to 50 "C over a period of 15 min and maintained at 50 °C until the reaction mass became a red color (approximately 30 min). A solution of 1 ,4-cyclohexaned.ione monoethytene ketal (1 , 30 g, 192 mmol) in dry dimethyl sulfoxide was added over a period of 45 min while keeping the temperature below 50 °C and the reaction was maintained at 50 "C for 16 h. The reaction mixture was cooled to room temperature, quenched with cold water (1 0 ml), and extracted with ethyl acetate (2x 160 mL). The combined organic layer was washed with water (2*200 mL), followed by brine (10%, 250 mL) and dried over sodium sulfate. The filtered organic layer was concentrated to give a solid which was triturated with hexanes (300 mL) and the precipitated triphenylphosphine oxide was filtered off. The hexane layer was concentrated to give an oil which was purified by column chromatography. [Column dimensions: dia: 6.0 cm, height: 12 cm, silica: 200 mesh, eluted hexanes (1.0 L) followed by hexane: ethyl acetate (97:3, 2.0 L)] The hexane: ethyl acetate fractions were combined and concentrated under vacuum to give an oil. Weight: 23.36 g. Weight yield: 66.7%. 'H-NMR (400 MHz, CDCb): <S 1.61.-1..63 (t, 4H), 1 ,64 (s, 6H), 2.29 (m, 4H), 3,97 (s, 4H), MS (APC1 method): No molecular ion peak was observed.

Preparation of 4- opropyiidme c l hex n e (3):

/j-Toluenesulfonic acid (31.16 g, 1 4 mmol) was added to a solution of ketal (2, 23.0 g,

126 mmol) in acetone (2,3 L) and water (1 S ml,). The reaction mixture was heated to reflux and maintained at reflux for 3.5 h. Tiie mixture was cooled to room temperature, treated with saturated sodium bicarbonate (60 mL) and concentrated under vacuum. Hie resulting oily residue was extracted widi ediyl acetate (2*130 mL), washed wi th water (100 mL), then brine (100 mL), and dried over sodium sulfate. The filtered organic layer was concentrated under vacuum to give an oil. Weight: 16 g. Weight yield: 92%. l H- MR (400 MHz, CDCia): δ 1.69 (s t 6H), 2,35 (t, 4H), 2.50 (t, 4H). MS (APCI method): No molecular ion peak was observed (Note: J H-NMR showed the presence o -2% of ketal 2 but used without purification), ft-eparaiion of4-isopropyU<iene~}~oxa-spiro[2.S}o ianc j (4): Potassium f-butoxide {3.3 g, 29,4 mmol) was added to a . mixture of ketone (3, 2.5 g, 18.1 mmol) and trimethylsulfoxonium iodide (6.45 g, 29.4 mmol) in dry dimetliyl sulfoxide (40 mL) under nitrogen atmosphere at room temperature. The mixture was stirred for 4.0 h at room temperature. The reaction was quenched by the addition of cold water (40 mL) and extracted with ethyl acetate (2x60 mL). The combined organic layer was washed with water (75 mL) followed by brine (75 mL) and dried over sodium sulfate. The filtered organic layer was concentrated under vacuum to give an oil. Weight; 2.13 g. Weight yield; 77%. Ή-NMR (400 MHz, CDCI3): S 1.42-1.50 (m, 2H), 1.55-1.61 (m, 2H), 1.65 (s, 6H), 2,31 (t, 4H), 2.61 (s, 2H). MS (APCi method): No molecular ion peak was observed.

Preparation of S -dimtrobemoic acid 4-isopropyUd e cycioh&rl ' eyiylmethyl ester (6):

Aluminum isopropoxide (5.93 g, 29.0 mmol) was added to a mixture of epoxide (4, .0 g, 26,2 mmol) in toluene (80 mL) and the mixture was heated to reflux for 7,0 h, The mixture was cooled to room temperature and quenched with saturated potassium sodium tartrate solution. The organic layer was separated, washed with water (40 mL), followed by brine (40 mL), and dried over sodium sulfate. The filtered organic layer was concentrated under vacuum to give crude isoperi!jyl alcohol (5) as an oil. Weight; 4.0 g, Weight yield: 100%, Purity: -85-90% (by GC area percent, Actual yield ca; 85%).

Triethylamine (5.1 mL, 36.6 mmol) was added to a solution of crude isoperiilyl alcohol (5, 4.0 g, 26.2 mmoi) in dichtoromethane (50 mL), After stirring for 15 min, 3,5-dinitrobeni50yl chloride (6.3 g, 27.5 mmol) was added over a period of 0.25 h. The reaction mixture was stirred for 3.0 h and quenched with water (30 mL). The organic layer was separated, washed with water (40 mL), and dried over sodium sulfate. The filtered organic layer was concentrated under vacuum to gi ve a pais yellow solid (8.5 g), which was recrystalHsed from acetone to gi ve pure ester 6 as a pale yellow solid. Mp: 138-1 0 °C (acetone). Weight: 5.7 g, Yield: 62% (from epoxide). l H-NMR (400 MHz, CDCI3): 0 ' 1.68 (s, 3H), 1.71 (s, 3*0, 2.18 <t, 2H), 2.40 (t, 2H), 2.87 (br s, 2H), 4.85 (s, 2M); 5.88 (s f 1 H), 9.17 (t, 1H), 9,24 (s, I K), MS (APCI method): m/e: 247.1 (5%), 149.07 (7%), 135,1 (100%), .107.1 (9%). Rep ration of isoperiilyl alcohol (7): Aqueous odium hydroxide (1.43 g, 35,7 mmol, dissol ved in 12.5 mL of water) was added to an ice cold solution of 3, S-dimfirotozok acid 4-isop.fopyl.idene-cyclohex-1-eny netiiy! ester (6, 5.63 g, 1 .2 mmol) in methanol (56 mL) over a period of 0.25 h. The reaction mixture was allowed to warm to room temperature and then stirred for 3.0 h. The methanol was concentrated under vacuum to a minimum stirring volume and the mixture was suspended in water (40 ml). The resulting mixture was extracted with ethyl acetate (2x50 mL), The organic layer was washed with, water (2 50 mL), then brine (50 mL), and dried over sodium sulfate. The filtered organic layer was concentrated under vacuum to give pure isoperiilyi. alcohol as an oil. Weight: 2.35 g f Yield: 95%, Purity: 97% (by GC AUC). l H-NMR (400 MHz, CDC ): S 1.65 (s, 3H), 1.69 (s, 3H), 1.77 (b$, OH), 2,09 (m, 2H), 2.33 (t- 2R), 2,79 (br s, 2H); i ;i C-NMR: 620,38, 20,80, 26.95, 27.60, 29.86, 67,49, 12188, 123.04, 127.92, 138,37. S (APCl method): m e: 152 (M", 3.5%), 135.07 (100 %), 107, 12 (5%). However, the mass spectrum showed four small peaks (-5%) at -k 207.06, 269.1 , 287.09 & 301. which were not characterized.

Example 2; Alternative Synthesis qflso-PQH

The reactio scheme is the following:

Preparation of trifluoromethanesulfonic acid 4~isv mppli nev?efa &K' l-enyt ester (8):

2.5 M solution of n-Butyl lithium in hexaries (5,6 mL, 14.1 mmol) was added t a solution of diisopropyiar ne (1.98 mL, 14, 1. mmol) in dry THF (30 mL) at -78 "C over a period of 0.5 hr. After stirring for 1 ,0 h at -78 C, a sohstion of ketone (3, 1 ,3g„ 9,4 mmol) in dry THF (1.0 mL) was added over a period of .10 min while maintaining the temperature below -78 '3 C, The reaction mixture was stirred for .! .0 h at -78 '3 C, A solution of hetryltriruniidc (3.53g, 9.86 mmol) in THF (1.5 mL) was added slowly while maintaining the temperature below -78 The reaction mixture was slowly warmed to 0 a C„ maintained for 2.0 h at 0 °C and then quenched with satd ammonium chloride solution, The separated orga ic layer was washed with water (15 mL), brine (1 mL) and dried over sodium sulfate. The filtered organic layer was concentrated under vacuum and th l i id ifi d b l h hy. [Column dimensions: dia: 6,0 cm, height: 12 era, silica; 200 mesh, eluted with hexanes (200 mL)] The similar fractions were combined and concentrated under vacuum which gave an oil. Weight: 0.9 g. Weight yield: 38%. *H-N R (400 MHz, CDCな): <S 1.68 (a, 3H), 1.71 (s, 3H), 2.37 (ra, 2H), 2.46 (m, 2H), 2.91 (ra, 2H), 5,73 (m, 1H), MS (APCI method): No molecular ion peak was observed.

Note-1 : Ή-NMR indicated the presence of aromatic peaks ( 5%) between 0 7.42-7.57 which were attributed to the by-product trifluoro- -phenyirr»^anesulfonainide.

Note-2: The compound 8 was also synthesized in iow yield (28%) using triflic anhydride in the presence of 2,6-di-ifc j /'/-butyl-4-methylpyridine as a base.

Preparation of 4~i$opropyUdene cyclohex-l-enecarboxyUc acid methyl est&r (9);

To a solution of compound 8 (0.2g, 0.74 mmoi) in N'N-dimethylformamide (1.5 raL) was added methanol ( 1 ,0 mL), triethylamine (0, 17 mL, 1 ,2 mmoi), 1 ,3- bis(diphenylphosphino)propane (0,03 g, 0.07 mmoi) and palladium acetate (0.04g, 0.07 mmoi). The reaction mixture was degassed and then stirred at room temperature under carbon monoxide (balloon pressure) for 5 h. The reaction mixture was diluted with ethyl acetate (15 mL) and washed with 0.5 N HG (15 mL), brine (1 mL) and dried over sodium sulfate. The filtered organic layer was concentrated under vacuum and the resulting residue was purified by column chromatography. [Column dimensions: dia: 6.0 cm, height: 12 cm, silica: 200 mesh, eluted with hexanes (.100 mL) followed by ethyl acetate: hexanes (2%, 150 mL)] The similar fractions were combined and concentrated under vacuum which gave an oil. While TLC analysis showed only a single spot, l H.-NMR and GC analysis indicated that the isolated material, was a mixture of two primary components that co-eluted by TLC. Weight: 0.1 1 g. Weight yield: 82%. Ή-N R (400 MHz, CDC ) indicated the presence of peaks corresponding to the methyl ester (9) along with an unknown impurity. GC analysis confirmed that it is mainly a mixture of two compounds with a ratio of 3: 1. MS (APCI method): nVe: 180 (M\ 5%), 180.9 QA* ' ( 100 %). The other peaks (< 5%) at M- : 197.8, 247,0 & 274.0 were not characterized. The crude mixture was taken forward without purification.

Reparation ofisoperi yl akokol (7): Methyl ester (9, .1 lg, 0.6 mmoi) in dry THF (10 mL) was added to a cold solution of LAil (0.03g, 0,78 mmoi) in dry THF (10 mL) over a period of 2 min. The reaction mixture was slowly heated to reflu and maintained for 3,0 h. The mixture was cooled to 5 °C and quenched with satd sodium sulfate {1.5 raL). The precipitated lithium salts were fi ltered off and washed with hot ethyl acetate (20 niL). The filtrate was dried over sodium sulfate, The filtered organic layer was concentrated under vacuum which gave an oil. Weight: 74 rag. Weight yield: 79%. While TLC analysis showed only a single spot, 'H-NMR and GC analysis indicated that the isolated material was a mixture of two primary components that co-eluted by TLC, Ή-NMR (400 MHz, CDCl,) indicated the presence of peaks

corresponding to the isoperillyl alcohol (7) along with an unknown impurity. MS (APCI method): m/e: 153 ( ' \ 40%), 152 (M ' \ 13 %), 135.09 (M-OH). The other peaks at M+: 169.03 (10%), 255,20, (1 %), 285,25 (15%), 287.19 (70%), 290 (68%), & 397,24 (15%) were not characterized. GC analysis confirmed the presence of isoperillyl alcohol (20,5%, (AUC)), compared with the iso-POH obtained from the epoxide route along with the unknown impurity (67.5%, (AUC)).

Example 3: In vitro Cytotoxicity Studies of iso-POH ((4-Isopropylidene cyciohex-I-enyl)- methanol)

The MTT cytotoxicit assays were carried out after cells were treated with iso-POH (e.g., synthesized by the method in Example 1) or other types of POH with different purity. Figure 1 shows the results of the MTT cytotoxicity assays demonstrating the efficacy of different types of POH and iso-POH in killing LN229 human glioma ceils. Sigma POH is the POH purchased from Sigma Chemicals having a purity of about 96%. SGP-527-155 was prepared from the WAKO POH by rwo-fbid crystallization from di-isopropyl ether solvent, and has a GC relative area purity of about 98.7% (area under the curve). WH0744 is the crude POH purchased from Wako having a purity of about 89.5%. SGP-527-130 was prepared from the WAKO POH by single crystallization from di-isopropyl ether solvent, and has a GC relative area purity of about 97.1% (area under the curve). Figure 2 shows the results of the MTT cytotoxicit assays demonstrating the efficacy of different types of VOE and iso-POH in killing U251 human glioma cells. Figure 3 shows the results of the MTT cytotoxicity assays demonstrating the efficacy of different types of POH and iso-POH in killing A 172 human glioma cells. The results suggest thai iso-POH exhibited much better cytotoxicity than POH with different purity. In vitro cytotoxicity of iso-POH in teraozolomide-sensitive or te ozolomide-resistant ceils were also studied Glioma cells were treated with Sigma POH, POH syndiesized to GLP quality (SGP-527-155) having a purity of about 98,7%, and iso-POH (SGP-561-79P, SGP-561 - 65P) for 48 hours and MTT assay performed Figure 4 demonstrates that A 172 ceils had the greatest cytotoxic response to iso-POH compared to GLP POH and Sigma POH (Fig 4A). This response pattern was also seen in Al 72 temozoiomide resistant ceils (Figure 4B). Similarly, Figure 5 demonstrates that U251 cells had the greatest response to iso-POH (Figure 5 A), and that this response was also seen in U251 temozoiomide resistant cells (Figure 5B). The same response to iso-POH was seen in LN229 temozoiomide sensitive (Figure 6A) and temozoiomide resistant cells (Figure 6B). U87 ceils, both temozoiomide sensitive (Figure 7A) and resistant (Figure 7B), had the greatest response to iso-POH, albeit less tha LN229 and U251 cells.

Figure 8 shows the results of the MTT assays performed using ΎΜΖ sensiti ve U251 glioma cells over 24 hours using Sigma POH, GLP POH having a purity of about 98.7%, iso- POH (ISO-POH65, IS -POH79) all at 0 ra - 3 mM concentration. Iso-POH had greater cytotoxicity compared to Sigma POH and GLP POH (Figure 8 A). U2 1 temozoiomide resistant ceil line (U251 -TR2) treated under same conditions demonstrated greater cytotoxicity with iso- POH compared to Sigma and GLP POH (Figure 88). Another U251 temozoiomide resistant cell line (U251 -TH1) treated under same conditions also demonstrated greater cytotoxicity with iso- POH (iso-POH65, iso-POH79) compared to GLP POH or Sigma POH (Figure 8C).

Figure 9 shows die resuits of die MTT assay performed using glioblastoma cancer stem cell line USC04 treated with both Gi -POH and iso-POH (3so-POH65) over 24 hrs. GLP POH and iso-POH demonstrated similar cytoxicity on the ceils.

U25.1 T Z-sensirive and T Z-resistant (U251-TR 1, U25.1-TR2) cells were treated with Sigma POH ( 1.5mM) or GLP POH (1.5 mM) for 18 hours, dien Western blot was performed. The results show that Sigma POH and GLP POH increased expression of glucose-regulatory protein 78 (GRP-78) and the apoptosis marker CHOP, suggesting increased endoplasmic reticulum (ER) stress after treatment (Figure 10a), Under the same conditions, iso-ϊΌΗ (iso- POH^, iso~POH79) also increased ER stress (Figure 10b).

U251 glioma celts were treated with 500 μ Sigma POH, GLP POH or iso-POH

(isoPOH65, isoPOH79) for 24 hours, then Western blot was performed. The results (Figure i i) demonstrate tliat all treatments decreased Kras expression. Example 4; Synthesis of iso-POH Conjugated with Teroosolamide (TMZ)

The reaction scheme is die following:

Prejpar&fi<m *,| Y#~ &/& ^

mrbmnic id -4-i pn>py me &yGfahex*l « e yim& ' thyi ite :

Oxalyl chloride (0.26 g, 2,0 mraol) will be added slowly to a mixture of Temo- lamide (Source: OChem Incorporation, Lot # 07.1 .1 185A; 0.2 g, .1 ,0 mmoi) in 1 ,,2-dichloroethane (13 mL) over a period of 5 mitt while maintaining the temperature at 10 °C under Nj. The reaction mixture will be allowed to warm to room temperature and then heated to reflux for 2.5 h. The excess of oxalyl chloride and 1 ,2-dichloroethane will be removed by concentration under vacuum. The resulting residue will be redissoKed in 1 ,2-dichloroethane (20 mL) and the reaction mixture cooled to 5°C under N^, A solution of isoperillyl alcohol (0.1.7 g„ 1 , 12 mmoi) in .1 ,2-dichloroethane (5 ml.) will be added over a period of 10 min. The reaction mixture will be allowed to warm to room temperature and stirred for 12 h. 1 ,2-Dichloroetbane will he concentrated under vacuum to gi ve a residue which will be triturated with hexanes, The resulting pale yellow solid will be filtered and washed with hexanes.

Exam le 5: Synthesis of Jso-FQH Conjugated with Rolipram

The reaction scheme is as follows.

mpmpy^de yclo ex^l^nyfm^yi ester;

Phosgene (20% in. toluene, 19.5 ml, 39.4 mmol) will be added to a mixture of isoperiUyl alcohol (3.0 g, 19.7 mmol) and potassium carbonate (8.1 g 5 58.6 mmol) in dry toluene (45 ml,) over a period of 45 mm while maintaining the temperature between 10-12 "C. The reaction mixture will, be allowed to warm to room temperature ami stirred for .10 h under Nj. The reaction mixture will be quenched wit water (40 m.L) and the organic layer separated. The aqueous layer will be extracted with, toluene (30 mL) and the combined organic layer washed with water (40 mL x 2), brine (.10%, 40 mL), and dried over sodium sulfate (25 g). The filtered organic layer will be concentrated under vacuum to give isoperiUyl chloor formate as an oil.

Butyl lithium (2,5 M s 0,36 mL, 0,90 mmol) will be added to a solution of rolipram (Source: GL synthesis, inc. Lot # GLS-SH-l 10809; 0.2g, 0.72 mmol) in dry THF (8 ml,) at -72 ' , over a period of 10 m under j. After the reaction mixture being stirred for 1.0 h. at. -72 "C, isoperillyi chlorotbrmate (0.16 g, 0,76 mmol, dissolved in 4 mL THF) will be added o ver a period of 10 nun whi le maintaining the temperature at -72 "C. The reaction mixture will be stirred fo 3 h and quenched with saturated ammonium chloride (10 mL), The reaction mixture will be allowed to warm to room temperature and extracted with ethyl acetate (2x20 mL). The combined organic layer will be washed with water (20 mL), brine ( 10%, 25 mL), and dried over sodium sulfate. The filtered organic layer will be concentrated to give an oil. which will be purified by column chromatography [Column dimensions; dia: 1 ,5 cm, height: 1 cm, silica: 230-400 mesh] and elated wi th a mixture of 5% ethyl, acetate hexanes (120 mL) followed by 10% ethyl aceiate/hexaaes ( 150 mL). The 10% ethyl acetate /bexanes fractions will, be combined and concentrated under vacuum to give a gummy solid.

Example 6: Synthesis of Dimethyl Celecoxib bis iso-POH carbamate conjugate

The reaction scheme is as follows.

PMparatlm of 4-0is~N, <V '^~L<iopmjtyliii&te cyvfo &x*^eftylmethyi xy e rhonyl ~(Χ 5-

Phosgene (20% in. toluene, 10.5 ml, 39.4 mraol) will be added to a mixture of isoperil!yi alcohol (3.0 g, 19.7 mmol) and potassium carbonate (8.1 g } 58.6 mmol.) in dry toluene (45 ml) over a period of 45 min while ma ntaining the temperature between .10-12 tl C. The reaction mixture will, be allowed to warm to room temperature and stirred for .10 h under Nj. The reaction mixture will be quenched with water (40 mL) and the organic layer separated. The aqueou layer will be extracted with toluene (30 mL) and the combined organic layer washed with water (40 mL x 2), brine (10%, 40 mL), and dried over sodium sulfate (25 g). The filtered organic layer will be concentrated under vacuum to gi e isoperiliyl chloroformate as an oil.

Isoperil!yi chloroformate (0.22 g, 1.0 mmol) will be added slowly to a mixture of dimethyl celecoxib (0.2 g, 0.50 mmol) and potassium carbonate (0.14 g, 1.0 mraol) in dry acetone (25 mL) over a period of 5 min under j. The reaction mixture will be heated to reflux and maintained t r 4 h. The reaction mixture will be cooled and the acetone concentrated under vacuum. The resulting residue will be suspended in water (25 ml) and extracted with ethyl acetate {3x20 mL), The combined organic layer will be washed with water (40 mL), followed by brine (10%, 30 mL), and dried over sodium sulfate. The filtered organic layer will be concentrated under vacuum to give a residue which will be purified by column chromatography [Column dimensions; dia: 1.5 cm, height: 15 cm, silica: 230-400 mesh] and eiuted with hexanes (100 mL) followed by a mixture of hexanes ethyl acetate (95:5, 100 mL). The hexane/ethyl acetate fractions will be combined and concentrated under vacuum to give a gummy mass.

The scope of the present invention is not limited by what has bee specifically shown and described hereinabove. Those skilled in the art will recognize that there are suitable alternatives to the depicted examples of materials, configurations, constructions and dimensions. Numerous references, including patents and various publications, are cited and discussed in the description of this invention, The citation and discussion of such references is provided merely to clarify the description of the present invention and is not an admission that any reference is prior art to the invention described herein. AH references cited and discussed in this specification are incorporated herein by reference in their entirety. Variations, modifications and other implementations of what is descri bed herein will occur to those of ordinary skill in the art without departing from the spirit and scope of the invention. While certain embodiments of the present invention have been shown and described, it will be obvious to those skilled in the art thai changes and modifications may be made without departing from the spi rit and scope of die invention. The matter set forth in the foregoing description and accompanying drawings is offered by way of illustration only and not as a limitati on.