Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ANTIOXIDANT CAMPTOTHECIN DERIVATIVES AND ANTIOXIDANT ANTINEOPLASTIC NANOSPHERES THEREOF
Document Type and Number:
WIPO Patent Application WO/2010/060098
Kind Code:
A1
Abstract:
The present invention is directed to antioxidant derivatives of camptothecin and antioxidant derivatives of camptothecin analogs and the preparation of nanometer-sized camptothecin prodrugs. Methods of synthesizing the antioxidant derivatives of camptothecin and antioxidant derivatives of camptothecin analogs, spontaneous emulsification or nanoprecipitation thereof to produce antioxidant camptothecin nanosphere prodrugs and their use in treating cancerous diseases are also provided. A further aspect of this invention is the use of these antioxidant camptothecin nanosphere prodrugs for the preparation of delivery devices of other pharmaceuticals and/or drugs.

Inventors:
YU JOHN S (US)
LEE BONG SEOP (US)
Application Number:
PCT/US2009/065776
Publication Date:
May 27, 2010
Filing Date:
November 24, 2009
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CEDARS SINAI MEDICAL CENTER (US)
YU JOHN S (US)
LEE BONG SEOP (US)
International Classes:
C07D471/00
Domestic Patent References:
WO2008012365A22008-01-31
WO2009086547A12009-07-09
Foreign References:
US5122526A1992-06-16
US6387945B22002-05-14
US6664287B22003-12-16
US6629995B12003-10-07
US20060013882A12006-01-19
US6090842A2000-07-18
US6013663A2000-01-11
US6117899A2000-09-12
US6127394A2000-10-03
US6150358A2000-11-21
US6204288B12001-03-20
US6235772B12001-05-22
US6288106B12001-09-11
US6353011B12002-03-05
US6369098B12002-04-09
US6387945B22002-05-14
US6605637B12003-08-12
US6887891B22005-05-03
US6900338B12005-05-31
US6936715B22005-08-30
US6387945B22002-05-14
US6664287B22003-12-16
US6605637B12003-08-12
US6887891B22005-05-03
US6936715B22005-08-30
US5693664A1997-12-02
US5948810A1999-09-07
US6884420B22005-04-26
US5925668A1999-07-20
US6251935B12001-06-26
US6472432B12002-10-29
US6586472B22003-07-01
USPP1874908P2008-01-03
US20090039956W2009-04-08
Other References:
THOMAS ET AL.: "Camptothecin: Current perspectives", B . M . C ., vol. 12, 2004, pages 1585 - 1604
PIZZOLATO ET AL.: "The Camptothecin", THE LANCET, vol. 361, 2003, pages 2235 - 2242
PACKER ET AL., FREE RADICAL BIOLOGY & MEDICINE, vol. 19, no. 2, 1995, pages 227 - 250
DUNCAN; HEALES: "Nitric Oxide and Neurological Disorders", MOLECULAR ASPECTS OF MEDICINE, vol. 26, 2005, pages 67 - 96
KERWIN ET AL.: "Nitric Oxide: A New Paradigm For Second Messengers", J. M . C ., vol. 38, 1995, pages 4343 - 4362
PACKER ET AL., FREE RADICAL BIOLOGY & MEDICINE, vol. 19, 1995, pages 227 - 250
PACKER ET AL., FREE RADICAL BIOLOGY & MEDICINE, vol. 22, 1997, pages 359 - 378
SINGLETON ET AL.: "Dictionary of Microbiology and Molecular Biology", 2001, J. WILEY & SONS
MARCH: "Advanced Organic Chemistry Reactions, Mechanisms and Structure", 2001, J. WILEY & SONS
REMINGTON: "The Science and Practice of Pharmacy", 2000, WILLIAMS & WILKINS
See also references of EP 2370435A4
Attorney, Agent or Firm:
TRUONG, Linda, B. et al. (865 South Figueroa StreetSuite 240, Los Angeles California, US)
Download PDF:
Claims:
CLAIMS

WHAT IS CLAIMED IS:

1. A compound comprising:

Formula Il wherein A and B are independently selected from the group consisting of — OC(O)-, -OC(O)O-, and -OC(O)N(R)-, wherein R is a hydrogen atom, or a substituted, unsubstituted. branched or unbranched chain of carbon atoms;

X and Y are linkers, each independently comprising a substituted, unsubstituted, branched or unbranched chain of carbon atoms and may optionally contain a heteroatom; and

R1, R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, alkyl. aryl, cycloaliphatic, and aralkyl and may each optionally contain a hetero atom. 2. The compound of claim 1 , having Formula IV:

wherein Li is a moiety formed by esterificatϊon of two free esterifiable hydroxyl groups on a dioi; and

R1, R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, alkyl, aryl, cycioaliphatic, and aralkyl group, and may optionally contain a hetero atom. 3. The compound claim 2, wherein the diol is selected from the group consisting of whereιn w is a hydrocarbon group, wherein n is an integer between 1 and 100,

HO-J-CHΓ^OH wherein n is an integer between 2 and 12,

4. The compound of claim 2, selected from the group consisting of Formula V

Formula Vl

Formula VII

Formula ViIi

Formula IX

Formula X

Formula XLVI

5. A compound, comprising:

Formula IH wherein A is selected from the group consisting of — OC(O) — , -OC(O)O-, and -OC(O)N(R)-, wherein R is a hydrogen atom, or a substituted, unsubstituted, branched or unbranched chain of carbon atoms:

P is selected from the group consisting of — OC(O) — , and -N(R)C(O)-, wherein R is a hydrogen atom, or a substituted, unsubstituted, branched or unbranched chain of carbon atoms;

X is a linker comprising a substituted, unsubstituted, branched or unbranched chain of carbon atoms and may optionally contain a heteroatom, and

R1, R2. R3, R4, and R5 are each independently selected from the group consisting of hydrogen, alkyl. aryl, cycloaliphatic. and aralkyl and may each optionally contain a hetero atom.

6. The compound of claim 5, having Formula Xl

R2 ' Tr < 0(COlN(H) L. N(HlClO)

R3-V - RS. V R4 wherein L2 is a moiety formed by using a diamine as the linker in the process of producing the compound; and

R1, R2, R3, 43, and R5 are each independently selected from the group consisting of hydrogen, aikyl, aryl, cycloaiiphatic, and aralkyl group, and may optionally contain a hetero atom. 7. The compound of claim 6, wherein the diamine is selected from the group consisting of: wherein X is a hydrocarbon group, , wherein n is an integer between 1 and 100, and wherein n is an integer between 2 and 12. 8. The compound of claim 6, selected from the group consisting of: Formula XII

Formula XIIl

Formuta XIV

Formula XV

Formula XVI

Formula XVIt

and

Formula XLVII

9. The compound of claim 5, having Formula XVIII

wherein L3 is a moiety formed by using an aminoalcohol as the linker in the process of producing the compound and R1 , R2, R3, R4 and R5 are each independently selected from the group consisting of hydrogen, alky!, aryl, cycloaliphatic, and aralkyl and may each contain a hetero atom The compound of claim 9, wherein the aminoalcohol is selected from the group consisting of wherefn the γ |S a hydrocarbon group, and may optionally contain a hetero atom wherein n ts an integer between 1 and 100 and wherein n is an integer between 2 and 12 The compound of claim 9 selected from the group consisting of Formula XIX

Formula XX

Formula XXI

Formula XXIl

Formula XXIiI

Formula XXiV

and

Formula XLVlII

A compound produced by conjugation of an α-lipoic acid and camptothecm or a camptothecin analog modified by reacting with succinic anhydride or glutaric anhydride, wherein the camptothecin analog is represented by Formula I

wherein R1, R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, aikyl, aryl, cycloaliphatic, and aralkyl, and may optionally contain a hetero atom The compound of claim 12, selected from the group consisting of

Formula XXV

Formula XXVI

Formula XXVl

Formula XXVIII

Formula XXIX

Formula XXX

Formula XXXI Formula XXXiI

Formula XXXIII

Formula XXXIV

Formula XXXV

Formula XXVI

Formula XXXVII Formula XXXVIII

Formula XXXlX

Formuia XL

Formula XLI

Formula XLl

Formula XLIiI Formula XLIV

, and

Formula XLV

wherein R1, R2, R3, R4 and R5 are each independently selected from the group consisting of hydrogen alkyl, aryl, cycioaiiphatic, and aralkyl, and may optionally contain a hetero atom A compound selected from the group consisting of

Compound 23

Compound 1

Compound 2

Compound 10

Compound 3

Compound 4

Compound 5

Compound 11

Compound 6

Compound 7

Compound 8 Compound 12

Compound 9

Compound 13

Compound 14

Compound 15

Compound 16

Compound 17 Compound 18

Compound 19

Compound 20

Compound 21

, and

Compound 22

A nanosphere, comprising the compound of claim 1 The nanosphere of claim 15, further comprising a compound selected from the group consisting of a multiple α-lιpoιc acid-containing hydrophobic compound, α-tocopherol a nonsteroidal antiinflammatory drug (NSAID) derivative and combinations thereof A nanosphere comprising the compound of claim 5 The nanosphere of claim 17, further comprising a compound selected from the group consisting of a multiple α-lipoic acid-containing hydrophobic compound, α-tocopherol, a nonsteroidal antiinflammatory drug (NSAID) derivative, and combinations thereof A nanosphere, comprising the compound of claim 12 The nanosphere of claim 19, further comprising a compound selected from the group consisting of a multiple α-lιpoιc acid-containing hydrophobic compound, α-tocopherol a nonsteroidal antiinflammatory drug (NSAID) derivative, and combinations thereof A method of treating cancer in a subject in need thereof comprising providing a composition comprising the compound of claim 1 , and administering a therapeutically effective amount of the composition to the subject to treat the cancer A method of treating cancer in a subject in need thereof, comprising providing a composition comprising the compound of claim 5, and administering a therapeutically effective amount of the composition to the subject to treat the cancer A method of treating cancer in a subject in need thereof, comprising providing a composition comprising the compound of claim 12, and administering a therapeutically effective amount of the composition to the subject to treat the cancer A method of treating cancer in a subject in need thereof, comprising providing the nanosphere of claim 15, and administering a therapeutically effective amount of the nanosphere to the subject to treat the cancer A method of treating cancer in a subject in need thereof, comprising providing the nanosphere of claim 17, and administering a therapeutically effective amount of the nanosphere to the subject to treat the cancer A method of treating cancer in a subject in need thereof comprising providing the nanosphere of claim 19 and administering a therapeutically effective amount of the naπosphere to the subject to treat the cancer. A pharmaceutical composition, comprising: a pharmaceutically acceptable carrier or excipient; and a compound selected from the group consisting of

Formula II wherein A and B are independently selected from the group consisting of — OC(O)-, -OC(O)O-, and -OC(O)N(R)-, wherein

R is a hydrogen atom, or a substituted, unsubstituted, branched or unbraπched chain of carbon atoms,

X and Y are linkers, each independently comprising a substituted, unsubstituted, branched or unbranched chain of carbon atoms and may optionally contain a heteroatom, and

R1, R2, R3, R4. and R5 are each independently selected from the group consisting of hydrogen, alkyl, aryl, cycloaliphatic. and aralkyl and may each optionally contain a hetero atom;

Formula IV wherein L1 is a moiety formed by esteriflcation of two free esterifiable hydroxy! groups on a dioj, and

R1, R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, alkyl, aryl, cycloaliphatic, and aralkyl group, and may optionally contain a hetero atom;

Formula III wherein A ts selected from the group consisting of — OC(O) — , -OC(O)O-, and -OC(O)N(R)-, wherein R is a hydrogen atom, or a substituted, unsubstituted, branched or unbranched chain of carbon atoms,

P is selected from the group consisting of -OC(O) — , and -N(R)C(O)-, wherein R is a hydrogen atom, or a substituted, unsubstituted, branched or unbranched chain of carbon atoms,

X is a linker comprising a substituted, unsubstituted, branched or unbranched chain of carbon atoms and may optionally contain a heteroatom, and

R1, R2, R3, R4. and Rs are each independently selected from the group consisting of hydrogen, alkyl, aryl, cycloaliphatic, and aralkyl, and may each optionally contain a hetero atom;

Formula Xl, wherein L2 is a moiety formed by using a diamine as the linker in the process of producing the compound, and

R1 , R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, alkyl, aryl, cycloaliphatic, and aralkyl group, and may optionally contain a hetero atom,

Formula XVIII wherein L3 is a moiety formed by using an aminoalcohol as the linker in the process of producing the compound, and

R1, R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, alkyl, aryl, cycloaliphatic, and aralkyl, and may each contain a hetero atom, and a compound produced by conjugation of an α-lipotc acid and camptothecin or a camptothecin analog modified by reacting with succinic anhydride or giutaric anhydride, wherein the camptothecin analog is represented by Formula I

wherein R1, R2, R3, R4, and R5 are each independently selected from the group consisting of hydrogen, alky!, aryi, cycloaliphatic, and araikyi, and may optionally contain a hetero atom.

Description:
ANTIOXIDANT CAMPTOTHECIN DERIVATIVES AND ANTIOXIDANT ANTINEOPLASTIC NANOSPHERES THEREOF

FIELD OF INVENTION This invention relates to antioxidant camptothecin derivatives and naπospheres thereof.

BACKGROUND

AIi publications herein are incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference. The following description includes information that may be useful in understanding the present invention. It is not an admission that any of the information provided herein is prior art or relevant to the presently claimed invention, or that any publication specifically or implicitly referenced is prior art.

Antineoplastic effect of camptothecin

Camptothecin is a plant alkaloid first isolated from the wood and barks of Camptotheca acuminate (Nyssaceae), and exhibits its antineoplastic effect by the inhibition of DNA relaxation by DNA topoisomerase I. However, camptothecin is essentially insoluble in water, and therefore, numerous derivatives have been developed to increase the water solubility (Thomas et al., Camptothecin: Current perspectives. BIOORG. MED. CHEM.. 12, 2004, 1585-1604: PizzoJato et a!.. The Camptothecin THE LANCET, 361. 2003, 2235-2242). Camptothecin consists of a pentacyclic structure having a lactone in the E- ring. which is essential for antitumor effects of the molecule. It has been demonstrated that the main transformation and elimination pathways of the drug comprise lactone hydrolysis and urinary excretion. In fact, the lactone form is 50% hydrolyzed to an open ring 30 minutes after administration The sodium salt showed a lower activity than camptothecin, because at pH 7.4 the inactive form (open ring) predominates on the lactone active form {closed ring) Antioxidant effect of α-lipoic acid

Molecules containing a dithiolane moiety are widely investigated due to their antioxidant properties α-Lipoic acid {thioctic acid, 1 ,2-dιthιolane-3-pentanoιc acid), which has dithiolane ring in its molecule is a widely distributed natural substance which was originally discovered as a growth factor Physiologically it acts as a coenzyme of the oxidative decarboxylation of o>keto carboxylic acid (e g , pyruvates) and as an antioxidant, and it is able to regenerate vitamin C, vitamin E, glutathione and coenzyme Q10 In pathological conditions, lipoic acid is applied in the treatment of diabetic polyneuropathy, liver cirrhosis and metal intoxications

α-Lipoic acid Dihydrolipoic acid

Lipoic acid and dihydrolipoic acid are capable of trapping a number of radicals both in a lipid and in an aqueous environment Lipoic acid and dihydrolipoic acid act as antioxidants not only by direct radical trapping and/or metal chelation but also by recycling other antioxidants (e g , vitamin C vitamin E) and by reducing glutathione, which in turn recycles vitamin E The two thiol groups present in [1 ,2]-dιthιolane ring system confer it a unique antioxidant potential The disulfides with a cyclic five- member ring such as lipoic acid have been found to be more effective in reductive and/or nucleophilic attack than open-chain derivatives such as cystine or glutathione The antioxidant potential of a compound may be evaluated based on the properties such as (1 ) specificity of free radical scavenging, (2) interaction with other antioxidants, (3) metal-chelating activity, (4) effects on gene expression (5) absorption and bioavailability, (6) location (in aqueous or membrane domains, or both), and (7) ability to repair oxidative damage (Packer et al , FREE RADICAL BIOLOGY & MEDICINE 19(2) 227-250, 1995) According to the above criteria, the [1 ,2]- dithiolane containing lipoic acid/dihydrolipoic acid redox system has been regarded as a universal antioxidant

There have been many attempts to develop lipoic acid derivatives or complexes having antioxidant activity U S Patent Nos 6 090,842 6 013 663, 6 117,899 6, 127,394 6, 150,358 6,204 288 6,235,772 6,288,106, 6 353,011 , 6,369,098, 6,387,945, 6,605,637, 6,887,891 6,900,338, and 6,936,715 are some examples

In many other U S patents the natural and synthetic lipoic acid derivatives and their metabolites are disclosed for use in preventing skin aging and in the treatment of free radical mediated diseases, including inflammatory, proliferative neurodegenerative metabolic and infectious diseases

Inhibitory activity on NO-synthase and trapping the reactive oxygen species (ROS) Various conditions or disease conditions have demonstrated a potential role of nitric oxide (NO) and the ROS's and the metabolism of glutathione in their physiopathology Conditions or disease conditions where nitrogen monoxide and the metabolism of glutathione as well as the redox status of thiol groups are involved include but are not limited to cardiovascular and cerebrovascular disorders (e g , atherosclerosis, migraine, arterial hypertension, septic shock, ischemic or hemorrhagic cardiac or cerebral infarctions, ischemias and thromboses), disorders of the central or peripheral nervous system (e g , neurodegenerative nervous system), neurodegenerative diseases including cerebral infarctions, sub-arachnoid hemorrhaging, ageing, senile dementias (e g , Alzheimer's disease), Huntmgton's chorea, Parkinson's disease, prion disease (e g , Creutzfeld Jacob disease), amyotrophic lateral sclerosis, pain, cerebral and spinal cord traumas, proliferative and inflammatory diseases (e g , atherosclerosis), amyloidoses, and inflammations of the gastro-intestinai system, organ transplantation, diabetes and its complications (e g , retinopathies, nephropathies and polyneuropathies, multiple sclerosis, myopathies); cancer, autosomal genetic diseases (e g , Unvemcht-Lundborg disease), neurological diseases associated with intoxications (e g , cadmium poisoning inhalation of n-hexane, pesticides, herbicides), associated with treatments (e g radiotherapy) or disorders of genetic origin (e g , Wilson's disease), and impotence linked to diabetes

These conditions and disease conditions are characterized by an excessive production or a dysfunction of nitrogen monoxide and/or the metabolism of glutathione and of the redox status of the thiol groups (Duncan and Heales, Nitric Oxide and Neurological Disorders, MOLECULAR ASPECTS OF MEDICINE 26 67-96 2005; Kerwin et ai, Nitric Oxide: A New Paradigm For Second Messengers, J. MED. CHEM. 38:4343-4362, 1995; Packer et a/., FREE RADICAL BIOLOGY & MEDICINE 19:227-250, 1995). U.S. Pat. Nos. 6,605,637, 6,887,891 , and 6.936,715 disclose that lipoic acid derivatives inhibit the activity of NOsynthase enzymes producing nitrogen monoxide NO and regenerate endogenous antioxidants which trap the ROS and which intervene in a more general fashion in the redox status of thiol groups. U.S. Pat. Nos. 5,693,664, 5,948,810, and 6,884,420 disclose the use of racemic α-iipoϊc acid or their metabolites, salts, amides or esters for the synthesis of drugs for the treatment of diabetes meilϊtus of types i and II. U.S. Pat. No. 5,925,668 discloses a method of treating free radical mediated diseases, and/or reducing the symptoms associated with such diseases whereby the compounds with antioxidant activity contain 1 ,2-dithiolane, reduced or oxidized forms. U.S. Pat. No. 6,251 ,935 discloses methods for the prevention or treatment of migraine comprising the administration of an active ingredient selected from the group consisting of racemic alpha-lipoic acid, enantiomers and pharmaceutically acceptable salts, amides, esters or thioesters thereof. U S. Pat. Nos. 6,472,432 and 6,586,472 disclose the treatment of a chronic inflammatory disorder rosacea by application of a composition containing lipoic acid and/or lipoic acid derivatives. There is also strong evidence that the neuroprotective effects of lipoic acid and dihydrolipoic acid are mediated by antioxidant and free radical scavenging mechanisms (Packer et a/., FREE RADICAL BIOLOGY & MEDICINE. 22:359-378, 1997}

SUMMARY OF THE INVENTION

The following embodiments and aspects thereof are described and illustrated in conjunction with compositions and methods which are meant to be exemplary and illustrative, not limiting in scope.

One embodiment of present invention provides for a compound comprising:

Formula II. wherein A and B may be independently selected from the group consisting of — OC(O)--, --OC(O)O-. and -OC(O)N(R)-, wherein R may be a hydrogen atom, or a substituted, unsubstituted, branched or unbranched chain of carbon atoms. X and Y may be linkers, each independently comprising a substituted, unsubstituted, branched or unbranched chain of carbon atoms and may optionaily contain a heteroatom, and R 1 , R 2 , R 3 , R 4 , and R 5 may each be independently selected from the group consisting of hydrogen, alkyl, aryl, cycioaiiphatic, and aralkyl and may each optionally contain a hetero atom.

In one embodiment, the compound may be represented by

Formula IV, wherein L 1 may be a moiety formed by esterification of two free esterifiable hydroxy! groups on a diol, and R 1 , R 2 , R 3 , R 4 , and R 5 may each be independently selected from the group consisting of hydrogen, alkyl, aryl. cycioaiiphatic. and aralkyl group, and may optionally contain a hetero atom.

In various embodiments, the diol may be selected from the group consisting

of wherein W is a hydrocarbon group, wherein n is an

integer between 1 and 100, wherein n is an integer between 2 and 12,

, and . in various embodiments, the compound may be selected from the group consisting of Formula V

Another embodiment of the present invention also provides for a compound,

comprising Formula 111, wherein A may be selected from the group consisting of — OC(O)- , -OC(O)O-. and -OC(O)N(R) — , wherein R may be a hydrogen atom, or a substituted, unsubstituted, branched or unbranched chain of carbon atoms, P may be selected from the group consisting of — OC(O) — , and — N(R)C(O) — , wherein R may be a hydrogen atom, or a substituted, unsubstituted, branched or unbranched chain of carbon atoms; X may be a linker comprising a substituted, unsubstituted, branched or unbranched chain of carbon atoms and may optionally contain a heteroatom; and R 1 , R 2 , R 3 , R 4 , and R 5 may each be independently selected from the group consisting of hydrogen, alky!, aryl, cycloaliphatic, and aralkyl, and may each optionally contain a hetero atom

In one embodiment, the compound may be

, Formula Xl, wherein L 2 may be a moiety formed by using a diamine as a linker in the process of producing the compound; and R 1 , R 2 , R 3 , R 4 , and R 5 may each be independently selected from the group consisting of hydrogen, alkyl, aryl, cycloaliphatic, and aralky! group, and may optionally contain a hetero atom.

In various embodiments, the diamine may be selected from the group consisting of: wherein X is a hydrocarbon group,

wherein n is an integer between 1 and 100, and wherein n is an integer between 2 and 12

In various embodiments, the compound may be selected from the group consisting of Formula XLVfi

In various embodiments, the compound may be represented as follows

Formula XVIII, wherein L 3 may be a moiety formed by using an ammoalcohol as a linker in the process of producing the compound, and R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from the group consisting of hydrogen, alkyl, aryl, cycloaliphatic, and aralkyl, and may each contain a hetero atom

In various embodiments, the ammoalcohol may be selected from the group consisting of wherein the Y is a hydrocarbon group, and may

optionally contain a hetero atom, wherein n is an integer between

1 and 100, and wherein n is an integer between 2 and 12

In various embodiments, the compound may be selected from the group consisting of Formula XIX

Formula XX

Another embodiment of the present invention also provides for a compound produced by conjugation of an α-lιpoιc acid and camptothecin or a camptothecin analog modified by reacting with succinic anhydride or glutaric anhydride, wherein the camptothecin analog is represented by Formula I

, wherein R 1 , R 2 , R 3 , R 4 , and R 5 may each be independently selected from the group consisting of hydrogen, alkyl, aryl, cycloaiiphatic, and araikyl, and may optionally contain a hetero atom

In various embodiments, the compound may be selected from the group consisting of

Formula XXV

Formula XXVl

Formula XXVII

Formula XXVIIi

wherein R 1 , R 2 R 3 , R 4 and R 5 may each be independently selected from the group consisting of hydrogen, alkyl, aryl, cycloaliphatic, and aralkyl, and may optionally contain a hetero atom The present embodiments also provide for the following compounds

The present invention also provides a nanosphere, comprising a compound of the present invention. In a further embodiment, the nanosphere may further comprise a compound selected from the group consisting of: a multiple α-lipoic acid- containing hydrophobic compound, α-tocopherol, a nonsteroidal anti-inflammatory drug (NSAID) derivative, and combinations thereof.

The present invention also provides for a method of treating cancer in a subject in need thereof, comprising: providing a composition comprising a compound of the present invention; and administering a therapeutically effective amount of the composition to the subject to treat the cancer.

The present invention also provides a method of treating cancer in a subject in need thereof, comprising: providing a nanosphere of the present invention: and administering a therapeutically effective amount of the nanosphere to the subject to treat the cancer. The present invention also provides a pharmaceutical composition, comprising: a pharmaceutically acceptable carrier or excipient; and a compound of the present invention.

Other features and advantages of the invention will become apparent from the following detailed description, taken in conjunction with the accompanying drawings. which illustrate, by way of example, various features of embodiments of the invention

BRIEF DESCRIPTION OF THE FIGURES Exemplary embodiments are illustrated in the referenced figures It is intended that the embodiments and figures disclosed herein are to be considered illustrative rather than restrictive

Figure 1 depicts the 1 H spectrum of the compound ALA-TEG-OH in accordance with an embodiment of the present invention Figure 2 depicts the 13 C spectrum of the compound ALA-TEG-OH in accordance with an embodiment of the present invention

Figure 3 depicts the 1 H spectrum of the compound ALA 2 (1 , 12-dodecanedιol) in accordance with an embodiment of the present invention

Figure 4 depicts the 13 C spectrum of the compound ALA 2 (I 12-dodecanedιol) in accordance with an embodiment of the present invention

Figure 5 depicts the effect of nanospheres comprising Compound 10 on glioma cell viability in accordance with an embodiment of the present invention Control nanospheres prepared from α-tocopherol only in the absence of Compound 10 (Toco), cells are treated with nanospheres prepared from a mixture of α- tocopherol and Compound 10, which contained 0 1-2 0 μM of Compound 10 Error bar represents ± S D calculated from triplicate determinations

Figure 6 depicts the effect of nanospheres comprising Compound 10 on glioma cell viability in accordance with an embodiment of the present invention Control nanospheres prepared from ALA 2 (1 , 12-dodecanedιol) only in the absence of Compound 10 (DD(ALA2)), cells treated with nanospheres prepared from a mixture of ALA 2 (1 , 12-dodecanedιoS) and Compound 10, which contained 0 1-2 0 μM of Compound 10 Error bar represents ± S D calculated from triplicate determinations

Figure 7 depicts the effect of nanospheres comprising Compound 10 and IbU 2 TEG on glioma cell viability Cells treated with nanospheres prepared from a mixture of IbU 2 TEG and Compound 10, which contained 0 1-2 0 μM of Compound 10 For control experiment with nanospheres prepared from IbU 2 TEG only in the absence of Compound 10, see Fig. 8. Error bar represents ± S. D. calculated from triplicate determinations.

Figure 8 depicts the effect of nanospheres containing IbU 2 TEG on glioma cell viability Cells were treated with nanospheres prepared from IbU 2 TEG only which contained 16-160 μM of IbU 2 TEG. The amount of IbU 2 TEG in this experiment is equal to the amount of IbU 2 TEG contained in the nanospheres prepared from a mixture of IbU 2 TEG and Compound 10 (Fig. 7). Thus, the results serve as a control for the experiment of Fig. 7 in the absence of Compound 10. Error bar represents ± S.D. calculated from triplicate determinations.

DESCRIPTION OF THE INVENTION

All references cited herein are incorporated by reference in their entirety as though fully set forth. Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Singleton et a/., Dictionary of Microbiology and Molecular Biology 3 rd ed., J. Wiley & Sons (New York. NY 2001 ); and March, Advanced Organic Chemistry Reactions, Mechanisms and Structure 5 th ed. , J. Wiley & Sons (New York, NY 2001 ) provide one skilled in the art with a general guide to many of the terms used in the present application. One skilled in the art will recognize many methods and materials similar or equivalent to those described herein, which could be used in the practice of the present invention. Indeed, the present invention is in no way limited to the methods and materials described. For purposes of the present invention, the following terms are defined below. The abbreviation "CPT" as used herein refers to camptothecin {(S)-4-ethyl-4- hydroxy-1 H-pyrano-[3', 4':6, 7]indolizino[1 ,2-b]quinoline-3, 14(4H, 12H)-dione}, which is shown below. The compound is commercially available from numerous sources: e.g., from Sigma Chemical Co. (St. Louis, Mo).

"Camptothecin analogs" as used herein refer to compounds of Formula I

wherein R 1 , R 2 , R 3 , R 4 , and R 5 may each be independently selected from hydrogen or a substituent selected from an alkyl, aryf, cycloaliphatic, and aralkyl group, may be saturated or unsaturated, and may contain hetero atoms (e g , nitrogen, oxygen, sulfur, halogens, etc)

"Antioxidant derivative of camptothecin" and "antioxidant camptothecin derivative " as used herein refer to a derivative of camptothecin that contains an antioxidant [1 ,2]-dιthιolane ring "Antioxidant derivative of a camptothecin analog" and "antioxidant camptothecin analog derivative" as used herein refer to a derivative of a camptothecin analog that contains an antioxidant [1 ,2]-dιthιolane ring

"Camptothecin nanosphere" and " camptothecin nanosphere prodrug" as used herein refer to a nanosphere comprising an antioxidant derivative of camptothecin or an antioxidant derivative of a camptothecin analog The nanosphere may further comprise a multiple α-iipoic acid-containing hydrophobic compound, α-tocopherol, a nonsteroidal anti-inflammatory drug (NSAID) derivative, or combinations thereof

"Cancer" and ' cancerous" refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth Examples of cancer include, but are not limited to breast cancer, colon cancer, lung cancer, prostate cancer, hepatocellular cancer, gastric cancer, pancreatic cancer, cervical cancer ovarian cancer, liver cancer, bladder cancer, cancer of the urinary tract, thyroid cancer, renal cancer, carcinoma, melanoma head and neck cancer, and brain cancer, including, but not limited to, gliomas, glioblastomas, glioblastoma multiforme (GBM), oligodendrogliomas primitive neuroectodermal tumors, low, mid and high grade astrocytomas ependymomas (e g , myxopapiltary ependymoma papillary ependymoma, subependymoma anaplastic ependymoma), oligodendrogliomas, medulloblastomas meningiomas pituitary carcinomas, neuroblastomas and craniopharyngiomas

"Mammal" as used herein refers to any member of the class Mammalia, including, without limitation, humans and nonhuman primates such as chimpanzees and other apes and monkey species, farm animals such as cattle, sheep, pigs, goats and horses, domestic mammals such as dogs and cats, laboratory animals including rodents such as mice, rats and guinea pigs, and the like The term does not denote a particular age or sex Thus, adult and newborn subjects, as well as fetuses, whether male or female, are intended to be included within the scope of this term "Nanosphere" as used herein refers to a particle with a size, in at least one dimension, between about 10 nm to about 1000 nm, and may also include a nanoernulsion

"Nanoprodrug" is used interchangeably with "nanosphere" throughout the application

"Non-steroidal" as used herein distinguishes the anti-inflammatory drugs from steroids, which have a similar anti-inflammatory action

"NSAID derivative" as used herein refers to a compound in which as least one NSAID molecule is coupled to a polyol, for example, through esteπfication

"Polyol" as used herein refers to a compound that contains at least two free esteπfiable hydroxyl groups "Therapeutic agent" as used herein refers to any substance used internally or externally as a medicine for the treatment, cure, prevention slowing down, or lessening of a disease or disorder, even if the treatment, cure, prevention, slowing down, or lessening of the disease or disorder is ultimately unsuccessful

"Therapeutically effective amount' as used herein refers to an amount which is capable of achieving beneficial results in a patient with a condition or a disease condition in which treatment is sought A therapeutically effective amount can be determined on an individual basis and will be based, at least in part, on consideration of the physiological characteristics of the mammal, the type of delivery system or therapeutic technique used and the time of administration relative to the progression of the disease

Treatment" and treating, ' as used herein refer to both therapeutic treatment and prophylactic or preventative measures wherein the object is to prevent slow down and/or alleviate the disease or disease condition even if the treatment is ultimately unsuccessful

The present invention provides for antioxidant derivatives of camptothecin and antioxidant derivatives of camptothecin analogs These derivatives are useful for treating various types of cancer In addition, the present invention provides antioxidant-antineoplastic nanospheres comprising the antioxidant derivatives of camptothecin or antioxidant derivatives of camptothectn analogs and methods of preparing the antioxidant-antineoplastic nanospheres These nanospheres can operate as prodrugs In one embodiment, the camptothecin nanosphere prodrugs are capable of releasing camptothecin or a camptothecin analog for a prolonged period of time In another embodiment, the camptothecin nanosphere prodrugs are capable of serving as a vehicle for the delivery of additional pharmaceuticals

In one embodiment, an antioxidant derivative of camptothecin and/or an antioxidant derivative of a camptothecin analog may be represented by Formula Il

wherein A and B may be independently selected from the group consisting of -OC(O)- -OC(O)O-, and -OC(O)N(R)-, wherein R may be a hydrogen atom, or a substituted, unsubstituted, branched or unbranched chain of carbon atoms and may contain heteroatoms (e g , nitrogen, oxygen, sulfur, etc ), wherein X and Y may be each be a linker that may be a substituted, unsubstituted, branched or unbranched chain of carbon atoms and may contain heteroatoms (e g , nitrogen, oxygen, sulfur, etc ) and wherein R 1 , R 2 , R 3 , R 4 , and R 5 may each be independently selected from hydrogen or a substituent selected from an alkyl aryl, cycloaliphatic, and aralkyl group may be saturated or unsaturated and may contain hetero atoms (e g , nitrogen oxygen, sulfur, halogens, etc)

In one embodiment, an antioxidant derivative of camptothecin and/or antioxidant derivative of a camptothecin analog is prepared by the conjugation of a camptothecin or a camptothecin analog and an α-lipoic acid and is represented by Formula III: ~v wherein A may be selected from the group consisting of — OC(O) — , — OC(O)O — . and — OC(O)N(R) — , wherein R may be a hydrogen atom, or a substituted, unsubstituted, branched or unbranched chain of carbon atoms and may contain heteroatoms (e.g., nitrogen, oxygen, sulfur, etc.); wherein P may be selected from the group consisting of — OC(O) — , and — N(R)C(O) — , wherein R may be a hydrogen atom, or a substituted, unsubstituted, branched or unbranched chain of carbon atoms and may contain heteroatoms (e.g., nitrogen, oxygen, sulfur, etc.); wherein X may be a linker that may be a substituted, unsubstituted, branched or unbranched chain of carbon atoms and may contain heteroatoms (e.g.. nitrogen, oxygen, sulfur, etc.); and wherein R 1 , R 2 , R 3 , R 4 , and R 5 may each be independently selected from hydrogen or a substituent selected from an alky!, aryl, cycloaliphatic, and aralkyl group, may be saturated or unsaturated, and may contain hetero atoms (e.g., nitrogen, oxygen, sulfur, halogens, etc).

In another embodiment, an antioxidant derivative of camptothecin and/or antioxidant derivative of a camptothecin analog is prepared by the conjugation of camptothecin or a camptothecin analog and α-lipoic acid via a diol and is represented Formula IV:

wherein Li may be a moiety formed by esterification of two free esterifiable hydroxy! groups on a diol; and wherein R 1 , R 2 , R 3 , R 4 , and R 5 may each be independently selected from hydrogen or a substituent selected from an alky!, aryl, cycloaiϊphatic. and aralkyl group, may be saturated or unsaturated, and may contain hetero atoms (e.g. , nitrogen, oxygen, sulfur, halogens, etc). In various embodiments diols that are useful in the present invention may be represented by the following formula wherein W may be a hydrocarbon group, for example, an alkyl, aryl, cycloatiphatic or aralkyl group, and may be saturated or unsaturated W may also contain hetero atoms (e g , nitrogen oxygen, sulfur, etc )

Additional examples of dtols that are useful in the present invention include, but are not limited to commercially available one as follows wherein n is an integer between 1 and 100 wherein n is an integer between 2 and 12 1 ,4-Bιs{2-hydroxyethyl)-pιperazιne 1 ,3-CycIopentanedιol

1 4-cyclohexanedιol

Examples of particularly useful antioxidant derivatives of camptothecin and/or antioxidant derivatives of camptothecin analogs of this embodiment are represented by the following formulas

wherein R 1 R 2 R 3 , R 4 , and R 5 may each be independently selected from hydrogen or a substttuent selected from an alkyl aryl cycloaliphatic and arafkyf group may be saturated or unsaturated, and may contain hetero atoms (e g , nitrogen, oxygen, sulfur halogens, etc)

One exemplary compound and its synthesis are shown below

In another embodiment an antioxidant derivative of a camptothecin and/or antioxidant derivative of a camptothecin analog is prepared by the conjugation of camptothecin or a camptothecin analog and an α-lspoic acid vsa a diamine and is represented by Formula Xl wherein L 2 may be a moiety formed by using a diamine as the linker in the process of producing the antioxidant camptothecin derivative or the antioxidant camptothecin analog derivative; and wherein R 1 , R 2 , R 3 , R 4 , R5 may each be independently selected from hydrogen or a substituent selected from an alkyl, aryl, cycloaliphatic, and aralkyl group, may be saturated or unsaturated, and may contain hetero atoms (e g , nitrogen, oxygen, sulfur, halogens, etc).

In one embodiment, diamines that are useful in the present invention may be represented by the following formula:

wherein X may be a hydrocarbon group; for example, an alkyl, aryl, cycloaliphatic or aralkyl group; and may be saturated or unsaturated. X may also contain hetero atoms (e.g., nitrogen, oxygen, sulfur, etc.). in other embodiments, diamines that are useful in the present inventive compounds include, but are not limited to commercially available ones as follows: wherein n is an integer between 1 and 100 wherein n is an integer between 2 and 12. Examples of particularly useful antioxidant derivatives of camptothecin and/or antioxidant derivatives of camptotheαn analogs of this embodiment are represented by the following formulas: Formula XIl

wherein R 1 , R 2 , R 3 , R A , and R 5 may each be independently selected from hydrogen or a substftuent selected from an alkyl aryt, cycloaliphatic, and aralkyl group, may be saturated or unsaturated, and may contain hetero atoms (e g , nitrogen, oxygen sulfur halogens, etc) One exemplary compound and its synthesis are shown below.

In another embodiment, an antioxidant derivative of camptothecin and/or antioxidant derivative of a camptothecin analog is prepared by the conjugation of camptothectn or a camptothecin analog and an α-lipoic acid via an aminoaicohoi and is represented by Formula XVIH:

wherein L 3 may be a moiety formed by using an aminoaicohoi as the linker in the process of producing the antioxidant camptothecin derivative or the antioxidant camptothecin analog derivative; and wherein R 1 , R 2 , R 3 , R 4 , and R 5 may each be independently selected from hydrogen or a substituent selected from an alky!, aryl, cycloaltphatic, and aralkyl group, may be saturated or unsaturated, and may contain hetero atoms (e.g., nitrogen, oxygen, sulfur, halogens, etc). Aminoalcohols that are useful in the present invention may be represented by the following formula wherein Y may be a hydrocarbon group, for example, an alkyl, aryl, cycloaliphatic or aralkyl group, and may be saturated or unsaturated Y may also contain hetero atoms {e g , nitrogen, oxygen sulfur, etc )

Examples of aminoalcohols that are useful in the present inventive compounds include, but are not limited to commercially available one as follows wherein n is an integer between 1 and 100 wherein n is an integer between 2 and 12

Examples of particularly useful antioxidant derivatives of camptothecin and/or antioxidant derivatives of camptothecin analogs of this embodiment are represented by the following formulas Formula XIX

Formula XX

Formula XXI

Formula XXlI

Formula XXIII

Formula XXIV

wherein R 1 , R 2 , R 3 , R 4 , and R 5 may each be independently selected from hydrogen or a substituent selected from an alky!, aryl, cycloaliphatic, and aralkyl group, may be saturated or unsaturated and may contain hetero atoms (e g , nitrogen, oxygen, sulfur, halogens, etc)

One exemplary compound and its synthesis are shown below

Additional embodiments of the present invention provide for the following compounds

In another embodiment, the camptothecin analogs are modified by reaction with succinic anhydride or glutaπc anhydride and an antioxidant derivative of camptothecin and/or antioxidant derivative of a camptothecin analog is prepared by the conjugation of an α-lipoιc acid and the modified camptothecin or camptothecin analog One exemplary compound and its synthesis are shown below.

Formula XXV wherein R 1 , R 2 , R 3 , R 4 , and R 5 may each be independentiy selected from hydrogen or a substituent selected from an alkyl, aryl, cyctoaliphatic, and aralkyl group may be saturated or unsaturated, and may contain hetero atoms (e g nitrogen, oxygen, sulfur, halogens, etc)

Additional examples of particularly useful antioxidant derivatives of camptothecin and/or antioxidant derivatives of camptothecin analogs are represented by formulas as follows Formula XXVI

Formula XXVIf

wherein R 1 R 2 R 3 R 4 , and R 5 may each be independently selected from hydrogen or a substituent selected from an alky! aryi cycloalsphatic and araikyl group may be saturated or unsaturated, and may contain hetero atoms (e g , nitrogen, oxygen, sulfur, halogens, etc)

In one particular embodiment, each of R 1 through R 5 of the formulas and/or compounds described above is H, and is shown below

A general scheme for the synthesis of the antioxidant derivatives of camptothecin and antioxidant derivatives of camptothecin analogs and preparation of the antioxidant-antineoplastic nanospheres are described in the ensuing examples The synthetic procedure is both simple and versatile and leads to the synthesis of the antioxidant derivatives of camptothecin and antioxidant derivatives of camptothecin analogs varying in size and hydrophobicity

The present invention also provides for methods of treating cancer In one embodiment, the antioxidant camptothecin derivatives and the antioxidant camptothecin analog derivatives are used for treating cancer The method comprises providing a pharmaceutical composition comprising an antioxidant camptothecin derivative or an antioxidant camptothecin analog derivative of the present invention, and administering a therapeutically effective amount of the pharmaceutical composition to a subject in need thereof In a further embodiment, the pharmaceutical composition comprises a pharmaceutically acceptable carrier or excipient

In one particular embodiment, the antioxidant camptothecin derivatives and the antioxidant camptothecin analog derivatives are used to treat a brain tumor The method comprises providing a pharmaceutical composition comprising an antioxidant camptothecm derivative or an antioxidant camptothecin analog derivative of the present invention, and administering a therapeutically effective amount of the pharmaceutical composition to a subject in need of treatment for a brain tumor Additional embodiments of the present invention provide for methods of preparing the camptothecin nanosphere prodrugs from the antioxidant derivatives of camptothecin and antioxidant derivatives of camptothecin analogs

In one embodiment, the antioxidant derivatives of camptothecin and antioxidant derivatives of camptothecin analogs are prepared into antioxidant-antineoplastic nanoparticles (e g., nanospheres) by blending with other antioxidant α-Iipoic acid-containing hydrophobic compounds. These compounds are disclosed in U S Provisional Application Serial No. 61/018,749, filed January 3, 2008, and International Application Publication No. WO 2009/086547, filed December 30, 2008, which are incorporated by reference in their entirety as though fully set forth. Examples of these antioxidant α-lipoic acid-containing hydrophobic compounds include, but are not limited to the following :

Antioxidant α-lipoic acid-containing hydrophobic compounds represented by Formula Ia

(Ia), wherein X may be selected from the group consisting of a substituted, unsubstituted, branched or unbranched chain of carbon atoms, and may optionally contain a heteroatom, Y may be selected from the group consisting of a branched and unbranched alkyl, branched and unbranched alkenyl, branched and unbranched alkynyl, heteroatom-containing branched and unbranched alkyl, heteroatom-containing branched and unbranched alkenyl, heteroatom- containing branched and unbranched alkynyl, aryl. cyclic aliphatic, cyclic aromatic, heterocyclic, and aromatic heterocyclic group; and n may be an integer of at least one. In particular embodiments, n may be an integer from 1 to 4; and X may be an unsubstituted, unbranched chain of 1 to 6 carbon atoms. In one embodiment, the dithiolane moiety in Formula Ia may be an α-lipoic acid and is represented by Formula Ha:

In various embodiments, Y may be a moiety formed by esteπfication of the hydroxyl groups of a polyol In various embodiments, the polyol may be selected from the group consisting of wherein n is an integer between 1 and 4 and wherein n is an integer between 3 and 16

One example of a particularly useful multiple α-hpoic acid-containing hydrophobic compound is represented as follows

In another embodiment, the antioxidant derivatives of camptothecin and antioxidant derivatives of camptothecin analogs are prepared into nanoparticles (e g , nanospheres) by blending with a non-steroidal anti-inflammatory drug (NSAID) derivative disclosed in International Application No PCT/US09/39956, filed on April 8, 2009, which is incorporated by reference in its entirety as though fully set forth Examples of these NSAID derivatives include but are not limited to the following NSAID derivatives of Formula Ib

(Ib), wherein the A may be selected from the group consisting of branched and unbranched alkyl, branched and unbranched alkenyl, branched and unbranched alkynyl, heteroatom-containing branched and unbranched alkyl heteroatom-containing branched and unbranched alkenyl, heteroatom- containing branched and unbranched alkynyl aryl, cyclic aliphatic cyclic aromatic, heterocyclic, and aromatfc heterocyclic groups, and n may be an integer of at least two

In various embodiments, A may be a moiety that is formed by esterifi cation of at least two free esterifiable hydroxyl groups on a polyol In one embodiment, the polyol may be , wherein n on the polyol may be an integer between 1

and 6 In another embodiment, the polyo) may be , wherein n on the polyol may be an integer between 3 and 16

In other embodiments, A may be formed from esterifi cation of a polyol selected from group consisting of an ethylene glycol (Methylene glycol, tπethylene glycol, tetraethylene glycol, pentaethylene glycol, hexaethylene glycol, 1 ,3- propanediol, and 1 ,4-butanedιol

In various embodiments, the NSAID may be selected from the group consisting of aspirin, ibuprofen, flurbiprofen, ketoprofen, fenoprofen, fenbufen, naproxen, indomethacin, diclofenac, ketorolac, tolmetin, flufenamic acid, mefenamic acid, tolfenamic aod, meclofenamic acid niflumic acid, sulindac, sulindac sulfide and combinations thereof

NSAID derivatives of Formula lib

(lib) wherein X may be selected from the group consisting of a substituted, unsubstituted, branched or unbranched chain of carbon atoms and may optionally contain a heteroatom, A may be selected from the group consisting of branched and unbranched alkyl, branched and unbranched alkenyl, branched and unbranched alkynyl, heteroatom-containing branched and unbranched alkyl, heteroatom-containing branched and unbranched alkenyl, heteroatom- containing branched and unbranched alkynyl, aryl, cyclic aliphatic, cyclic aromatic, heterocyclic, and aromatic heterocyclic groups, n may be an integer of at least one, and m may be an integer of at least one

In various embodiments A may be a moiety that is formed by esterificatfon of at least two free esterifiable hydroxyl g olyol In particular embodiments the polyol may be , wherein n on the polyol may be an integer

between 1 and 6, or , wherein n on the polyol may be an integer between 3 and 16

In various embodiments, the NSAID may be selected from the group consisting of aspirin, ibuprofen, flurbiprofen, ketoprofen, fenoprofen, fenbufen naproxen indomethacm, diclofenac, ketorolac, tolmetin, flufenamic acid, mefenamic acid, tolfenamic acid, meclofenamic acid, niflumic acid, sulindac, sulindac sulfide and combinations thereof

In one embodiment the dithiolane moiety may be an α-lιpoιc acid ("ALA') and is represented by formula 11 Ib

in another embodiment, the antioxidant derivatives of camptothecm and antioxidant derivatives of camptothecm analogs are prepared into nanopartides by blending with an antioxidant α-lιpoιc acid-containing hydrophobic compound and a non-steroidal anti-inflammatory drug (NSAID) derivative disclosed in U S Provisional Application Serial No 61/018,749, filed January 3, 2008, and International Application Publication No WO 2009/086547, filed December 30, 2008, and in International Application No PCT/US09/39956, filed on April 8, 2009, respectively, which are incorporated by reference in their entirety as though fully set forth In a further embodiment, α- tocopherol is also added to the mixture for the formation of nanopartides and camptothecin nanospheres of the present invention Its addition can provide additional stability to the nanopartides and nanospheres of the present invention

The camptothecm nanospheres described above can be used for treating cancer They can operate as prodrugs as discussed above The method comprises providing a composition comprising a camptothecm nanosphere of the present invention, and administering a therapeutically effective amount of the composition to a subject in need thereof

In one particular embodiment, the camptothecin nanospheres are used to treat a brain tumor The method comprises providing a composition comprising a camptothecin nanosphere, and administering a therapeutically effective amount of the composition to a subject in need of treatment for a brain tumor

In another embodiment, the camptothecin nanospheres may be used as a carrier of an additional therapeutic agent In one embodiment, the second therapeutic agent is a chemotherapeutic agent that is useful for cancer treatment Accordingly, one embodiment provides for a composition comprising a camptothecin nanosphere of the present invention and an additional therapeutic agent

In one embodiment, the present invention provides for a method to enhance the cytotoxicity of an antineoplastic drug for treatment of a disorder of abnormal ceil proliferation The method comprises providing a composition comprising a camptothecin nanosphere of the present invention, and administering a therapeutically effective amount of the composition and an antineoplastic drug to a subject in need of the treatment to enhance the cytotoxicity of the antineoplastic drug

in various embodiments, the present invention provides pharmaceutical compositions including a pharmaceutically acceptable excipient along with a therapeutically effective amount of the antioxidant derivatives of camptothecin and/or antioxidant derivatives of camptothecin analogs, or the camptothecin nanosphere prodrugs "Pharmaceutically acceptable excipient" means an excipient that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic, and desirable, and includes excipients that are acceptable for veterinary use as well as for human pharmaceutical use Such excipients may be solid, liquid, semisolid, or, in the case of an aerosol composition, gaseous

In various embodiments, the pharmaceutical compositions according to the invention may be formulated for delivery via any route of administration 'Route of administration" may refer to any administration pathway known in the art, including but not limited to aerosol nasal, oral, transmucosal transdermal, parenteral enteral or ocular "Transdermal" administration may be accomplished using a topical cream or ointment or by means of a transdermal patch 'Parenteral" refers to a route of administration that is generally associated with injection, including intraorbital infusion, intraarterial, intracapsular, intracardiac, intradermal, intramuscular, intraperitoneal, intra pulmonary, intraspinal, intrasternal, intrathecal, intrauterine, intravenous, subarachnoid, subcapsular, subcutaneous, transmucosat, or transtracheal Via the parenteral route, the compositions may be in the form of solutions or suspensions for infusion or for injection, or as lyophiltzed powders Via the enteral route, the pharmaceutical compositions can be in the form of tablets, gel capsules, sugar-coated tablets, syrups, suspensions, solutions, powders, granules, emulsions, microspheres or nanospheres or lipid vesicles or polymer vesicles allowing controlled release Via the parenteral route, the compositions may be in the form of solutions or suspensions for infusion or for injection Via the topical route, the pharmaceutical compositions based on compounds according to the invention may be formulated for treating the skin and mucous membranes and are in the form of ointments, creams, milks, salves powders, impregnated pads, solutions, gels, sprays, lotions or suspensions They can also be in the form of microspheres or nanospheres or lipid vesicles or polymer vesicles or polymer patches and hydrogels allowing controlled release These topical-route compositions can be either in anhydrous form or in aqueous form depending on the clinical indication Via the ocular route, they may be in the form of eye drops

The pharmaceutical compositions according to the invention can also contain any pharmaceutically acceptable carrier "Pharmaceutically acceptable carrier' as used herein refers to a pharmaceutically acceptable material composition, or vehicle that is involved in carrying or transporting a compound of interest from one tissue organ, or portion of the body to another tissue, organ, or portion of the body For example, the carrier may be a liquid or solid filler, diluent, exctpsent, solvent, or encapsulating material, or a combination thereof Each component of the carrier must be 'pharmaceutically acceptable" in that it must be compatible with the other ingredients of the formulation It must also be suitable for use in contact with any tissues or organs with which it may come in contact, meaning that it must not carry a risk of toxicity, irritation, allergic response, immunogenicity, or any other complication that excessively outweighs its therapeutic benefits

The pharmaceutical compositions according to the invention can also be encapsulated, tabfeted or prepared in an emulsion or syrup for oral administration Pharmaceutically acceptable solid or liquid carriers may be added to enhance or stabilize the composition, or to facilitate preparation of the composition Liquid carriers include syrup, peanut oil, olive oil, glycerin, saline, alcohols and water Solid carriers include starch, lactose, calcium sulfate, dihydrate, terra alba magnesium stearate or stearic acid, talc, pectin, acacia, agar or gelatin The earner may also include a sustained release material such as glyceryl monostearate or glyceryl distearate alone or with a wax

The pharmaceutical preparations are made following the conventional techniques of pharmacy involving milling, mixing, granulation, and compressing, when necessary, for tablet forms, or milling, mixing and filling for hard gelatin capsule forms When a liquid carrier is used, the preparation will be in the form of a syrup, elixir, emulsion or an aqueous or non-aqueous suspension Such a liquid formulation may be administered directly p o or filled into a soft gelatin capsule

The pharmaceutical compositions according to the invention may be delivered in a therapeutically effective amount The precise therapeutically effective amount is that amount of the composition that will yield the most effective results in terms of efficacy of treatment in a given subject This amount will vary depending upon a variety of factors including but not limited to the characteristics of the therapeutic compound (including activity, pharmacokinetics, pharmacodynamics, and bioavailability), the physiological condition of the subject (including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage, and type of medication), the nature of the pharmaceutically acceptable carrier or carriers in the formulation, and the route of administration One skilled in the clinical and pharmacological arts will be able to determine a therapeutically effective amount through routine experimentation, for instance by monitoring a subject's response to administration of a compound and adjusting the dosage accordingly For additional guidance, see Remington The Science and Practice of Pharmacy (Gennaro ed 20th edition Williams & Wilkins PA, USA) (2000) Typical dosages of an effective amount of the antioxidant derivatives of camptothecin and/or antioxidant derivatives of camptothecin analogs, or the camptothecin nanosphere prodrugs can be in the ranges recommended by the manufacturer where known therapeutic compounds are used, and also as indicated to the skilled artisan by the in vitro responses or responses in animal models Such dosages typically can be reduced by up to about one order of magnitude in concentration or amount without losing the relevant biological activity Thus, the actual dosage will depend upon the judgment of the physician, the condition of the patient, and the effectiveness of the therapeutic method based, for example on the in vitro responsiveness of the relevant primary cultured cells or histocultured tissue sample, such as biopsied malignant tumors, or the responses observed in the appropriate animal models, as previously described

The present invention is also directed to a kit to treat cancer The kit is an assemblage of materials or components, including at least one of the inventive compositions Thus in some embodiments the kit contains a composition including antioxidant derivatives of camptothecin and/or antioxidant derivatives of camptothecin analogs, or the camptothecin nanospheres of the present invention as described above

The exact nature of the components configured in the inventive kit depends on its intended purpose For example, some embodiments are configured for the purpose of treating cancer In one embodiment, the kit is configured particularly for the purpose of treating mammalian subjects In another embodiment, the kit is configured particularly for the purpose of treating human subjects In further embodiments, the kit is configured for veterinary applications treating subjects such as, but not limited to, farm animals, domestic animals, and laboratory animals

Instructions for use may be included in the kit "Instructions for use" typically include a tangible expression describing the technique to be employed in using the components of the kit to effect a desired outcome, such as to treat cancer Optionally, the kit also contains other useful components, such as, diluents, buffers, pharmaceutically acceptable carriers, syringes, catheters applicators, pipetting or measuring tools, or other useful paraphernalia as will be readily recognized by those of skill in the art The materials or components assembled in the kit can be provided to the practitioner stored in any convenient and suitable ways that preserve their operability and utility For example the components can be in dissolved, dehydrated, or lyophilized form, they can be provided at room, refrigerated or frozen temperatures The components are typically contained in suitable packaging mateπal(s) As employed herein, the phrase "packaging material" refers to one or more physical structures used to house the contents of the kit, such as inventive compositions and the like The packaging material is constructed by well known methods, preferably to provide a sterile, contaminant-free environment As used herein, the term "package" refers to a suitable solid matrix or material such as glass, plastic, paper, foil, and the like, capable of holding the individual kit components Thus, for example, a package can be a glass vial used to contain suitable quantities of an inventive composition containing antioxidant derivatives of camptothecin and/or antioxidant derivative of camptothecin analogs, or an inventive composition containing antioxidant-antineoplastic nanospheres The packaging material generally has an external label which indicates the contents and/or purpose of the kit and/or its components

EXAMPLES The following examples are provided to better illustrate the claimed invention and are not to be interpreted as limiting the scope of the invention To the extent that specific materials are mentioned, it is merely for purposes of illustration and is not intended to limit the invention One skilled in the art may develop equivalent means or reactants without the exercise of inventive capacity and without departing from the scope of the invention

Example 1

General Procedures and Materials

Unless otherwise noted, solvents and chemicals were obtained as highest purity from Sigma-Aldπch Chemical Co (St Louis, MO, USA) and used without further preparation Chromatographic purification of all newly synthesized compounds was performed using silica gel (60 A 200-400 mesh) The compounds were analyzed by thin layer chromatography (TLC): sificagel plates (Merck 60 F254); compounds were visualized by irritation by treatment with a solution of 1 .5 g of KMnO 4 , 10 g K 2 CO 3 , and 125 mL 10% NaOH in 200 mL of H 2 O, followed by gentle heating, HPLC analysis was performed on Merck-Hitachi analytical LaChrom D-7000

HPLC/UV detector system with CAPCELL PAK 1 Type SG 120 (phenomenex) Ci 8 reversed phase column (250/4.6 mm, 5 μm). The composition of the mobile phase (acetonitrile/water mixture containing 0.1 % (v/v) trifluoroacetic acid) was adjusted for camptothecin and α-lipoic acid derivatives, multiple α-lipoic acid-containing compounds and NSAID derivatives in order to provide an appropriate retention time and separation.

Example 2 Synthesis of the camptothecin and α-lipoic acid derivatives A α-Lϊpoic acid (ALA, 2.06 g, 10 mmol ) and a diol compound (tetraethylene glycol, TEG) (30 mmol) in 50 mL of anhydrous dichSoromethane (DCM) were reacted with 4~(dimethylamino)-pyridine (DMAP, 15 mmol) in the presence of a molecular sieve (60 Λ, 10-20 mesh beads) for 10 min at room temperature, N-(S- Dimethylamfnopropyl)-N-ethyfcarbodϋmide hydrochloride (EDCI, 2,3 g, 12 mmol) was added portionwise over 10 min and the reaction mixture was stirred for 5 h at room temperature in the dark, filtered, and then concentrated under vacuum to reduce the volume. The resulting reaction mixture was purified using silica gel by direct loading onto the column without further preparation. The solvent was removed under reduced pressure to give the products. A suspension of camptothecin (0.4 mmol), triphosgene (0, 15 mmol), and

DMAP (1.3 mmol) in anhydrous DCM was stirred for 10 min. The mono-ALA-TEG (0.4 mmoi) was added and the reaction mixture was stirred for 24 h. The reaction mixture was concentrated under vacuum to reduce the volume. The resulting reaction mixture was purified using silica get by direct loading onto the column without further preparation. The solvent was removed under reduced pressure to give the products. The same procedure was used for the synthesis of the compounds 1-3 (See Scheme 1). Scheme 1 Synthesis of the camptothecin and α-lιpoιc acid derivatives A

Compound 23

Compound 1 (Camptothecin)-diethylene glycol-(α-!ιpoιc acid)

Compound 2 (Camptothecιrι)-tπethylene glycoHα-I'POic acid) Compound 3, (Camptothecin)-hexaethy)ene glyco!-(α-lipoic acid) '

Example 3 Synthesis of the camptothecin and α-lipoic acid derivatives B

Mono-ALA-TEG was prepared as described in Example 1 Mono-ALA-TEG (10 mmol) and succinic anhydride (100 mmot) were dissolved in 100 mL of pyridine and the reaction mixture was stirred for 3 h at room temperature The reaction mixture was concentrated under vacuum to reduce the volume The resulting reaction mixture was purified using silica gel by direct loading onto the column without further preparation The solvent was removed under reduced pressure to give the products.

Mono-ALA-TEG-SA {5 mmol) and camptothecin (5 mmol) in 10 mL of anhydrous dϊchloromethane (DCM) were reacted with 4-(dimethylamino)-pyrϊdine (DMAP, 10 mmol) in the presence of molecular sieve {60 A. 10-20 mesh beads) for 10 min at room temperature. A/-(3-Dimethylaminopropyi)-N-ethylcarbodiimide hydrochloride (EDCt, 10 mmol) was added portionwise over 10 min and the reaction mixture was stirred for 5 h at room temperature in the dark, filtered, and then concentrated under vacuum to reduce the volume. The resuiting reaction mixture was purified using silica gei by direct loading onto the column without further preparation. The solvent was removed under reduced pressure to give the products. The same procedure was used for the synthesis of the compounds 4-6 (See Scheme 2) Scheme 2 Synthesis of the camptothecin and α-lipoic acid derivatives B

Compound 4 (Camptothecsn)-diethylene giycol-(α-Iιpoic acid)

Compound 5 (Camptotheαn)-tπethylene glycot-(α-Iιpoιc acιd) Compound 6 (Camptothecιn)-hexaethylene glycol-(α-lιpoιc acid)

Scheme 3

Synthesis of the camptothecin and α-lψoic acid derivatives C The derivatives C were prepared as described in Example 2

Compound 7, (Camptothecιn)-dιethylene glycol-(α-hpoιc acid) Compound 8, (Camptothecm)-triethylene glycol-(α-lιpoic acid):

Compound 9, (Camptothecin)-hexaethylene glycol-(α-lipoic acid)

Example 4

Synthesis of α-lipoic acid derivative ALA 2 (1, 12-dodecanedioi) α-ϋpoic acid (2.48 g, 12 mmol, 1 2 equiv.) and 1 ,12-dodecanediol (10 mmo1 OH, 1.0 equiv.) in 20 ml_ of anhydrous dichloromethane (DCM) were reacted with A- (dιmethylamino)-pyridine (DMAP, 1.47 g, 12 mmol, 1.2 equiv,) in the presence of molecular sieve (60 A, 10-20 mesh beads) for 10 min at room temperature, N-(3- DimethyIamιnopropyl)-N-ethylcarbodiimide hydrochloride (EDCI, 2.3 g, 12 mmol, 1.2 equiv,) was added portionwise over 10 min and the reaction mixture was stirred for 12 h at room temperature in the dark, filtered, and then concentrated under vacuum to reduce the volume. The resulting reaction mixture was purified using silica gel by direct loading onto the column without further preparation. The solvent was removed under reduced pressure to give the products. 1 H NMR and 13 C NMR spectra of the compound are provided U.S. Provisional Application Serial No. 61/018,749, filed January 3, 2008, and

International Application Publication No WO 2009/086547, filed December 30, 2008, herein incorporated by reference in their entirety as though fully set forth, provide additional examples of synthesizing α-hpoic acid derivatives that are used in the present invention. Scheme 4 Synthesis of α-Iipoic acid derivatives B

Example 5

Preparation of the Antioxidant-Antineoplastic Nanospheres Nanospheres were prepared according to the method using spontaneous emuisification with slight modification Briefly, 15 mg of the compounds (mixture of camptothecin derivatives and ALA 2 (1 , 12-dodecanediol) were dissolved in acetone (5 HnL 1 0 1% polysorbate 80) The organic solution was poured under moderate stirring on a magnetic plate into an aqueous phase prepared by dissolving 25 mg of Pluronic F68 in 10 mL bidistilled water (0 25% w/v) Following 15 mm of magnetic stirring, the acetone was removed under reduced pressure at room temperature The nanospheres were filtered through 0 8 μm hydrophilic syringe filter and stored at 4 °C The hydrodynamic size measurement and size distribution of the nanospheres was performed by the dynamic light scattering (DLS) using a Coulter N4~Pius Submicron Particle Sizer (Coulter Corporation, Miami FL)

Additionally, 25 mg of the compounds (mixture of the antioxidant camptothecin derivatives, multiple α-lιpoιc acid containing compounds and α- tocopherol) were dissolved tn acetone (5 mL, 0 1 % polysorbate 80) The organic solution was poured under moderate stirring on a magnetic plate into an aqueous phase prepared by dissolving 25 mg of Pluronic F68 in 10 mL bidistilled water (0 25% w/v) Following 15 mm of magnetic stirring the acetone was removed under reduced pressure at room temperature The nanospheres were filtered through 0 8 μm hydrophilic syringe filter and stored at 4 °C. The hydrodynamic size measurement and size distribution of the nanospheres was performed by the dynamic light scattering (DLS) using a Coulter N4-P!us Submicron Particle Sizer (Coulter Corporation, Miami, FL). Control nanosphere was prepared from multiple u- lipoic acid containing compounds and α-tocopheroi in the absence of camptothecin derivatives.

Table 1. Size and Polydispersitv Index (P.I.): Antioxidant-Antineoplastic Nanosphere I

Example 6

Preparation of the Antioxidant-Antineoplastic Nanospheres Nanospheres were prepared according to the method described in Example 5 using spontaneous emulsification from 25 mg of the compounds (mixture of camptothecin derivatives and α~tocopherol). Control nanosphere was prepared from α-tocopherol or IbU 2 TEG in the absence of camptothecin derivatives.

Table 2. Size and Poiydispersity Index (P.I.):

Antioxidant-Antineoplastic Nanosphere Il

Example 7

Preparation of the Anti-inflammatory-Antineoplastic Nanosphere Nanospheres were prepared according to the method described in Example 5 using spontaneous emulsification from 25 f th compounds (mixture of camptothecin derivatives, derivatives of non-steroidal antiinflammatory drugs (NSAIDs) and α-tocophero!) Control nanosphere was prepared from α-tocopherol or a mixture of α-tocopherol and derivatives of NSAIDs in the absence of camptothecin derivatives

Table 3 Size and Polydispersity Index (P.I. ) Anti-inflammatory-Antineoplastic

Nanosphere

Example 8 Anticancer and antiproliferative effects of the nanospheres comprising camptothecin derivatives

The U87-MG human glioma cell line was obtained from American Type Culture Collection (ATCC) (Rockville, Maryland, USA) The cells were grown and maintained in Minimum Essential Medium (MEM) (Invitrogen) containing antibiotics 100 U/mL penicillin (Invitrogen) and 100 μg/mL streptomycin (Invitrogen), and supplemented with 10% fetal bovine serum (FBS) (Invitrogen) Cells were kept at 37°C in a humidified atmosphere including 5% CO 2

Nanospheres were prepared from the mixture of Compound 10 (1 mg), α- tocopherol (25 mg), and multiple α-lipoic acid containing compound (ALA^Glycerol, or Compound 10 (1 mg) and α-tocopherol (25 mg), or Compound 10 (1 mg), α- tocophero! (25 mg), and NSAID derivative IbU 2 TEG as described in Examples 5, 6, and 7, and dialyzed in phosphate buffered saline (PBS) overnight The human glioma cells (U87-MG) were seeded in a 6-wel! flask at 10 5 cells/well and allowed to grow for 24 h The medium was changed and the cells were treated with nanospheres at final concentration ranging from 0 1 to 2 μM for the Compound 10 After a 4-day treatment the medium was remove, cells were washed with PBS and 1 mL of 0 25% trypsin/EDTA (Gibco) was added to detach the cells The cells were counted immediately in a hemacytometer (See FiGS. 5-8.). Control culture was grown in the absence of nanospheres.

Example 9 Synthesis of bifunctional derivatives of α-lipoic acid and NSAlDs α-Lipoic acid (ALA, 10 mrnol) and tetraethylene glycol (TEG, 30 mmof) in 50 ml of anhydrous dichloromethane (DCM) were reacted with 4-(dimethylamino)- pyridine (DMAP, 15 mmot) in the presence of a motecufar sieve (Fluka, 3 A, 10-20 rnesh beads) for 10 min at room temperature. N-(3-Dimethylaminopropyf)-N- ethylcarbodiimide hydrochloride (EDCi, 10 rnrnof) was added portionwise over 10 min and the reaction mixture was stirred for 5 h at room temperature in the dark, filtered, and then concentrated under vacuum to reduce the volume. The product ALA-TEG-OH and dimeric byproduct ALA-TEG-ALA were purified using cofumn chromatography by loading the concentrated reaction mixture on the column without prior preparation and characterized as described above. Mono-ALA derivatives of TEG (3.8 mmoi) and NSAiDs (4.1 mmoL indomethacin: Ind, ibuprofen: Ibu, naproxen: Npx) in 20 ml of anhydrous DCM were reacted with DMAP (4.1 mmoi) in the presence of molecular sieve for 10 min at room temperature. EDCI (4.1 mmoi) was added portionwise over 10 min and the reaction mixture was stirred for 5 h at room temperature in the dark, filtered, and then concentrated under vacuum at room temperature. The products were purified using column chromatography and characterized as described above. international Application No. PCT/US09/39956, filed on April 8, 2009, which is incorporated by reference in its entirety as though fully set forth, provides additional examples of synthesizing bifunctional derivatives of α-lipoic acid and NSAIDs,

Example 10

Synthesis of dimeric derivatives of NSAIDs

NSAIDs (6 mmoi) and TEG (2.5 mmoi) in 40 ml of anhydrous DCM were reacted with DMAP (6 mmoi) in the presence of molecular sieve for 10 min at room temperature. EDC! (6 mmoi) was added portionwise over 10 min and the reaction mixture was stirred for 5 h at room tem t i the dark, filtered, and then concentrated under vacuum The products were purified (column chromatography, 100 0 5 CH3CI MeOH) and characterized as described above

International Application No PCT/US09/39956 filed on April 8, 2009, which is incorporated by reference in its entirety as though fully set forth, provides additional examples of synthesizing dimeric derivatives of NSAIDs

Various embodiments of the invention are described above in the Detailed Description While these descriptions directly describe the above embodiments, it is understood that those skilled in the art may conceive modifications and/or variations to the specific embodiments shown and described herein Any such modifications or variations that fall within the purview of this description are intended to be included therein as well. Unless specifically noted, it is the intention of the inventors that the words and phrases in the specification and claims be given the ordinary and accustomed meanings to those of ordinary skill in the applicable art(s) The foregoing description of various embodiments of the invention known to the applicant at this time of filing the application has been presented and is intended for the purposes of illustration and description The present description is not intended to be exhaustive nor limit the invention to the precise form disclosed and many modifications and variations are possible in the light of the above teachings The embodiments described serve to explain the principles of the invention and its practical application and to enable others skilled in the art to utilize the invention in various embodiments and with various modifications as are suited to the particular use contemplated Therefore, it is intended that the invention not be limited to the particular embodiments disclosed for carrying out the invention While particular embodiments of the present invention have been shown and described, it will be obvious to those skilled in the art that, based upon the teachings herein, changes and modifications may be made without departing from this invention and its broader aspects and, therefore the appended claims are to encompass within their scope all such changes and modifications as are within the true spirit and scope of this invention It will be understood by those within the art that, in general, terms used herein are generally intended as "open" terms (e g the term "including" should be interpreted as "including but not limited to,' the term 'having" should be interpreted as 'having at least," the term 'includes" should be interpreted as ' includes but is not limited to," etc )