Language selection

Search

Patent 2556759 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2556759
(54) English Title: BETA-LAPACHONE AND S-PHASE DRUG COMBINATIONS FOR CANCER TREATMENT
(54) French Title: COMBINAISONS DE BETA-LAPACHONE ET DE MEDICAMENT EN PHASE S POUR LE TRAITEMENT DU CANCER
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 31/352 (2006.01)
  • A61K 31/7064 (2006.01)
  • A61P 35/00 (2006.01)
  • A61P 35/02 (2006.01)
  • A61P 43/00 (2006.01)
(72) Inventors :
  • LI, CHIANG J. (United States of America)
(73) Owners :
  • ARQULE, INC. (United States of America)
(71) Applicants :
  • ARQULE, INC. (United States of America)
(74) Agent: OSLER, HOSKIN & HARCOURT LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2005-02-23
(87) Open to Public Inspection: 2005-09-09
Examination requested: 2006-08-17
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2005/005620
(87) International Publication Number: WO2005/082354
(85) National Entry: 2006-08-17

(30) Application Priority Data:
Application No. Country/Territory Date
60/547,287 United States of America 2004-02-23
11/068,459 United States of America 2005-02-18
11/060,747 United States of America 2005-02-18

Abstracts

English Abstract




Cancers and /or malignancies can be treated by administration of a G1/S phase
drug, which is preferably .beta.-lapachone, or a pharmaceutically acceptable
salt, prodrug, metabolite, analog or derivative thereof, combined with an S
phase drug, which is advantageously gemcitabine. This combination of a G1/S
phase drug with an S phase drug results in an unexpectedly effective treatment
of cancer. The invention includes methods of treating cancers by administering
a combination of a G1/S phase drug and an S phase drug, pharmaceutical
compositions comprising the combination of drugs used in these methods, as
well as pharmaceutical kits.


French Abstract

Des cancers et/ou des tumeurs malignes peuvent être traités par administration d'un médicament en phase G1/S, de préférence un .beta.-lapachone, ou un sel pharmaceutiquement acceptable, un promédicament, un métabolite, un analogue ou un dérivé de celui-ci, combiné avec un médicament en phase S, avantageusement de la gemcitabine. Cette combinaison d'un médicament en phase G1/S avec un médicament en phase S permet d'obtenir un traitement incroyablement efficace du cancer. Cette invention porte sur des procédés de traitement de cancers par administration d'une combinaison d'un médicament en phase G1/S et d'un médicament en phase S, sur des compositions pharmaceutiques contenant la combinaison de médicaments utilisés dans ces procédés, ainsi que sur des kits pharmaceutiques.

Claims

Note: Claims are shown in the official language in which they were submitted.



What is claimed:

1. A method of treating cancer, comprising administering to a subject in need
thereof a
therapeutically effective amount of
a) .beta.-lapachone, or a pharmaceutically acceptable salt thereof, in
combination with a
pharmaceutically acceptable carrier, and
b) an S phase drug, or a pharmaceutically acceptable salt thereof, in
combination with a
pharmaceutically acceptable carrier,
wherein said cancer is treated.

2. The method of claim 1, wherein said S phase drug is an antimetabolite drug.

3. The method of claim 1, wherein said S phase drug is a nucleoside analog or
nucleotide
analog.

4. The method of claim 1, wherein said S phase drug is selected from the group
consisting
of gemcitabine, 5-fluorouracil, methotrexate, hydroxyurea, cladribine,
fludarabine, cytarabine,
azacitidine, 5-fluorodeoxyuridine, mercaptopurine, azathioprine, thioguanine
and capacitabine.

5. The method of claim 1, wherein said S phase drug is gemcitabine.

6. The method of claim 1, wherein said S phase drug is 5-fluorouracil.

7. The method of claim 1, wherein said S phase drug is methotrexate.

8. The method of claim 1, wherein said cancer is selected from the group
consisting of
pancreatic cancer, lung cancer, colon cancer, breast cancer, prostate cancer,
chronic
myelogenous leukemia, melanoma, and ovarian cancer.

9. The method of claim 1, wherein said cancer is pancreatic cancer.

10. The method of claim 1, wherein said cancer is lung cancer.

11. The method of claim 1, wherein said .beta.-lapachone, or a
pharmaceutically acceptable salt,
prodrug, metabolite, analog or derivative thereof, is administered
simultaneously with
administration of said S phase drug, or a pharmaceutically acceptable salt
thereof.

12. The method of claim 1, wherein said .beta.-lapachone, or a
pharmaceutically acceptable salt,
prodrug, metabolite, analog or derivative thereof, is administered prior to
administration of said
S phase drug, or a pharmaceutically acceptable salt thereof.

64



13. The method of claim 1, wherein said .beta.-lapachone, or a
pharmaceutically acceptable salt,
prodrug, metabolite, analog or derivative thereof, is administered following
administration of
said S phase drug, or a pharmaceutically acceptable salt thereof.

14. A method of treating pancreatic cancer, comprising administering to a
subject in need
thereof a therapeutically effective amount of
a) .beta.-lapachone, or a pharmaceutically acceptable salt thereof, in
combination with a
pharmaceutically acceptable carrier, and
b) an S phase drug, or a pharmaceutically acceptable salt thereof, in
combination with a
pharmaceutically acceptable carrier,
wherein said pancreatic cancer is treated.

15. The method of claim 14, wherein said S phase drug is an antimetabolite
drug.

16. The method of claim 14, wherein said S phase drug is a nucleoside analog
or nucleotide
analog.

17. The method of claim 14, wherein said S phase drug is selected from the
group consisting
of gemcitabine, 5-fluorouracil, methotrexate, hydroxyurea, cladribine,
fludarabine, cytarabine,
azacitidine, 5-fluorodeoxyuridine, mercaptopurine, azathioprine, thioguanine
and capacitabine.

18. The method of claim 14, wherein said S phase drug is gemcitabine.

19. The method of claim 14, wherein said S phase drug is 5-fluorouracil.

20. The method of claim 14, wherein said S phase drug is methotrexate.

21. The method of claim 14, wherein said .beta.-lapachone, or a
pharmaceutically acceptable
salt, prodrug, metabolite, analog or derivative thereof, is administered
simultaneously with
administration of said S phase drug, or a pharmaceutically acceptable salt
thereof.

22. The method of claim 14, wherein said .beta.-lapachone, or a
pharmaceutically acceptable
salt, prodrug, metabolite, analog or derivative thereof, is administered prior
to administration of
said S phase drug, or a pharmaceutically acceptable salt thereof.

23. The method of claim 14, wherein said .beta.-lapachone, or a
pharmaceutically acceptable
salt, prodrug, metabolite, analog or derivative thereof, is administered
following administration
of said S phase drug, or a pharmaceutically acceptable salt thereof.

24. A method of treating lung cancer, comprising administering to a subject in
need thereof a
therapeutically effective amount of




a) .beta.-lapachone, or a pharmaceutically acceptable salt thereof, in
combination with a
pharmaceutically acceptable carrier, and
b) an S phase drug, or a pharmaceutically acceptable salt thereof, in
combination with a
pharmaceutically acceptable carrier,
wherein said lung cancer is treated.

25. The method of claim 24, wherein said S phase drug is an antimetabolite
drug.

26. The method of claim 24, wherein said S phase drug is a nucleoside analog
or nucleotide
analog.

27. The method of claim 24, wherein said S phase drug is selected from the
group consisting
of gemcitabine, 5-fluorouracil, methotrexate, hydroxyurea, cladribine,
fludarabine, cytarabine,
azacitidine, 5-fluorodeoxyuridine, mercaptopurine, azathioprine, thioguanine
and capacitabine.

28. The method of claim 24, wherein said S phase drug is gemcitabine.

29. The method of claim 24, wherein said S phase drug is 5-fluorouracil.

30. The method of claim 24, wherein said S phase drug is methotrexate.

31. The method of claim 24, wherein said .beta.-lapachone, or a
pharmaceutically acceptable
salt, prodrug, metabolite, analog or derivative thereof, is administered
simultaneously with
administration of said S phase drug, or a pharmaceutically acceptable salt
thereof.

32. The method of claim 24, wherein said .beta.-lapachone, or a
pharmaceutically acceptable
salt, prodrug, metabolite, analog or derivative thereof, is administered prior
to administration of
said S phase drug, or a pharmaceutically acceptable salt thereof.

33. The method of claim 24, wherein said .beta.-lapachone, or a
pharmaceutically acceptable
salt, prodrug, metabolite, analog or derivative thereof, is administered
following administration
of said S phase drug, or a pharmaceutically acceptable salt thereof.

34. A method of treating cancer, comprising administering to a subject in need
thereof a
therapeutically effective amount of
a) .beta.-lapachone, or a pharmaceutically acceptable salt thereof, in
combination with a
pharmaceutically acceptable carrier, and
b) gemcitabine, or a pharmaceutically acceptable salt thereof, in combination
with a
pharmaceutically acceptable carrier,
wherein said cancer is treated.

66



35. The method of claim 34, wherein said cancer is selected from the group
consisting of
pancreatic cancer, lung cancer, colon cancer, breast cancer, prostate cancer,
chronic
myelogenous leukemia, melanoma, and ovarian cancer.

36. The method of claim 34, wherein said cancer is pancreatic cancer.

37. The method of claim 34, wherein said cancer is lung cancer.

38. The method of claim 34, wherein said .beta.-lapachone, or a
pharmaceutically acceptable salt
thereof, is administered simultaneously with administration of said
gemcitabine.

39. The method of claim 34, wherein said .beta.-lapachone, or a
pharmaceutically acceptable salt
thereof, is administered prior to administration of said gemcitabine.

40. A method for inducing cell death in a cancer cell, comprising contacting
said cancer cell
with an effective amount of
a) .beta.-lapachone, or a pharmaceutically acceptable salt thereof, and
b) an S phase drug, or a pharmaceutically acceptable salt thereof,
wherein said contacting induces said cell death in said cancer cell.

41. The method of claim 40, wherein said S phase drug is an antimetabolite
drug.

42. The method of claim 40, wherein said S phase drug is a nucleoside analog
or nucleotide
analog.

43. The method of claim 40, wherein said S phase drug is selected from the
group consisting
of gemcitabine, 5-fluorouracil, methotrexate, hydroxyurea, cladribine,
fludarabine, cytarabine,
azacitidine, 5-fluorodeoxyuridine, mercaptopurine, azathioprine, thioguanine
and capacitabine.

44. The method of claim 40, wherein said S phase drug is gemcitabine.

45. The method of claim 40, wherein said S phase drug is 5-fluorouracil.

46. The method of claim 40, wherein said S phase drug is methotrexate.

47. The method of claim 40, wherein said S phase drug, or a pharmaceutically
acceptable
salt thereof, is combined with a pharmaceutically acceptable carrier.

48. The method of claim 40, wherein said .beta.-lapachone, or a
pharmaceutically acceptable salt
thereof, is combined with a pharmaceutically acceptable carrier.

49. The method of claim 40, wherein said cancer cell is a pancreatic cancer
cell, lung cancer
cell, colon cancer cell, breast cancer cell, prostate cancer cell, chronic
myelogenous leukemia
cell, melanoma cell, or ovarian cancer cell.

67



50. The method of claim 40, wherein said cancer cell is a pancreatic cancer
cell.

51. The method of claim 40, wherein said cancer cell is a lung cancer cell.

52. A pharmaceutical composition comprising a therapeutically effective amount
of .beta.-
lapachone, or a pharmaceutically acceptable salt thereof, a therapeutically
effective amount of
gemcitabine, and a pharmaceutically acceptable carrier.

53. A pharmaceutical composition comprising a therapeutically effective amount
of .beta.-
lapachone, a therapeutically effective amount of gemcitabine, and a
pharmaceutically acceptable
carrier.

54. A kit comprising
a) a first container comprising a therapeutically effective amount of .beta.-
lapachone, or a
pharmaceutically acceptable salt thereof,
b) a second container comprising a therapeutically effective amount of an S
phase drug, or a
pharmaceutically acceptable salt thereof, and
c) instructions for using said .beta.-lapachone, or a pharmaceutically
acceptable salt thereof, and
said S phase drug, or a pharmaceutically acceptable salt thereof, to treat a
subject.

55. The kit of claim 54, further comprising one or more additional doses of a
therapeutically
effective amount of .beta.-lapachone, or a pharmaceutically acceptable salt
thereof.

56. The kit of claim 54, further comprising one or more additional doses of a
therapeutically
effective amount of an S phase drug, or a pharmaceutically acceptable salt
thereof.

57. The kit of claim 54, wherein said .beta.-lapachone, or a pharmaceutically
acceptable salt
thereof, is administered orally.

58. The kit of claim 54, wherein said .beta.-lapachone, or a pharmaceutically
acceptable salt
thereof, is administered intravenously.

59. The kit of claim 54, wherein said .beta.-lapachone, or a pharmaceutically
acceptable salt
thereof, is administered by injection.

60. The kit of claim 54, wherein said S phase drug, or a pharmaceutically
acceptable salt
thereof, is administered orally.

61. The kit of claim 54, wherein said S phase drug, or a pharmaceutically
acceptable salt
thereof, is administered intravenously.

68



62. The kit of claim 54, wherein said S phase drug, or a pharmaceutically
acceptable salt
thereof, is administered by injection.

63. The kit of claim 54, wherein said .beta.-lapachone, or a pharmaceutically
acceptable salt
thereof, is administered simultaneously with administration of said S phase
drug, or a
pharmaceutically acceptable salt thereof.

64. The kit of claim 54, wherein said .beta.-lapachone, or a pharmaceutically
acceptable salt
thereof, is administered prior to administration of said S phase drug, or a
pharmaceutically
acceptable salt thereof.

65. The kit of claim 54, wherein said .beta.-lapachone, or a pharmaceutically
acceptable salt
thereof, is administered following administration of said S phase drug, or a
pharmaceutically
acceptable salt thereof.

66. A kit comprising
a) a first container comprising a therapeutically effective of .beta.-
lapachone, or a pharmaceutically
acceptable salt thereof,
b) a second container comprising a therapeutically effective amount of
gemcitabine, and
c) instructions for using said .beta.-lapachone, or a pharmaceutically
acceptable salt thereof, and
said gemcitabine, to treat a subject.

67. The kit of claim 66, further comprising one or more additional doses of a
therapeutically
effective amount of .beta.-lapachone, or a pharmaceutically acceptable salt
thereof.

68. The kit of claim 66, further comprising one or more additional doses of a
therapeutically
effective amount of gemcitabine, or a pharmaceutically acceptable salt
thereof.

69. The kit of claim 66, wherein said .beta.-lapachone, or a pharmaceutically
acceptable salt
thereof, is administered orally.

70. The kit of claim 66, wherein said .beta.-lapachone, or a pharmaceutically
acceptable salt
thereof, is administered intravenously.

71. The kit of claim 66, wherein said .beta.-lapachone, or a pharmaceutically
acceptable salt
thereof, is administered by injection.

72. The kit of claim 66, wherein said gemcitabine is administered
intravenously.

73. The kit of claim 66, wherein said gemcitabine is administered by
injection.

69



74. The kit of claim 66, wherein said .beta.-lapachone, or a pharmaceutically
acceptable salt
thereof, is administered simultaneously with administration of said
gemcitabine.

75. The kit of claim 66, wherein said .beta.-lapachone, or a pharmaceutically
acceptable salt
thereof, is administered prior to administration of said gemcitabine.

76. The kit of claim 66, wherein said .beta.-lapachone, or a pharmaceutically
acceptable salt
thereof, is administered following administration of said gemcitabine.

77. A kit comprising
a) a container comprising a pharmaceutical composition comprising a
therapeutically effective
amount of .beta.-lapachone, or a pharmaceutically acceptable salt thereof, a
therapeutically effective
amount of gemcitabine, and a pharmaceutically acceptable carrier, and
b) instructions for using said pharmaceutical composition to treat a subject.

78. The kit of claim 77, further comprising one or more additional doses of
said
pharmaceutical composition.

79. The kit of claim 77, wherein said pharmaceutical composition is
administered
intravenously.

80. The kit of claim 77, wherein said pharmaceutical composition is
administered by
injection.



Description

Note: Descriptions are shown in the official language in which they were submitted.



CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
BETA-LAPACHONE AND S-PHASE DRUG COMBINATIONS
FOR CANCER TREATMENT
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims the benefit under 35 U.S.C. ~ 119(e) of U.S.
Application No.
60/547,287, filed February 23, 2004.
BACKGROUND OF THE INVENTION
Cancer is the second leading cause of death in the United States, exceeded
only by heart
disease. Cancer Facts and Figures 2004, American Cancer Society, Inc. Despite
recent
advances in cancer diagnosis and treatment, surgery and radiotherapy may be
curative if a
cancer is found early, but current drug therapies for metastatic disease are
mostly palliative and
seldom offer a long-term cure. Even with new chemotherapies entering the
market, the need
continues for new drugs effective in monotherapy or in combination with
existing agents as first
line therapy, and as second and third line therapies in treatment of resistant
tumors.
Cancer cells are by definition heterogeneous. For example, within a single
tissue or cell
type, multiple mutational 'mechanisms' may lead to the development of cancer.
As such,
heterogeneity frequently exists between cancer cells taken from tumors of the
same tissue and
same type that have originated in different individuals. Frequently-observed
mutational
'mechanisms' associated with some cancers may differ between one tissue type
and another (e.g.,
frequently-observed mutational 'mechanisms' leading to colon cancer may differ
from
frequently-observed 'mechanisms' leading to leukemias). It is therefore often
difficult to predict
whether a particular cancer will respond to a particular chemotherapeutic
agent. (Cancer
Medicine, 5th Edition, Bast et al. eds., B.C. Decker Inc., Hamilton, Ontario).
Multiple checkpoints are built into the machinery of the cell proliferation
cycle where
cells make a commitment to repair DNA damage or to undergo cell death. Many
cancer cells
have lost checkpoint control and have an uncontrolled proliferation drive.
Major checkpoints
occur at G1/S phase and at the G2/M phase transitions where cells make a
commitment to repair
DNA or undergo cell death (e.g., apoptosis). Cells may undergo cell death
(e.g., apoptosis)
when DNA damage is irreparable (Li, CJ et al. (1999) Pf-oc. Natl. Acad. Sci.
USA 96:13369-
13374).
Identification of therapeutic agents modulating the checkpoint control may
improve
cancer treatment. Indeed, recent reports suggest that activation of cell cycle
checkpoints may


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
represent an important new paradigm in the treatment of cancer (see, e.g., Y.
Li et al., Proc.
Nat!. Acad. Sci. USA (2003), 100(5), 2674-8).
The cell cycle checkpoint activator (3-lapachone, which acts at the G1/S phase
transition,
is an agent with a reported anti-cancer activity in a limited number of
cancers. For example,
there is reported a method and composition for the treatment of tumors, which
comprises the
administration of an effective amount of (3-lapachone, in combination with a
taxane derivative
(W000/61142). Additionally, U.S. Pat. No. 6,245,807 discloses the use of (3-
lapachone,
amongst other (3-lapachone derivatives, for use in treatment of human prostate
disease. As a
single agent, (3-lapachone has also been reported to decrease the number of
tumors, reduce tumor
size, or increase survival time, ox a combination of these in xenotransplant
mouse models of
human ovarian cancer (Li, C.J. et al., (1999) Proc. Nat!. Acad. Sci. USA,
96(23): 13369-13374),
human prostate cancer (Li, C.J. et al., (1999) Proc. Nat!. Acad. Sci. USA,
96(23): 13369-13374),
human breast cancer (Li, C.J. et al., (2000) AACR Proc., p. 9), and human
multiple myeloma
(WO 03!011224). In addition, it has been reported that j3-lapachone induces
necrosis in human
breast cancer cells, and apoptosis in ovary, colon, and pancreatic cancer
cells through induction
of caspase (Li, YZ et al., (1999) Molecular Medicine 5:232-239).
It has also been reported that J3-lapachone, when combined with Taxol~
(paclitaxel;
Bristol-Myers Squibb Co., N.Y., N.Y.) at moderate doses, has effective anti-
tumor activity in
human ovarian, prostate and breast cancer xenograft models in nude mice. No
signs of toxicity
to the mice were observed, and no weight loss was recorded during the
subsequent two months
following treatment during which the tumors did not reappear (See Li, CJ et
al. (1999) Proc.
Nat!. Acad. Sci. USA 96:13369-13374). Taxol is believed to act at the G2lM
phase transition of
the cell cycle.
It has now been discovered that the combination of a Gl/S phase drug, such as
(i-
lapachone, or a pharmaceutically acceptable salt, prodrug, metabolite, analog
or derivative
thereof, with an S-phase drug (such as gemcitabine, available from Eli Lilly
under the trade
name GEMZAR~) provides unexpectedly effective treatment for certain cancers,
such as
pancreatic cancer. GEMZAR~ (gemcitabine HCl) is a nucleoside analog that
exhibits antitumor
activity. Gemcitabine may be used in monotherapy, or in combination with other
agents (e.g.,
cisplatin, carboplatin, TAXOL~ (paclitaxel)), to treat various cancers,
including pancreatic
cancer, breast cancer, non-small cell lung cancer, ovarian cancer, and bladder
cancer.
Gemcitabine exhibits cell phase specificity, primarily killing cells
undergoing DNA synthesis
(S-phase) and also blocking the progression of cells through the Gl/S
boundary. Without being
limited by theory, it is believed that after a gemcitabine nucleotide is
incorporated into DNA,
2


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
only one additional nucleotide may be added to the growing DNA strands. Again
not limited by
theory, it is believed that DNA polymerase epsilon is unable to remove the
gemcitabine
nucleotide and repair the growing DNA strand (e.g., masked chain termination).
In CEM T
lymphoblastoid cells, gemcitabine induces internucleosomal DNA fragmentation,
one of the
characteristics of programmed cell death (e.g., apoptosis).
SUMMARY OF THE INVENTION
The present invention provides a method of treating cancer, comprising
administering to
a subject in need thereof a therapeutically effective amount of a) (3-
lapachone, or a
pharmaceutically acceptable salt thereof, in combination with a
pharmaceutically acceptable
carrier, and b) an S phase drug, or a pharmaceutically acceptable salt
thereof, in combination
with a pharmaceutically acceptable carrier, where the cancer is treated.
The present invention also provides a method of treating pancreatic cancer,
comprising
administering to a subject in need thereof a therapeutically effective amount
of a) [3-lapachone,
or a pharmaceutically acceptable salt thereof, in combination with a
pharmaceutically acceptable
Garner, and b) an S phase drug, or a pharmaceutically acceptable salt thereof,
in combination
with a pharmaceutically acceptable carrier, where the pancreatic cancer is
treated.
The present invention also provides a method of treating lung cancer,
comprising
administering to a subject in need thereof a therapeutically effective amount
of a) (3-lapachone,
or a pharmaceutically acceptable salt thereof, in combination with a
pharmaceutically acceptable
Garner, and b) an S phase drug, or a pharmaceutically acceptable salt thereof,
in combination
with a pharmaceutically acceptable carrier, where the lung cancer is treated.
The present invention also provides a method of treating cancer, comprising
administering to a subject in need thereof a therapeutically effective amount
of a) (3-lapachone,
or a pharmaceutically acceptable salt thereof, in combination with a
pharmaceutically acceptable
carrier, and b) gemcitabine, or a pharmaceutically acceptable salt thereof, in
combination with a
pharmaceutically acceptable Garner, where the cancer is treated.
The present invention also provides a method of treating pancreatic cancer,
comprising
administering to a subject in need thereof a therapeutically effective amount
of a) [3-lapachone,
or a pharmaceutically acceptable salt thereof, in combination with a
pharmaceutically acceptable
Garner, and b) gemcitabine, or a pharmaceutically acceptable salt thereof, in
combination with a
pharmaceutically acceptable carrier, where the pancreatic cancer is treated.


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
The present invention also provides a method of treating lung cancer,
comprising
administering to a subject in need thereof a therapeutically effective amount
of a) (3-lapachone,
or a pharmaceutically acceptable salt thereof, in combination with a
pharmaceutically acceptable
Garner, and b) gemcitabine, or a pharmaceutically acceptable salt thereof, in
combination with a
pharmaceutically acceptable carrier, where the lung cancer is treated.
The present invention also provides a method for inducing cell death in a
cancer cell,
comprising contacting the cancer cell with an effective amount of a) (3-
lapachone, or a
pharmaceutically acceptable salt thereof, and b) an S phase drug, or a
pharmaceutically
acceptable salt thereof, where the contacting induces the cell death in the
cancer cell.
The present invention also provides a method for inducing cell death in a
cancer cell,
comprising contacting the cancer cell with an effective amount of a) (3-
lapachone, or a
pharmaceutically acceptable salt thereof, and b) gemcitabine, or a
pharmaceutically acceptable
salt thereof, where the contacting induces the cell death in the cancer cell.
The present invention also provides a pharmaceutical composition comprising a
therapeutically effective amount of (3-lapachone, or a pharmaceutically
acceptable salt thereof, a
therapeutically effective amount of gemcitabine, and a pharmaceutically
acceptable Garner.
The present invention also provides a pharmaceutical composition comprising a
therapeutically effective amount of (3-lapachone, a therapeutically effective
amount of
gemcitabine, and a pharmaceutically acceptable carrier.
The present invention also provides a kit comprising a) a first container
comprising a
therapeutically effective amount of [3-lapachone, or a pharmaceutically
acceptable salt thereof,
b) a second container comprising a therapeutically effective amount of an S
phase drug, or a
pharmaceutically acceptable salt thereof, and c) instructions for using the (3-
lapachone, or a
pharmaceutically acceptable salt thereof, and the S phase drug, or a
pharmaceutically acceptable
salt thereof, to treat a subject.
The present invention also provides a kit comprising a) a first container
comprising a
therapeutically effective of (3-lapachone, or a pharmaceutically acceptable
salt thereof, b) a
second container comprising a therapeutically effective amount of gemcitabine,
and c)
instructions for using the (3-lapachone, or a pharmaceutically acceptable salt
thereof, and the
gemcitabine, to treat a subject.
The present invention also provides a kit comprising a) a container comprising
a
pharmaceutical composition comprising a therapeutically effective amount of (3-
lapachone, or a
pharmaceutically acceptable salt thereof, a therapeutically effective amount
of gemcitabine, and
4


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
a pharmaceutically acceptable earner, and b) instructions for using the
pharmaceutical
composition to treat a subject.
The present invention also provides a method of treating a cell proliferative
disorder,
comprising administering to a subject in need thereof a therapeutically
effective amount of a) (3-
lapachone, or a pharmaceutically acceptable salt, prodrug, metabolite, analog
or derivative
thereof, in combination with a pharmaceutically acceptable carrier, and b) an
S phase drug, or a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof, in
combination with a pharmaceutically acceptable carrier, where the cell
proliferative disorder is
treated.
The present invention also provides a method of treating a cell proliferative
disorder,
comprising administering to a subject in need thereof a therapeutically
effective amount of a) (3-
lapachone, or a pharmaceutically acceptable salt, prodrug, metabolite, analog
or derivative
thereof, in combination with a pharmaceutically acceptable carrier, and b)
gemcitabine, or a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof, in
combination with a pharmaceutically acceptable carrier, where the cell
proliferative disorder is
treated.
The present invention also provides a method of treating cancer, comprising
administering to a subject in need thereof a therapeutically effective amount
of a) ~3-lapachone,
or a pharmaceutically acceptable salt, prodrug, metabolite, analog or
derivative thereof, in
combination with a pharmaceutically acceptable carrier, and b) an S phase
drug, or a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof, in
combination with a pharmaceutically acceptable earner, where the cancer is
treated.
The present invention also provides a method of treating pancreatic cancer,
comprising
administering to a subject in need thereof a therapeutically effective amount
of a) (3-lapachone,
or a pharmaceutically acceptable salt, prodrug, metabolite, analog or
derivative thereof, in
combination with a pharmaceutically acceptable carrier, and b) an S phase
drug, or a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof, in
combination with a pharmaceutically acceptable carrier, where the pancreatic
cancer is treated.
The present invention also provides a method of treating lung cancer,
comprising
administering to a subject in need thereof a therapeutically effective amount
of a) (3-lapachone,
or a pharmaceutically acceptable salt, prodrug, metabolite, analog or
derivative thereof, in
combination with a pharmaceutically acceptable earner, and b) an S phase drug,
or a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof, in
combination with a pharmaceutically acceptable carrier, where the lung cancer
is treated.


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
The present invention also provides a method of treating cancer, comprising
administering to a subject in need thereof a therapeutically effective amount
of a) (3-lapachone,
or a pharmaceutically acceptable salt, prodrug, metabolite, analog or
derivative thereof, in
combination with a pharmaceutically acceptable Garner, and b) gemcitabine, or
a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof, in
combination with a pharmaceutically acceptable Garner, where the cancer is
treated.
The present invention also provides a method of treating pancreatic cancer,
comprising
administering to a subject in need thereof a therapeutically effective amount
of a) (3-lapachone,
or a pharmaceutically acceptable salt, prodrug, metabolite, analog or
derivative thereof, in
combination with a pharmaceutically acceptable Garner, and b) gemcitabine, or
a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof, in
combination with a pharmaceutically acceptable carrier, where the pancreatic
cancer is treated.
The present invention also provides a method of treating lung cancer,
comprising
administering to a subject in need thereof a therapeutically effective amount
of a) (3-lapachone,
or a pharmaceutically acceptable salt, prodrug, metabolite, analog or
derivative thereof, in
combination with a pharmaceutically acceptable carrier, and b) gemcitabine, or
a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof, in
combination with a pharmaceutically acceptable carrier, where the lung cancer
is treated.
The present invention also provides a method for inducing cell death in a
cancer cell,
comprising contacting the cancer cell with an effective amount of a) (3-
lapachone, or a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof, and b) an S
phase drug, or a pharmaceutically acceptable salt, prodrug, metabolite, analog
or derivative
thereof, where the contacting induces the cell death in the cancer cell.
The present invention also provides a method for inducing cell death in a
cancer cell,
comprising contacting the cancer cell with an effective amount of a) [3-
lapachone, or a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof, and b)
gemcitabine, where the contacting induces the cell death in the cancer cell.
The present invention also provides a pharmaceutical composition comprising a
therapeutically effective amount of (3-lapachone, or a pharmaceutically
acceptable salt, prodrug,
metabolite, analog or derivative thereof, a therapeutically effective amount
of gemcitabine, and a
pharmaceutically acceptable carrier.
The present invention also provides a kit comprising a) a first container
comprising a
therapeutically effective amount of (3-lapachone, or a pharmaceutically
acceptable salt, prodrug,
metabolite, analog or derivative thereof, b) a second container comprising a
therapeutically
6


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
effective amount of an S phase drug, or a pharmaceutically acceptable salt,
prodrug, metabolite,
analog or derivative thereof, and c) instructions for using the ~3-lapachone,
or a pharmaceutically
acceptable salt, prodrug, metabolite, analog or derivative thereof, and the S
phase drug, or a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof, to treat a
subj ect.
The present invention also provides a kit comprising a) a first container
comprising a
therapeutically effective of [3-lapachone, or a pharmaceutically acceptable
salt, prodrug,
metabolite, analog or derivative thereof, b) a second container comprising a
therapeutically
effective amount of gemcitabine, and c) instructions for using the [3-
lapachone, or a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof, and the
gemcitabine, to treat a subject.
The present invention also provides a kit comprising a) a container comprising
a
pharmaceutical composition comprising a therapeutically effective amount of /3-
lapachone, or a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof, a
therapeutically effective amount of gemcitabine, and a pharmaceutically
acceptable Garner, and
b) instructions for using the pharmaceutical composition to treat a subject.
The present invention also provides a method of treating individuals afflicted
with
cancerous or pre-cancerous cells, tumors and/or malignancies. This method
comprises
administering to a subject (e.g., a subject in need of such treatment, e.g.,
an individual afflicted
with cancer) an effective amount of a G1/S phase drug, such as (3-lapachone or
a derivative or
analog thereof, together with an effective amount of an S phase drug such as
gemcitabine, such
that the cancer is treated. The combination of the present invention results
in a surprising
efficacy which is beneficial in reducing tumor burden load and/or regressing
tumor growth,
especially in patients with metastatic disease. In an embodiment, the human
malignancy treated
is a cancer such as pancreatic cancer, although the invention is not limited
in this respect, and
other metastatic diseases may be treated by the combination of the present
invention.
The present invention also provides a method for treating cancer in a subject
by
administering to the subject a G1/S phase drug, which is preferably (3-
lapachone, or a derivative
or analog thereof, together with an S-phase drug, such as gemcitabine, in a
therapeutically
effective amount, under conditions such that the cancer is treated. In certain
embodiments, the
cancer is pancreatic cancer.
In another embodiment, the invention provides compositions useful for the
treatment of
cancer. In a preferred embodiment, the composition comprises a therapeutically
effective
amount of a combination of a G1/S phase drug such as (3-lapachone, or a
derivative or analog
7


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
thereof, together with an S-phase drug, such as gemcitabine. In preferred
embodiments, the
composition further includes a pharmaceutically-acceptable solvent, earner,
diluent, or
excipient.
In another embodiment, the invention provides kits for treating cancer. The
kits include
a GlIS phase drug such as [3-lapachone, or a derivative or analog thereof, and
an S-phase drug,
such as gemcitabine. The Gl/S phase drug and the S-phase drug are present in
amounts
effective in combination for treating cancer.
The combination of the present invention is particularly advantageous in the
treatment of
patients who have chemotherapeutically refractive metastatic cancers,
including pancreatic
cancer. The method of the present invention comprises administering to the
patient, an effective
amount, in combination, of a Gl/S phase drug (such as ~3-lapachone) and an S
phase drug.
Preferably, the combination is (1) a cell cycle checkpoint activator such as
(3-lapachone or
derivatives or analogs thereof (a G1 and/or S phase drug) and (2)
antimetabolite drugs such as
gemcitabine, 5-fluorouracil (5-FU), capacitabine, methotrexate, 5-
fluorodeoxyuridine, and
cytarabine (araC); and the like (S phase drug), and pharmaceutically
acceptable salts thereof. In
a preferred embodiment, the cell cycle checkpoint activator is [3-lapachone.
In preferred
embodiments, the antimetabolite drug is a pyrimidine antagonist such as
gemcitabine, 5-FU,
capacitabine or araC, and most preferably gemcitabine.
The above description sets forth rather broadly the more important features of
the present
invention in order that the detailed description thereof that follows may be
understood, and in
order that the present contributions to the art may be better appreciated.
Other objects and
features of the present invention will become apparent from the following
detailed description
considered in conjunction with the accompanying drawings. It is to be
understood, however,
that the drawings are designed solely for the purposes of illustration and not
as a definition of
the limits of the invention, for which reference should be made to the
appended claims.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 sets forth an effect of (3-lapachone, administered in monotherapy or
in
combination with gemcitabine, on the growth of xenografted Panc-1 human
pancreatic tumors in
an athymic nude mouse model.
Figure 2 sets forth an effect of (3-lapachone, administered in monotherapy or
in
combination with gemcitabine, on the growth of xenografted A549 human lung
tumors in an
athymic nude mouse model.


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides a method of treating a cell proliferative
disorder, such as
cancer, comprising administering to a subject in need thereof a
therapeutically effective amount
of a) (3-lapachone, or a pharmaceutically acceptable salt, prodrug,
metabolite, analog or
derivative thereof, in combination with a pharmaceutically acceptable Garner,
and b) an S phase
drug, or a pharmaceutically acceptable salt, prodrug, metabolite, analog or
derivative thereof
(e.g., gemcitabine), in combination with a pharmaceutically acceptable
carrier, where the cell
proliferative disorder is treated. The invention also provides the use of (3-
lapachone, or a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof, and an S
phase drug, or a pharmaceutically acceptable salt, prodrug, metabolite, analog
or derivative
thereof (e.g., gemcitabine) for the preparation of a medicament useful for the
treatment of
cancer. The invention also provides for kits comprising (3-lapachone, or a
pharmaceutically
acceptable salt, prodrug, metabolite, analog or derivative thereof and an S
phase drug, or a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof (e.g.,
gemcitabine).
While not limited by theory, the present invention includes and is based in
part on an
understanding of, and methods for, the activation of cell cycle checkpoints by
modulators of cell
cycle checkpoint activation (e.g., (3-lapachone, or a pharmaceutically
acceptable salt, prodrug,
metabolite, analog or derivative thereof). The activation of cell cycle
checkpoints in general is
referred to as Activated Checkpoint TherapyTM, or ACTTM. Briefly, many cancer
cells are
defective in their cell cycle checkpoint functions secondary to mutations in
one of their
molecular modulators, e.g., p53. It is in part, for this reason, that cancer
cells have accumulated
genetic errors during the carcinogenic process. Therapeutic agents that
activate cell cycle
checkpoint functions can selectively promote cell death in cancer cells, since
cell death appears
to be induced at least in part by the conflict between the uncontrolled-
proliferation drive in
cancer cells and the checkpoint delays induced artificially. ACTTM takes
advantage of the
tendency of cell death to occur at checkpoints during the cell proliferation
cycle by activating
one or more checkpoints, thereby producing conflicting signals regarding cell
cycle progression
versus arrest. If more than one checkpoint is activated, cancer cells with
uncontrolled
proliferation signals and genetic abnormalities are blocked at multiple
checkpoints, creating
"collisions" that promote synergistic cell death.
ACTTM offers selectivity against cancer cells as compared to normal cells and
is
therefore safer than less selective therapies. First, the ACTTM method
activates but does not
disrupt cell cycle checkpoints. Second, normal cells with well-controlled
proliferation signals
9


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
can be delayed at checkpoints in a regulated fashion, resulting in no cell
death-prone collisions.
Third, normal cells with intact G1 checkpoint control are expected to arrest
in G1. Cancer cells,
on the other hand, are expected to be delayed in S-, G2-, and M-phases, since
most cancer cells
harbor Gl checkpoint defects, making cancer cells more sensitive to drugs
imposing S and M
phase checkpoints. (3-lapachone is a G1 and S phase compound, and contacting a
cell with [3-
lapachone results in activation of a G1 or S cell cycle checkpoint.
In preferred embodiments, the combination of a G1/S phase drug (e.g., ~3-
lapachone, or a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof) with an S
phase drug results in synergistic treatment of cancer cells and/or tumors. The
combinations of
the present invention are particularly advantageous using (3-lapachone and
gemcitabine, where
synergistic results may be obtained. Without intending to be limited by
theoary, molecular
changes underlying cell cycle delay at checkpoints, for example Gl/S phase and
S phase, appear
to result in the synergistic induction of cell death (e.g., apoptosis) in
malignant cells. Preferably,
the G1/S phase compound (or compounds) is administered prior to, or
simultaneously with,
compounds that target a cell at the S phase checkpoint. More preferably, the
G1/S phase
compounds) is/are administered prior to the compounds that target a cell at
the S phase
checkpoint.
The term "modulator of cell cycle checkpoint activation," as used herein,
refers to a
compound capable of altering checkpoint activation in cells (in preferred
embodiments,
activating one or more cell cycle checkpoints), preferably by activating
checkpoint-mediated
DNA repair mechanisms, or by reinstating checkpoint activity that has been
lost due to a
malfunction or mutation in the cellular pathways that regulate cell cycle
activity. As is known in
the art, major cell cycle checkpoints occur at Gl/S phase and at the GZ/Mphase
transitions. In a
model, four major cell cycle checkpoints monitor the integrity of genetic
material. DNA
synthesis begins only past the restriction point (R point), where the cell
determines if preparation
during G1 has been satisfactory for cell cycle continuation. The second
checkpoint occurs
during replication initiation in S phase. The third and fourth checkpoints
take place in G2 phase
and M phase, respectively. Modulation of cell cycle checkpoint activation is
further discussed
in, e.g., C.J. Li et al. Ps°oc. Natl. Acad. Sci. USA (1999), 96(23),
13369-13374, and Y. Li et al.
Proc. Natl. Acad. Sci. USA (2003), 100(5), 2674-2678, and PCT Publication WO
00/61142
(Pardee et al.). Preferred modulators of cell cycle checkpoint activation for
use in the present
invention induce checkpoint activation (i.e., activate one or more cell cycle
checkpoints,
preferably at Gl/S phase), preferably without causing substantial DNA damage.
In addition,
certain preferred modulators of cell cycle checkpoint activation are capable
of increasing the


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
level or activity of E2F (more preferably E2F1) in a cell. Methods for
screening for modulators
of cell cycle checkpoint activation, including compounds capable of elevating
E2F activity or
levels in a cell, include those that are disclosed in PCT Patent Application
No. PCT/LTS03/22631
to Li et al. In certain embodiments, preferred modulators of cell cycle
checkpoint activation are
capable of increasing the level or activity of E2F in a cell by an amount
sufficient to cause cell
death (e.g., apoptosis) if the cell is a cancerous cell. More preferred
modulators of cell cycle
checkpoint activation are capable of raising the level or activity of E2F1 in
a cell by an amount
sufficient to cause cell death (e.g., apoptosis) if the cell is a cancerous
cell. In one aspect, a
modulator of cell cycle checkpoint activation is not (3-lapachone.
Again not limited by theory, cellular response to DNA damage is regulated by
the
ATM/ATR signal transduction pathway, in which ATM and ATR are protein kinases
of the
phosphatidyl-inositol-3 kinase family (PI3K). In response to DNA damage, ATM
and ATR
phosphorylate Chk2 and Chkl respectively, which in turn activate a variety of
substrates
involved in arresting cells at the GlIS phase of the cell cycle, as well as
inducing and activating
proteins involved in DNA repair. Chk2 has been shown to activate proteins
involved in DNA
repair including the tumor suppressor BRCA1, thereby enhancing DNA repair
capacity
following DNA damage. Chk2 has also been shown to stabilize p53 both by
directly
phosphorylating p53, and by inhibiting Mdm2, a ubiquitin ligase that targets
p53 for
degradation. Under such conditions, increased levels of p53 lead to Gl/S
arrest, DNA repair,
and apoptosis in cells with irreparable DNA damage. Again not limited by
theory, it is believed
that Chk2 is an important cell cycle regulator, which, depending on the
conditions, can induce
cell cycle arrest and DNA repair, or initiate cell death (e.g., apoptosis) if
DNA damage is too
severe. In certain embodiments, preferred modulators of cell cycle checkpoint
activation are
capable of increasing the level or activity of Chk2 in a cell by an amount
sufficient to cause cell
death (e.g., apoptosis) if the cell is a cancerous cell.
Again not limited by theory, E2F1 is one of related proteins in the E2F family
of nuclear
transcription factors, which family is critically important in regulation of
the cell cycle. E2F1 is
required for cellular proliferation by promoting passage through the Gl/S
checkpoint. During
proliferation of normal cells, transcriptionally active E2F1 is liberated from
an inactive E2FllRb
complex following phosphorylation of Rb. E2F1 levels rise, promoting
progression through Gl.
As the cell moves toward the end of S phase, E2F1 levels must decline for
progress to continue.
Sustained elevation of E2F1 at this point in the cell cycle causes activation
of the S phase
checkpoint, and subsequent cell death (e.g., by apoptosis). Thus, depending on
the phase of the
cell cycle and dynamics of E2F1 elevation, this regulatory protein may either
promote cellular
11


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
proliferation, induce cell cycle delay, DNA repair or cell death. During the
G1 phase of the cell
cycle, phosphorylation of Rb results in dissociation of Rb-E2F1 complexes,
liberating active
E2F1, which then stimulates entry into S phase by promoting transcription of
key cell cycle
effectors. During S-phase, E2F1 must be degraded for progress to continue. In
the presence of
DNA damage, however, E2F1 levels increase rather than decrease, causing cell
cycle delay,
DNA repair, and, if damage is severe, cell death. As used herein, "E2F" is the
E2F transcription
factor family (including but not limited to E2F-1, E2F-2, E2F-3).
As used herein, "a cell cycle checkpoint pathway" refers to a biochemical
pathway that is
involved in modulation of a cell cycle checkpoint. A cell cycle checkpoint
pathway may have
stimulatory or inhibitory effects, or both, on one or more functions
comprising a cell cycle
checkpoint. A cell cycle checkpoint pathway is comprised of at least two
compositions of
matter, preferably proteins, both of which contribute to modulation of a cell
cycle checkpoint. A
cell cycle checkpoint pathway may be activated through an activation of one or
more members
of the cell cycle checkpoint pathway. Preferably, a cell cycle checkpoint
pathway is a
biochemical signaling pathway.
As used herein, "cell cycle checkpoint regulator" refers to a composition of
matter that
can function, at least in part, in modulation of a cell cycle checkpoint. A
cell cycle checkpoint
regulator may have stimulatory or inhibitory effects, or both, on one or more
functions
comprising a cell cycle checkpoint. In one aspect, a cell cycle checkpoint
regulator is a protein.
In another aspect, a cell cycle checkpoint regulator is a not a protein. In
one aspect, a cell cycle
checkpoint regulator is selected from the group consisting of ATM, ATR, Chkl,
Chk2, E2F1,
BRCAl, Rb, p53, p21, Mdm2, Cdc2, Cdc25, and 14-4-3[sigma].
The term "cell cycle checkpoint activator," as used herein, refers to a
compound capable
of activating cell cycle checkpoints, e.g., by activating checkpoint-mediated
DNA repair
mechanisms, or by reinstating checkpoint activity that has been lost due to a
malfunction or
mutation in the cellular pathways that regulate cell cycle activity. As is
known in the art, major
cell cycle checkpoints occur at Gl/S phase and at the G2/M phase transitions.
Modulation of cell
cycle checkpoint activation is further discussed in, e.g., C.J. Li et al.
Proc. Natl. Acad. Sci. USA
(1999), 96(23), 13369-13374, and Y. Li et al. Proc. Natl. Acad. Sci. USA
(2003), 100(5),
2674-2678, and PCT Publication WO 00/61142 (Pardee et al.). Preferred cell
cycle checkpoint
activators for use in the present invention induce checkpoint activation
(i.e., activate one or more
cell cycle checkpoints, preferably at Gl/S phase), preferably without causing
substantial DNA
damage. In addition, certain preferred cell cycle checlcpoint activators are
capable of increasing
the level or activity of E2F (more preferably E2F1) in a cell. Methods for
screening for cell
12


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
cycle checkpoint activators, including compounds capable of elevating E2F
activity or levels in
a cell, include those that are disclosed in PCT Patent Application No.
PCT/LJS03/22631 to Li et
al. In certain embodiments, preferred cell cycle checkpoint activators are
capable of increasing
the level of E2F in a cell by an amount sufficient to cause cell death (e.g.,
apoptosis) if the cell is
a cancer cell. More preferred cell cycle checkpoint activators are capable of
raising the level or
activity of E2F1 in a cell by an amount sufficient to cause cell death (e.g.,
apoptosis) if the cell
is a cancer cell. In one aspect, a cell cycle checkpoint activator is not [i-
lapachone.
I. Compositions
As used herein, the phrase "(3-lapachone" refers to 3,4-dihydro-2,2-dimethyl-
2H-
naphtho[1,2-b]pyran-5,6-dione and derivatives and analogs thereof, and has the
chemical
structure:
Formula I
Preferred derivatives and analogs are discussed below.
As used herein, a Gl/S-phase drug refers to a drug that modulates the cell
cycle (e.g.,
causes cell death) at the G1 and S-phase checkpoints, e.g., by activating a
cell cycle checkpoint.
Beta-lapachone is a preferred Gl/S-phase drug. Similarly, an S phase drug is a
drug that
modulates the cell cycle (e.g., causes cell death) at the S-phase checkpoint.
In one embodiment,
an S phase drug exerts a therapeutic effect by inhibiting DNA synthesis. In
one embodiment,
the invention is directed to a method for treating a subject having malignant
cells or inhibiting
further growth of such malignant cells by administering a drug or compound
that targets such
cells at the Gl/S phase checkpoints in the cell cycle. A second drug or
compound that acts at
the S phase checkpoint in the cell cycle is administered simultaneously with,
or following, the
Gl/S phase drug or compound. Individual compounds satisfying these criteria
are known to
those of ordinary skill in the art, and may be found in reference texts such
as the Physician's
Desk Reference, 59th Edition, Thomson PDR (2005). For example, (3-lapachone
and its
derivatives are G1/S phase drugs, while gemcitabine and other antimetabolite
drugs are S phase
drugs. A list of representative compounds is set forth below in Table 1.
13


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
TABLE 1
Twe Cateeory Compound Name Chemical Formula
1. G1/ S phase drug (i-lapachone 3,4-dihydro-2,2-dimethyl-2H-naphtho[1,2-
b]pyran-
5,6-dione
Reduced (3-lapachone
Analogs and
derivatives of (3-
lapachone
2. S phase drugs Gemcitabine 2',2'difluorodeoxycytidine; 2'-deoxy-2',2'-
difluorocytidine monohydrochloride ((3-isomer)
5-FU 5-fluorouracil
MTX Methotrexate; N-[4[[(2,4-Diamino-6-
pteridinyl)methyl]methylamino]benzoyl]-L-glutamic
acid
Cytarabine
5-fluorodeoxyuridine
Capacitabine
Hydroxyurea
Cladribine
Fludaribine
Azacytadine
Mercaptopurine
Azathioprine
Thioguanine
As used herein, an "antimetabolite drug" is any compound that exerts a
therapeutic effect
by inhibiting the utilization of an endogenous cellular metabolite. Exemplary
antimetabolite
drugs include folic acid antagonists (e.g., methotrextate), purine antagonists
(e.g., thioguanine)
and pyrimidine antagonists (e.g., fluorouracil). Exemplary antimetabolite
drugs are known to
those of ordinary skill in the art, and may be found in reference texts such
as the Physician 's
Desk Reference, 59th Edition, Thomson PDR (2005). In one aspect, an
antimetabolite drug is an
S phase antimetabolite drug that modulates the cell cycle, e.g., by causing
cell death, at the S-
phase checkpoint.
As used herein, a "nucleoside analog" is any compound that that exerts a
therapeutic
effect by acting as an analog to an endogenous cellular nucleoside, including
analogs of
deoxyribonucleosides and ribonucleosides, such as but not limited to analogs
of
deoxyadenosine, deoxyguanosine, deoxycytidine, deoxyuridine, thymidine,
adenosine,
guanosine, cytidine, uridine, and thymine ribosides. For example, Gemcitabine
HCl
(GEMZAR~) is 2'-deoxy-2',2'-difluorocytidine monohydrochloride ((3-isomer), a
nucleoside
analog that exhibits antitumor activity. As used herein, a "nucleotide analog"
is any compound
that acts as an analog to an endogenous cellular nucleotide, including analogs
of
14


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
deoxyribonucleotides and ribonucleotides, such as but not limited to analogs
of deoxyadenylic
acid, deoxyguanylic acid, deoxycytidylic acid, thymidylic acid, 2'-adenylic
acid, 3'-adenylic
acid, 5'-adenylic acid, 3'-guanylic acid, 3'-cytidylic acid, 3'-uridylic acid,
and phosphate-
bearing derivatives thereof. Nucleoside analogs and nucleotide analogs may be
useful as
chemotherapeutic agents for cell proliferative disorders such as cancer.
Without intending to be
limited by theory, nucleoside analogs and nucleotide analogs are believed to
inhibit DNA
synthesis by inhibition of DNA polymerases and prevention of elongation of DNA
strands
through direct incorporation into the DNA molecule. High levels of nucleoside
analog drugs
may lead to DNA strand breaks, inhibition of DNA synthesis, accumulation of
cells in S phase
or at the G1/S junction, or cell death. Exemplary nucleoside analogs and
nucleotide analogs are
known to those of ordinary skill in the art, and may be found in reference
texts such as the
Plzysician's DerkRefey~ehce, 59th Edition, Thomson PDR (2005).
Preferred Gl/S phase checkpoint targeting compounds include Gl/S phase drugs
(for
example, (3-lapachone), G1 phase drugs (for example, lovastatin, mimosine,
tamoxifen, and the
like) and S phase drugs (for example, gemcitabine, 5-FU, MTX, and the like).
Preferred G1/S
phase drugs for use in this invention preferably do not cause significant DNA
damage in normal
cells. (3-lapachone, its derivatives and analogs are most preferred Gl/S phase
drugs. Preferred
Gl/S phase compounds are direct activators of cell cycle checkpoint activation
(see, e.g., Y. Li
et al., P~oc. Natl. Acad. Sci. USA (2003), 100(5), 2674-8). Preferred G1/S
phase compounds
are capable of elevating the amount or activity of E2F (preferably E2F1) in a
cancer cell, thereby
activating the checkpoint and causing cell death in cancerous or pre-cancerous
cells.
(3-Lapachone (3,4-dihydro-2, 2-dimethyl-2H-naphtho [1,2-b] pyran-5, 6-dione),
a simple
non-water soluble orthonapthoquinone, was first isolated in 1882 by Paterno
from the heartwood
of the lapacho tree (See Hooker, SC, (1936) I. Am. Chem. Soc. 58:1181-1190;
Goncalves de
Lima, O, et al., (1962) Rev. Inst. Antibiot. Univ. Recife. 4:3-17). The
structure of (3-Lapachone
was established by Hooker in 1896 and it was first synthesized by Fieser in
1927 (Hooker, SC,
(1936) I. Am. Chem. Soc. 58:1181-1190). a-Lapachone can, for example, be
obtained by simple
sulfuric acid treatment of the naturally occurnng lapachol, which is readily
isolated from
Tabebuia avelleraedae growing mainly in Brazil, or is easily synthesized from
seeds of lomatia
growing in Australia (Li, CJ, et al., (1993) J. Biol. Claem. 268:22463-33464).
Methods for
formulating (3-Lapachone or its derivatives or analogs can be accomplished as
described in U.S.
Patent No. 6,458,974 and U.S. Publication No. US-2003-0091639-Al.
As used herein, derivatives or analogs of (3-Lapachone include, for example,
3,4-dihydro-
2,2-dimethyl-3-(3-methyl-2-butenyl)-2H-naphtho[1,2-b]pyran-5,6-dione, 3,4-
dihydro-2,2-


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
dimethyl-2H-naphtho[1,2-b]thiopyran-5,6-dione and 3,4-dihydro-4,4-dimethyl-2H-
naphtho[1,2-
b]thiopyran-5,6-dione. Other derivatives or analogs of (3-lapachone are
described in PCT
International Application PCT/US93/07878 (W094/04145), and U.S. Pat. No.
6,245,807. PCT
International Application PCT/US00/10169 (WO 00/61142), discloses (3-
lapachone, which may
have a variety of substituents at the 3- position as well as in place of the
methyl groups attached
at the 2-position. U.5. Patent Nos. 5,763,625, 5,824,700, and 5,969,163,
disclose analogs and
derivatives with a variety of substituents at the 2-, 3- and 4-positions.
Furthermore, a number of
journals report (3-lapachone analogs and derivatives with substituents at one
or more of the
following positions: 2-, 3-, 8- and/or 9-positions, (See, Sabba et al., (1984)
JMed Chem 27:990-
994 (substituents at the 2-, 8- and 9- positions); (Portela and Stoppani,
(1996) Biochern Pharm
51:275-283 (substituents at the 2- and 9- positions); Goncalves et al., (1998)
Molecular and
Biochemical Parasitology 1:167-176 (substituents at the 2- and 3- positions)).
Other derivatives
or analogs of (3-lapachone have sulfur-containing hetero-rings in the "a" and
"(3" positions of
lapachone (Kurokawa S, (1970) Bulletifz of The Chemical Society of.Iapan
43:1454-1459;
Tapia, RA et al., (2000) Heterocycles 53(3):585-598; Tapia, RA et al., (1997)
Tetrahedron
Letters 38(1):153-154; Chuang, CP et al., (1996) Heterocycles 40(10):2215-
2221; Suginome H
et al., (1993) Journal of t7ze Chemical Society, Chemical Communications 9:807-
809; Tonholo J
et al., (1988) .Iourraal of the Brazilian Chemical Society 9(2):163-169; and
Krapcho AP et al.,
(1990) .Iournal ofMedicinal Chemistry 33(9):2651-2655).
Further, G1/S phase checkpoint targeting drugs include reduced [3-lapachone
(e.g.,
Formula la, in which R' and R" are both hydrogen) and derivatives of reduced
(3-lapachone
(see,e.g., Formula la, in which R' and R" are each independently hydrogen, C1-
C6 alkyl, C1-C6
alkylcarbonyl, or a pharmaceutically acceptable salt).
Formula Ia
As used herein, the term "combination of the present invention" means a Gl/S-
phase
drug of the present invention and an S phase drug of the present invention. As
used herein, the
term "compound of the present invention" means a Gl/S-phase drug of the
present invention or
16


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
an S phase drug of the present invention. Preferred combinations of the
present invention
include (3-lapachone with gemcitabine; ~i-lapachone with 5-FU; (3-lapachone
with methotrexate;
(3-lapachone with 5-fluorodeoxyuridine; or (3-lapachone with cytarabine. (3-
lapachone with
gemcitabine is the most preferred combination. Preferred combinations of the
present invention
also include reduced [3-lapachone with gemcitabine; reduced ~-lapachone with 5-
FU; reduced (3-
lapachone with methotrexate; reduced (3-lapachone with 5-fluorodeoxyuridine;
or reduced (3-
lapachone with cytarabine. In preferred embodiments, ~3-lapachone, or
derivatives or analogs
thereof (as the G1/S-phase drug) are not combined with cis-platinum.
While [3-lapachone is the preferred G1/S-phase compound for use in the
composition in
accordance with the present invention, the invention is not limited in this
respect, and (3-
lapachone derivatives or analogs, such as lapachol, and pharmaceutical
compositions and
formulations thereof are part of the present invention. Such (3-lapachone
analogs include those
recited in PCT International Application PCT/US93/07878 (WO 94/04145), which
discloses
compounds of the formula:
R2
Formula II
where R and Rl are each independently hydrogen, substituted and unsubstituted
aryl, substituted
and unsubstituted alkenyl, substituted and unsubstituted alkyl and substituted
or unsubstituted
alkoxy. The alkyl groups preferably have from 1 to about 15 carbon atoms, more
preferably
from 1 to about 10 carbon atoms, still more preferably from 1 to about 6
carbon atoms. The
term alkyl unless otherwise modified refers to both cyclic and noncyclic
groups, although of
course cyclic groups will comprise at least three carbon ring members.
Straight or branched
chain noncyclic alkyl groups are generally more preferred than cyclic groups.
Straight chain
alkyl groups are generally more preferred than branched. The alkenyl groups
preferably have
from 2 to about 15 carbon atoms, more preferably from 2 to about 10 carbon
atoms, still more
preferably from 2 to 6 carbon atoms. Especially preferred alkenyl groups have
3 carbon atoms
(i.e., 1-propenyl or 2-propenyl), with the allyl moiety being particularly
preferred. Phenyl and
napthyl are generally preferred aryl groups. Allcoxy groups include those
alkoxy groups having
one or more oxygen linkage and preferably have from 1 to 15 carbon atoms, more
preferably
17


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
from 1 to about 6 carbon atoms. The substituted R and Rl groups may be
substituted at one or
more available positions by one or more suitable groups such as, for example,
alkyl groups such
as alkyl groups having from 1 to 10 carbon atoms or from 1 to 6 carbon atoms,
alkenyl groups
such as alkenyl groups having from 2 to 10 carbon atoms or 2 to 6 carbon
atoms, aryl groups
having from six to ten carbon atoms, halogen such as fluoro, chloro and bromo,
and N, O and S,
including heteroalkyl, e.g., heteroalkyl having one or more hetero atom
linkages (and thus
including alkoxy, aminoalkyl and thioalkyl) and from 1 to 10 carbon atoms or
from 1 to 6
carbon atoms.
Other (3-lapachone analogs contemplated in accordance with the present
invention
include those described in U.S. Patent No. 6,245,807, which discloses ~3-
lapachone analogs and
derivatives having the structure:
R~
R
Formula III
where R and Rl are each independently selected from hydrogen, hydroxy,
sulfhydryl, halogen,
substituted alkyl, unsubstituted alkyl, substituted alkenyl, unsubstituted
alkenyl, substituted aryl,
unsubstituted aryl, substituted alkoxy, unsubstituted alkoxy, and salts
thereof, where the dotted
double bond between the ring carbons represents an optional ring double bond.
Additional (3-lapachone analogs and derivatives are recited in PCT
International
Application PCT/LTS00110169 (W000161142), which disclose compounds of the
structure:
18


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
Formula IV
where RS and R6 may be independently selected from hydroxy, Cl-C6 alkyl, C1-C6
alkenyl, C1-
C6 alkoxy, C1-C6 alkoxycarbonyl, --(CHZ)"-phenyl; and R7 is hydrogen,
hydroxyl, C1-C6 alkyl,
CI-C6 alkenyl, C1-C6 alkoxy, C1-C6 alkoxycarbonyl, --(CH2)n amino, --(CH2)"-
aryl, --(CHZ)"_
heteroaryl, --(CH2)"-heterocycle, or --(CH2)"-phenyl, wherein n is an integer
from 0 to 10.
Other (3-lapachone analogs and derivatives are disclosed in U.S. Pat. No.
5,763,625, U.S.
Pat. No. 5,824,700, and U.S. Pat. No. 5,969,163, as well is in scientific
journal articles, such as
Sabba et al., JMed C7aem 27:990-994 (1984), which discloses [3-lapachone with
substitutions at
one or more of the following positions: 2-, 8- and/or 9- positions. See also
Portela et al.,
Biochem Pharm 51:275-283 (1996) (substituents at the 2- and 9- positions);
Maruyama et al.,
Clzem Lett 847-850 (1977); Sun et al., Tetrahedron Lett 39:8221-8224 (1998);
Goncalves et al.,
Molecular and Biochemical Parasitology 1:167-176 (1998) (substituents at the 2-
and 3-
positions); Gupta et al., Indian Jouf°raal of Chemistry 16B: 35-37
(1978); Gupta et al., Cuf°r Sci
46:337 (1977) (substituents at the 3- and 4- positions); DiChenna et al., JMed
Chem 44: 2486-
2489 (2001) (monoarylamino derivatives).
More preferably, analogs and derivatives contemplated by the present
application are
intended to encompass compounds having the general formula V and VI:
19


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
Formula V Formula VI
where the dotted double bond between the ring carbons represents an optional
ring double bond
and where R and Rl are each independently selected from hydrogen, hydroxy,
sulfhydryl,
halogen, substituted alkyl, unsubstituted alkyl, substituted alkenyl,
unsubstituted alkenyl,
substituted aryl, unsubstituted aryl, substituted alkoxy, unsubstituted
alkoxy, and salts thereof.
The alkyl groups preferably have from 1 to about 15 carbon atoms, more
preferably from 1 to
about 10 carbon atoms, still more preferably from 1 to about 6 carbon atoms.
The term alkyl
refers to both cyclic and noncyclic groups. Straight or branched chain
noncyclic alkyl groups
are generally more preferred than cyclic groups. Straight chain alkyl groups
are generally more
preferred than branched. The alkenyl groups preferably have from 2 to about 15
carbon atoms,
more preferably from 2 to about 10 carbon atoms, still more preferably from 2
to 6 carbon
atoms. Especially preferred alkenyl groups have 3 carbon atoms (a.e., 1-
propenyl or 2-
propenyl), with the allyl moiety being particularly preferred. Phenyl and
napthyl are generally
preferred aryl groups. Alkoxy groups include those alkoxy groups having one or
more oxygen
linkage and preferably have from 1 to 15 carbon atoms, more preferably from 1
to about 6
carbon atoms. The substituted R and Rl groups may be substituted at one or
more available
positions by one or more suitable groups such as, for example, alkyl groups
having from 1 to 10
carbon atoms or from 1 to 6 carbon atoms, alkenyl groups having from 2 to 10
carbon atoms or
2 to 6 carbon atoms, aryl groups having from six to ten carbon atoms, halogen
such as fluoro,
chloro and bromo, and N, O and S, including heteroalkyl, e.g., heteroalkyl
having one or more
hetero atom linkages (and thus including alkoxy, aminoalkyl and thioalkyl) and
from 1 to 10
carbon atoms or from 1 to 6 carbon atoms; and where RS and R6 may be
independently selected
from hydroxy, C1-C6 alkyl, C1-C~ alkenyl, C1-C~ alkoxy, C1-C6 alkoxycarbonyl, -
-(CHa)"-aryl, --
(CHa)"-heteroaryl, --(CHa)"-heterocycle, or --(CHZ)"-phenyl; and R7 is
hydrogen, hydroxyl, C1-


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
C6 alkyl, C1-C6 alkenyl, C1-C6 alkoxy, C1-C6 alkoxycarbonyl, --(CH2)"-amino, --
(CH2)"-aryl, --
(CH2)"-heteroaryl, --(CHa)n-heterocycle, or --(CH2)"-phenyl, wherein n is an
integer from 0 to
10.
Preferred analogs and derivatives also contemplated by the invention include
compounds
of the following general formula VII:
O
Formula VII
where Rl is (CH2)"-R2, where n is an integer from 0-10 and RZ is hydrogen, an
alkyl, an aryl, a
heteroaromatic, a heterocyclic, an aliphatic, an alkoxy, an allyloxy, a
hydroxyl, an amine, a thiol,
an amide, or a halogen.
Analogs and derivatives also contemplated by the invention include 4-acetoxy-
(3-
lapachone, 4-acetoxy-3-bromo-/3-lapachone, 4-lceto-(i-lapachone, 7-hydroxy-~-
lapachone, 7-
methoxy-(3-lapachone, 8-hydroxy-~3-lapachone, 8-methoxy-(3-lapachone, 8-chloro-
~-lapachone,
9-chloro-(3-lapachone, 8-methyl-(3-lapachone and 8,9-dimethoxy-(3-lapachone.
Preferred analogs and derivatives also contemplated by the invention include
compounds
of the following general formula VIII:
O
Formula VIII
where Rl-R4 are each, independently, selected from the group consisting of H,
C1-C6 alkyl, C1-
C6 alkenyl, C1-C~ alkoxy, C1-C6 alkoxycarbonyl, --(CHZ)"-aryl, --(CHZ)n-
heteroaryl, --(CH2)"
heterocycle, or --(CH2)"-phenyl; or Rl and R2 combined are a single
substituent selected from
21


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
the above group, and R3 and R4 combined are a single substituent selected from
the above
groups, in which case ---- is a double bond.
Preferred analogs and derivatives also contemplated by this invention include
dunnione
and 2-ethyl-6-hydroxynaphtho[2,3-b]-furan-4,5-dione.
Preferred analogs and derivatives also contemplated by the invention include
compounds
of the following general formula IX:
Formula IX
where Rl is selected from H, CH3, OCH3 and NOa.
Additional preferred (3-lapachone analogs useful in the methods and kits of
the invention
are represented by Formula X (see also the co-owned PCT patent application
entitled "NOVEL
LAPACHONE COMPOUNDS AND METHODS OF USE THEREOF", PCT/US2003/037219,
filed November 18, 2003, and claiming priority to U.S. provisional application
no. 60/427,283,
filed November 18, 2002):
R$
R
Formula X
or pharmaceutically acceptable salts thereof, or a regioisomeric mixture
thereof, wherein
R1-R6 are each, independently, selected from the group consisting of H, OH,
substituted and
unsubstituted Cl-C~ alkyl, substituted and unsubstituted Ci-C6 alkenyl,
substituted and
22


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
unsubstituted C1-C6 alkoxy, substituted and unsubstituted C1-C6
alkoxycarbonyl, substituted and
unsubstituted Cl-C6 acyl, -(CH2)"-amino, -(CH2)"-aryl, -(CH2)"-heterocycle,
and -(CH2)"-phenyl;
or one of R1 or R2 and one of R3 or R4; or one of R3 or R4 and one of RS or R6
form a fused
ring, wherein the ring has 4-8 ring members; R7-R10 are each, independently,
hydrogen,
hydroxyl, halogen, substituted or unsubstituted alkyl, substituted or
unsubstituted alkoxy, vitro,
cyano or amide; and n is an integer from 0 to 10.
In a preferred embodiment, Rl and R2 are alkyl, R3-R6 are, independently, H,
OH,
halogen, alkyl, alkoxy, substituted or unsubstituted acyl, substituted alkenyl
or substituted alkyl
carbonyl, and R7-R10 are hydrogen. In another preferred embodiment, Rl and R2
are each
methyl and R3-R10 are each hydrogen. In another preferred embodiment, Rl-R4
are each
hydrogen, RS and R6 are each methyl and R7-R10 are each hydrogen.
Additional preferred [3-lapachone analogs useful in the methods and kits of
the invention
are represented by Formula XI (see also the co-owned PCT patent application
entitled "NOVEL
LAPACHONE COMPOUNDS AND METHODS OF USE THEREOF", PCT/US2003/037219,
filed November 18, 2003):
R4
Formula XI
or pharmaceutically acceptable salts thereof, or a regioisomeric mixture
thereof, wherein
Rl-R4 are each, independently, selected from the group consisting of H, OH,
substituted and
unsubstituted C1-C6 alkyl, substituted and unsubstituted C1-C6 alkenyl,
substituted and
unsubstituted Cl-C6 alkoxy, substituted and unsubstituted C1-C6
alkoxycarbonyl, substituted and
unsubstituted CI-C6 acyl, -(CH2)~-amino, -(CH2)~-aryl, -(CHz)n-heterocycle,
and -(CHZ)"-phenyl;
or one of Rl or R2 and one of R3 or R4 form a fused ring, wherein the ring has
4-8 ring
members; RS-R8 are each, independently, hydrogen, hydroxyl, halogen,
substituted or
unsubstituted alkyl, substituted or unsubstituted alkoxy, vitro, cyano or
amide; and n is an
integer from 0 to 10. In certain embodiments of Formula XI, R1, R2, R3, R4,
R5, R6, R7 and
R8 are not each simultaneously H.
23


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
In an embodiment, the Gl/S phase drug is selected from the group consisting of
3,4-
dihydro-2,2-dimethyl-3-(3-methyl-2-butenyl)-2H-naphtho[1,2-b]pyran-5,6-dione,
3,4-dihydro-
2,2-dimethyl-2H-naphtho[1,2-b]thiopyran-5,6-dione and 3,4-dihydro-4,4-dimethyl-
2H-
naphtho[ 1,2-b]thiopyran-5,6-dione.
All stereoisomers of the compounds of the instant invention are contemplated,
either in
admixture or in pure or substantially pure form, including crystalline forms
of reacemic mixtures
and crystalline forms of individual isomers. The definition of the compounds
according to the
invention embraces all possible stereoisomers (e.g., the R and S
configurations for each
asymmetric center) and their mixtures. It very particularly embraces the
racemic forms and the
isolated optical isomers having a specified activity. The racemic forms can be
resolved by
physical methods, such as, for example, fractional crystallization, separation
or crystallization of
diastereomeric derivatives, separation by chiral column chromatography or
supercritical fluid
chromatography. The individual optical isomers can be obtained from the
racemates by
conventional methods, such as, for example, salt formation with an optically
active acid
followed by crystallization. Furthermore, all geometric isomers, such as E-
and Z-
configurations at a double bond, are within the scope of the invention unless
otherwise stated.
Certain compounds of this invention may exist in tautomeric forms. All such
tautomeric forms
of the compounds are considered to be within the scope of this invention
unless otherwise stated.
The present invention also includes one or more regioisomeric mixtures of an
analog or
derivative of (3-lapachone.
In additional aspects, a compound or combination of the present invention may
be
administered in combination with a chemotherapeutic agent. Exemplary
chemotheraputics with
activity against cell proliferative disorders, such as pancreatic cancer, are
known to those of
ordinary skill in the art, and may be found in reference texts such as the
Physician's Desk
Reference, 59th Edition, Thomson PDR (2005). For example, the chemotherapeutic
agent can be
a taxane, an aromatase inhibitor, an anthracycline, a microtubule targeting
drug, a topoisomerase
poison drug, a targeted monoclonal or polyconal antibody, an inhibitor of a
molecular target or
enzyme (e.g., a kinase inhibitor), or a cytidine analogue drug. In preferred
aspects, the
chemotherapeutic agent can be, but is not restricted to, tamoxifen,
raloxifene, anastrozole,
exemestane, letrozole, HERCEPTIN" (trastuzumab), GLEEVEC~ (imatanib), TAXOL~
(paclitaxel), IRESSA° (gefitinib), TARCEVATM (erlotinib),
cyclophosphamide, lovastatin,
minosine, araC, 5-fluorouracil (5-FLT), methotrexate (MTX), TAXOTERE~
(docetaxel),
ZOLADEX~ (goserelin), AVASTIN'~M (bevacizumab), vincristin, vinblastin,
nocodazole,
teniposide, etoposide, epothilone, navelbine, camptothecin, daunonibicin,
dactinomycin,
24


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
mitoxantrone, amsacrine, doxorubicin (adriamycin), epirubicin or idarubicin or
agents listed in
www.cancer.org/docroot/cdg/cdg O.asp. In another aspect, the chemotherapeutic
agent can be a
cytokine such as G-CSF (granulocyte colony stimulating factor). In another
aspect, (3-
lapachone, or a pharmaceutically acceptable salt, metabolite, analog or
derivative thereof may be
S administered in combination with radiation therapy. In yet another aspect,
(3-lapachone, or a
pharmaceutically acceptable salt, metabolite, analog or derivative thereof may
be administered
in combination with standard chemotherapy combinations such as, but not
restricted to, CMF
(cyclophosphamide, methotrexate and 5-fluorouracil), CAF (cyclophosphamide,
adriamycin and
5-fluorouracil), AC (adriamycin and cyclophosphamide), FEC (5-fluorouracil,
epirubicin, and
cyclophosphamide), ACT or ATC (adriamycin, cyclophosphamide, and paclitaxel),
or CMFP
(cyclophosphamide, methotrexate, 5-fluorouracil and prednisone).
As used herein, the term "salt" is a pharmaceutically acceptable salt and can
include acid
addition salts including hydrochlorides, hydrobromides, phosphates, sulphates,
hydrogen
sulphates, alkylsulphonates, arylsulphonates, acetates, benzoates, citrates,
maleates, fumarates,
succinates, lactates, and tartrates; alkali metal cations such as Na+, K+,
Li+, alkali earth metal
salts such as Mg or Ca, or organic amine salts.
As used herein, the term "metabolite" means a product of metabolism of a
compound of
the present invention, or a pharmaceutically acceptable salt, analog or
derivative thereof, that
exhibits a similar activity ih vivo to a compound of the present invention.
As used herein, the term "prodrug" means a compound of the present invention
covalently linked to one or more pro-moieties, such as an amino acid moiety or
other water
solubilizing moiety. A compound of the present invention may be released from
the pro-moiety
via hydrolytic, oxidative, and/or enzymatic release mechanisms. In an
embodiment, a prodrug
composition of the present invention exhibits the added benefit of increased
aqueous solubility,
improved stability, and improved pharmacokinetic profiles. The pro-moiety may
be selected to
obtain desired prodrug characteristics. For example, the pro-moiety, e.g., an
amino acid moiety
or other water solubilizing moiety may be selected based on solubility,
stability, bioavailability,
and/or in vivo delivery or uptake.
II. Methods of Treatment
As used herein, a "subject" can be any mammal, e.g., a human, a primate,
mouse, rat,
dog, cat, cow, horse, pig, sheep, goat, camel. In a preferred aspect, the
subject is a human.
As used herein, a "subj ect in need thereof ' is a subj ect having a cell
proliferative
disorder, or a subject having an increased risk of developing a cell
proliferative disorder relative
to the population at large. In one aspect, a subject in need thereof has a
precancerous condition.


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
In a preterrect aspect, a subject in need thereof has cancer. In an aspect,
the subject may be
suffering from a known (i.e., diagnosed) condition characterized by cell
hyperproliferation (e.g.,
cancer).
As used herein, the term "cell proliferative disorder" refers to conditions in
which
unregulated or abnormal growth, or both, of cells can lead to the development
of an unwanted
condition or disease, which may or may not be cancerous. In one aspect, a cell
proliferative
disorder includes a non-cancerous condition, e.g., rheumatoid arthritis;
inflammation;
autoimmune disease; lymphoproliferative conditions; acromegaly; rheumatoid
spondylitis;
osteoarthritis; gout, other arthritic conditions; sepsis; septic shock;
endotoxic shock; gram-
negative sepsis; toxic shock syndrome; asthma; adult respiratory distress
syndrome; chronic
obstructive pulmonary disease; chronic pulmonary inflammation; inflammatory
bowel disease;
Crohn's disease; psoriasis; eczema; ulcerative colitis; pancreatic fibrosis;
hepatic fibrosis; acute
and chronic renal disease; irritable bowel syndrome; pyresis; restenosis;
cerebral malaria; stroke
and ischemic injury; neural trauma; Alzheimer's disease; Huntington's disease;
Parkinson's
disease; acute and chronic pain; allergic rhinitis; allergic conjunctivitis;
chronic heart failure;
acute coronary syndrome; cachexia; malaria; leprosy; leishmaniasis; Lyme
disease; Reiter's
syndrome; acute synovitis; muscle degeneration, bursitis; tendonitis;
tenosynovitis; herniated,
ruptures, or prolapsed intervertebral disk syndrome; osteopetrosis;
thrombosis; restenosis;
silicosis; pulmonary sarcosis; bone resorption diseases, such as osteoporosis;
graft-versus-host
reaction; Multiple Sclerosis; lupus; fibromyalgia; AIDS and other viral
diseases such as Herpes
Zoster, Herpes Simplex I or II, influenza virus and cytomegalovirus; and
diabetes mellitus. In
another aspect, a cell proliferative disorder includes a precancer or a
precancerous condition. In
another aspect, a cell proliferative disorder includes cancer. In another
aspect, a cell
proliferative disorder includes a non-cancerous cell proliferative disorder.
Various cancers to be
treated include but are not limited to breast cancer, lung cancer, colon
cancer, colorectal cancer,
pancreatic cancer, ovarian cancer, prostate cancer, renal carcinoma, liver
cancer, hepatoma,
brain cancer, skin cancer, melanoma, multiple myeloma, chronic myelogenous
leukemia,
hematologic tumor, and lymphoid tumor, including metastatic lesions in other
tissues or organs
distant from the primary tumor site. Cancers to be treated include but are not
limited to
sarcoma, carcinoma, and adenocarcinoma. In one aspect, a "precancer cell" or
"precancerous
cell" is a cell manifesting a cell proliferative disorder that is a precancer
or a precancerous
condition. In another aspect, a "cancer cell" or "cancerous cell" is a cell
manifesting a cell
proliferative disorder that is a cancer. Any reproducible means of measurement
may be used to
identify cancer cells or precancerous cells. In a preferred aspect, cancer
cells or precancerous
26


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
cells are identified by histological typing or grading of a tissue sample
(e.g., a biopsy sample).
In another aspect, cancer cells or precancerous cells are identified through
the use of appropriate
molecular markers.
In one aspect, a solid tumor is formed as a result of a cancer selected from
the group
consisting of breast cancer, lung cancer, colon cancer, colorectal cancer,
pancreatic cancer,
ovarian cancer, prostate cancer, renal carcinoma, liver cancer, hepatoma,
brain cancer, skin
cancer, and melanoma.
A "cell proliferative disorder of the hematologic system" is a cell
proliferative disorder
involving cells of the hematologic system. In one aspect, a cell proliferative
disorder of the
hematologic system includes lymphoma, leukemia, myeloid neoplasms, mast cell
neoplasms,
myelodysplasia, benign monoclonal gammopathy, lymphomatoid granulomatosis,
lymphomatoid papulosis, polycythemia vera, chronic myelocytic leukemia,
agnogenic myeloid
metaplasia, and essential thrombocythemia. In another aspect, a cell
proliferative disorder of the
hematologic system includes hyperplasia, dysplasia, and metaplasia of cells of
the hematologic
system. In a preferred aspect, compositions of the present invention may be
used to treat a
cancer selected from the group consisting of a hematologic cancer of the
present invention or a
hematologic cell proliferative disorder of the present invention. In one
aspect, a hematologic
cancer of the present invention (i.e., a hematologic tumor) includes multiple
myeloma,
lymphoma (including Hodgkin's lymphoma, non-Hodgkin's lymphoma, childhood
lymphomas,
and lymphomas of lymphocytic and cutaneous origin), leukemia (including
childhood leukemia,
hairy-cell leukemia, acute lymphocytic leukemia, acute myelocytic leukemia,
chronic
lymphocytic leukemia, chronic myelocytic leukemia, chronic myelogenous
leukemia, and mast
cell leukemia), myeloid neoplasms and mast cell neoplasms.
A "cell proliferative disorder of the lung" is a cell proliferative disorder
involving cells
of the lung. In one aspect, a cell proliferative disorder includes a pre-
cancer or precancerous
condition of the lung. In one aspect, a cell proliferative disorder of the
lung includes a non-
cancerous cell proliferative disorder of the lung. In another aspect, a cell
proliferative disorder
includes lung cancer, including metastatic lesions in other tissues or organs
distant from the
primary tumor site. In one aspect, a "precancer cell" or "precancerous cell"
is a cell manifesting
a cell proliferative disorder that is a precancer or a precancerous condition.
In another aspect, a
"cancer cell" or "cancerous cell" is a cell manifesting a cell proliferative
disorder that is a
cancer. Any reproducible means of measurement may be used to identify cancer
cells or
precancerous cells. In a preferred aspect, cancer cells or precancerous cells
are identified by
histological typing or grading of a tissue sample (e.g., a biopsy sample). In
another aspect,
27


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
cancer cells or precancerous cells are identified through the use of
appropriate molecular
markers.
In a preferred aspect, the cell proliferative disorder of the lung is lung
cancer. In a
preferred aspect, compositions of the present invention may be used to treat
lung cancer or cell
proliferative disorders of the lung. In one aspect, lung cancer includes all
forms of cancer of the
lung. Cancers to be treated include but axe not limited to sarcoma, carcinoma,
and
adenocarcinoma. In another aspect, lung cancer includes malignant lung
neoplasms, carcinoma
in situ, typical carcinoid tumors, and atypical carcinoid tumors. In another
aspect, lung cancer
includes small cell lung cancer ("SCLC"), non-small cell lung cancer
("NSCLC"), squamous cell
carcinoma, adenocarcinoma, small cell carcinoma, large cell carcinoma,
adenosquamous cell
carcinoma, and mesothelioma. In another aspect, lung cancer includes "scar
carcinoma,"
bronchioalveolar carcinoma, giant cell carcinoma, spindle cell carcinoma, and
large cell
neuroendocrine carcinoma. In another aspect, lung cancer includes lung
neoplasms having
histologic and ultrastructual heterogeneity (e.g., mixed cell types). In one
aspect, lung cancer
includes mixed small cell/large cell carcinoma.
In one aspect, cell proliferative disorders of the lung include all forms of
cell
proliferative disorders affecting lung cells. In one aspect, cell
proliferative disorders of the lung
include lung cancer and precancerous conditions of the lung. In one aspect,
cell proliferative
disorders of the lung include hyperplasia, metaplasia, and dysplasia of the
lung. In one aspect,
cell proliferative disorders to be treated include sporadic and hereditary
cell proliferative
disorders of the lung. In one aspect, cell proliferative disorders of the lung
include benign
tumors of the lung. In another aspect, cell proliferative disorders of the
lung include asbestos-
induced hyperplasia, squamous metaplasia, and benign reactive mesothelial
metaplasia. In
another aspect, cell proliferative disorders of the lung include replacement
of columnar
epithelium with stratified squamous epithelium, and mucosal dysplasia. In
another aspect,
individuals exposed to inhaled injurious environmental agents such as
cigarette smoke and
asbestos may be at increased risk for developing cell proliferative disorders
of the lung. In
another aspect, prior lung diseases that may predispose individuals to
development of cell
proliferative disorders of the lung include chronic interstitial lung disease,
necrotizing
pulmonary disease, scleroderma, rheumatoid disease, sarcoidosis, interstitial
pneumonitis,
tuberculosis, repeated pneumonias, idiopathic pulmonary fibrosis, granulomata,
asbestosis,
fibrosing alveolitis, and Hodgkin's disease.
In one aspect, a lung cancer that is to be treated has arisen in a subject
equal to or older
than 30 years old, or a subject younger than 30 years old. In one aspect, a
lung cancer that is to
2~


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
be treated has arisen in a subject equal to or older than 50 years old, or a
subject younger than 50
years old. In one aspect, a lung cancer that is to be treated has arisen in a
subject equal to or
older than 70 years old, or a subject younger than 70 years old. In one
aspect, a lung cancer that
is to be treated has been typed to identify a familial or spontaneous mutation
in p53, Rb,
CDI~N2A (P16INK4A), FHIT, myc, ras, TP73, MADH2, MADH4, PPP2Rlb, or PTEN. In
another aspect, a lung cancer that is to be treated is associated with a GSTM1
null allele. In
another aspect, a lung cancer that is to be treated is associated with a
mutation selected from the
group consisting of del(3p), del(9p) and del(1p36). In one aspect, a lung
cancer that is to be
treated is associated with elevated levels of CEA (carcinoembryonic antigen)
or NSE (neuron-
specific enolase), or an upregulation of one or more components of telomerase.
In another
aspect, a lung cancer that is to be treated is associated with an increased
level of a marker
selected from the group consisting of MOC-l, MOC-21, MOC-31, MOC-32, and MOC-
52.
In one aspect, a lung cancer that is to be treated includes a localized tumor
of the lung. In
one aspect, a lung cancer that is to be treated includes a tumor of the lung
that is associated with
a negative regional lymph node biopsy. In one aspect, a lung cancer that is to
be treated includes
a tumor of the lung that is associated with a positive regional lymph node
biopsy. In another
aspect, a lung cancer that is to be treated includes a tumor of the lung that
has been typed as
having nodal negative status (e.g., node-negative) or nodal positive status
(e.g., node-positive).
In another aspect, a lung cancer that is to be treated includes a tumor of the
lung that has
metastasized to other locations in the body. In one aspect, a lung cancer that
is to be treated is
classified as having metastasized to a location selected from the group
consisting of lymph node,
stomach, bile duct, lung, liver, bone, and brain. In another aspect a lung
cancer that is to be
treated is classified according to a characteristic selected from the group
consisting of metastatic,
limited stage, extensive stage, unresectable, resectable, localized, regional,
local-regional,
locally advanced, distant, multicentric, bilateral, ipsilateral,
contralateral, newly diagnosed,
recurrent, and inoperable.
In one aspect, a lung cancer that is to be treated has been staged according
to the
American Joint Committee on Cancer (AJCC) TNM classification system, where the
tumor (T)
has been assigned a stage of Tis, T1, T2, T3,T4; and where the regional lymph
nodes (I~ have
been assigned a stage of NX, N0, Nl, N2, N2a, N2b, N3, N3a, N3b, or N3c; and
where distant
metastasis (M) has been assigned a stage of MX, M0, or M1. In another aspect,
a lung cancer
that is to be treated has been staged according to an American Joint Committee
on Cancer
(AJCC) classification as Stage 0, I, IA, IB, II, IIA, IIB, III, IIIA, IIIB,
IIIC and IV lung cancer.
In another aspect, a lung cancer that is to be treated has been assigned a
grade according to an
29


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
AJCC classification as Grade GX (e.g., grade cannot be assessed), Grade 1,
Grade 2, Grade 3 or
Grade 4.
In one aspect, a lung cancer that is to be treated includes a tumor that has
been
determined to be less than or equal to about 3 centimeters in diameter. In
another aspect, a lung
cancer that is to be treated includes a tumor that has been determined to be
from about 3 to about
centimeters in diameter. In another aspect, a lung cancer that is to be
treated includes a tumor
that has been determined to be greater than or equal to about 3 centimeters in
diameter. In
another aspect, a lung cancer that is to be treated includes a tumor that has
been determined to be
greater than 5 centimeters in diameter. In another aspect, a lung cancer that
is to be treated is
classified by microscopic appearance as well differentiated, moderately
differentiated, poorly
differentiated, or undifferentiated. In another aspect, a lung cancer that is
to be treated is
classified by microscopic appearance with respect to mitosis count (e.g.,
amount of cell division)
or nuclear pleiomorphism (e.g., change in cells). In another aspect, a lung
cancer that is to be
treated is classified by microscopic appearance as being associated with areas
of necrosis (e.g.,
areas of dying or degenerating cells). In one aspect, a lung cancer that is to
be treated is
classified as having an abnormal karyotype, having an abnormal number of
chromosomes, or
having one or more chromosomes that are abnormal in appearance. In one aspect,
a lung cancer
that is to be treated is classified as being aneuploid, triploid, tetraploid,
or as having an altered
ploidy. In one aspect, a lung cancer that is to be treated is classified as
having a chromosomal
translocation, or a deletion or duplication of an entire chromosome, or a
region of deletion,
duplication or amplification of a portion of a chromosome.
A "cell proliferative disorder of the colon" is a cell proliferative disorder
involving cells
of the colon. In one aspect, a cell proliferative disorder includes a pre-
cancer or precancerous
condition of the colon. In one aspect, a cell proliferative disorder of the
colon includes a non-
cancerous cell proliferative disorder of the colon. In another aspect, a cell
proliferative disorder
includes colon cancer, including metastatic lesions in other tissues or organs
distant from the
primary tumor site. In one aspect, a "precancer cell" or "precancerous cell"
is a cell manifesting
a cell proliferative disorder that is a precancer or a precancerous condition.
In another aspect, a
"cancer cell" or "cancerous cell" is a cell manifesting a cell proliferative
disorder that is a
cancer. Any reproducible means of measurement may be used to identify cancer
cells or
precancerous cells. In a preferred aspect, cancer cells or precancerous cells
are identified by
histological typing or grading of a tissue sample (e.g., a biopsy sample). In
another aspect,
cancer cells or precancerous cells are identified through the use of
appropriate molecular
markers.


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
In a preferred aspect, the cell proliferative disorder of the colon is colon
cancer. In a
preferred aspect, compositions of the present invention may be used to treat
colon cancer or cell
proliferative disorders of the colon. In one aspect, colon cancer includes all
forms of cancer of
the colon. In one aspect, a colon cancer to be treated includes carcinoma,
sarcoma, and
adenocarcinoma. In another aspect, colon cancer includes sporadic and
hereditary colon
cancers. In another aspect, colon cancer includes malignant colon neoplasms,
carcinoma in situ,
leiomyosarcomas, typical carcinoid tumors, and atypical carcinoid tumors. In
another aspect,
colon cancer includes adenocarcinoma, squamous cell carcinoma, signet ring
cell
adenocarcinoma and adenosquamous cell carcinoma. Tn another aspect, colon
cancer is
associated with a hereditary syndrome selected from the group consisting of
hereditary
nonpolyposis colorectal cancer (HNPCC), familial adenomatous polyposis (FAP),
Gardner's
syndrome, Peutz-Jeghers syndrome, Turcot's syndrome and juvenile polyposis. In
another
aspect, colon cancer is caused by a hereditary syndrome selected from the
group consisting of
hereditary nonpolyposis colorectal cancer (HNPCC), familial adenomatous
polyposis (FAP),
Gardner's syndrome, Peutz-Jeghers syndrome, Turcot's syndrome and juvenile
polyposis.
In one aspect, cell proliferative disorders of the colon include all forms of
cell
proliferative disorders affecting colon cells. In one aspect, cell
proliferative disorders of the
colon include colon ca~zcer, precancerous conditions of the colon, adenomatous
polyps of the
colon and metachronous lesions of the colon. In one aspect, a cell
proliferative disorder of the
colon includes adenoma. In one aspect, cell proliferative disorders of the
colon are characterized
by hyperplasia, metaplasia, and dysplasia of the colon. In another aspect,
prior colon diseases
that may predispose individuals to development of cell proliferative disorders
of the colon
include prior colon cancer. In another aspect, current disease that may
predispose individuals to
development of cell proliferative disorders of the colon include Crohn's
disease and ulcerative
colitis. In one aspect, a cell proliferative disorder of the colon is
associated with a mutation in a
gene selected from the group consisting of p53, t°as, FAP and DCC. In
another aspect, an
individual has an elevated risk of developing a cell proliferative disorder of
the colon due to the
presence of a mutation in a gene selected from the group consisting of p53,
ras, FAP and DCC.
In one aspect, a colon cancer that is to be treated has arisen in a subject
equal to or older
than 30 years old, or a subject younger than 30 years old. W one aspect, a
colon cancer that is to
be treated has arisen in a subject equal to or older than 50 years old, or a
subject younger than 50
years old. In one aspect, a colon cancer that is to be treated has arisen in a
subject equal to or
older than 70 years old, or a subject younger than 70 years old. In one
aspect, a colon cancer
that is to be treated has been typed to identify a familial or spontaneous
mutation in p53, Rb,
31


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
myc or ras. In one aspect, a colon cancer that is to be treated is associated
with a mutation in a
gene selected from the group consisting of MSH2, MSH6, MLH1, PMS 1, PMS2,
TGFBR2,
BAX, and APC. In another aspect, a colon cancer that is to be treated is
associated with a
mutation selected from the group consisting of de(lp),del(~p), del(~q21),
del(17p), LOHl7p,
and chrmosome 18q allelic loss. In one aspect, a colon cancer that is to be
treated has been
typed as having the replication error phenotype RER+. In one aspect, a colon
cancer that is to
be treated has been typed as CpG island methylator phenotype positive (CIMP+)
or CpG island
methylator phenotype negative (CINIP-). In one aspect, a colon cancer that is
to be treated is
associated with elevated levels of CEA (carcinoembryonic antigen).
In one aspect, a colon cancer that is to be treated includes a localized tumor
of the colon.
In one aspect, a colon cancer that is to be treated includes a tumor of the
colon that is associated
with a negative regional lymph node biopsy. In one aspect, a colon cancer that
is to be treated
includes a tumor of the colon that is associated with a positive regional
lymph node biopsy. In
another aspect, a colon cancer that is to be treated includes a tumor of the
colon that has been
typed as having nodal negative status (e.g., node-negative) or nodal positive
status (e.g., node-
positive). In another aspect, a colon cancer that is to be treated includes a
tumor of the colon
that has metastasized to other locations in the body. In one aspect, a colon
cancer that is to be
treated is classified as having metastasized to a location selected from the
group consisting of
lymph node, stomach, bile duct, liver, bone, ovary, peritoneum and brain. In
another aspect a
colon cancer that is to be treated is classified according to a characteristic
selected from the
group consisting of metastatic, limited stage, extensive stage, unresectable,
resectable, localized,
regional, local-regional, locally advanced, distant, multicentric, bilateral,
ipsilateral,
contralateral, newly diagnosed, recurrent, and inoperable.
111 one aspect, a colon cancer that is to be treated has been staged according
to the
American Joint Committee on Cancer (AJCC) TNM classification system, where the
tumor (T)
has been assigned a stage of Tx, Tis, T1, T2, T3, T4; and where the regional
lymph nodes (N)
have been assigned a stage of NX, N0, N1, N2, N2a, N2b, N3, N3a, N3b, or N3c;
and where
distant metastasis (M) has been assigned a stage of MX, M0, or Ml . In another
aspect, a colon
cancer that is to be treated has been staged according to an American Joint
Committee on Cancer
(AJCC) classification as Stage 0, I, II, IIA, IIB, III, IIIA, IIIB, IIIC and
IV colon cancer. In
another aspect, a colon cancer that is to be treated has been assigned a grade
according to an
AJCC classification as Grade GX (e.g., grade cannot be assessed), Grade 1,
Grade 2, Grade 3 or
Grade 4. In another aspect, a colon cancer that is to be treated has been
assigned a grade
according to the Dukes staging system of A, B, or C. In another aspect, a
colon cancer that is to
32


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
be treated has been assigned a grade according to the Astler-Coller staging
system of A, B 1, B2,
B3 or C1, C2, C3, or D.
In one aspect, a colon cancer that is to be treated includes a tumor that has
been
determined to be less than or equal to about 2 centimeters in diameter. In
another aspect, a colon
cancer that is to be treated includes a tumor that has been determined to be
from about 2 to about
centimeters in diameter. In another aspect, a colon cancer that is to be
treated includes a tumor
that has been determined to be greater than or equal to about 2 centimeters in
diameter. In
another aspect, a colon cancer that is to be treated includes a tumor that has
been determined to
be greater than 5 centimeters in diameter. In another aspect, a colon cancer
that is to be treated
is classified by microscopic appearance as well differentiated, moderately
differentiated, poorly
differentiated, or undifferentiated. Iii another aspect, a colon cancer that
is to be treated is
classified by microscopic appearance with respect to mitosis count (e.g.,
amount of cell division)
or nuclear pleiomorphism (e.g., change in cells). In another aspect, a colon
cancer that is to be
treated is classified by microscopic appearance as being associated with areas
of necrosis (e.g.,
areas of dying or degenerating cells). In one aspect, a colon cancer that is
to be treated is
classified as having an abnormal karyotype, having an abnormal number of
chromosomes, or
having one or more chromosomes that are abnormal in appearance. In one aspect,
a colon
cancer that is to be treated is classified as being aneuploid, triploid,
tetraploid, or as having an
altered ploidy. In one aspect, a colon cancer that is to be treated is
classified as having a
chromosomal translocation, or a deletion or duplication of an entire
chromosome, or a region of
deletion, duplication or amplification of a portion of a chromosome.
A "cell proliferative disorder of the breast" is a cell proliferative disorder
involving cells
of the breast. In one aspect, cell proliferative disorders of the breast
include all forms of cell
proliferative disorders affecting breast cells. In one aspect, cell
proliferative disorders of the
breast include breast cancer, a precancer or precancerous condition of the
breast, benign growths
or lesions of the breast, and malignant growths or lesions of the breast, and
metastatic lesions in
tissue and organs in the body other than the breast. In another aspect, cell
proliferative disorders
of the breast include hyperplasia, metaplasia, and dysplasia of the breast.
In one aspect, a cell proliferative disorder of the breast is a precancerous
condition of the
breast. In one aspect, compositions of the present invention may be used to
treat a precancerous
condition of the breast. In one aspect, a precancerous condition of the breast
includes atypical
hyperplasia of the breast, ductal carcinoma in situ (DCIS), intraductal
carcinoma, lobular
carcinoma in situ (LCIS), lobular neoplasia, and stage 0 or grade 0 growth or
lesion of the breast
(e.g., stage 0 or grade 0 breast cancer, or carcinoma in situ). In another
aspect, a precancerous
33


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
condition of the breast has been staged according to the TNM classification
scheme as accepted
by the American Joint Committee on Cancer (AJCC), where the primary tumor (T)
has been
assigned a stage of TO or Tis; and where the regional lymph nodes (I~ have
been assigned a
stage of N0; and where distant metastasis (M) has been assigned a stage of M0.
In a preferred aspect, the cell proliferative disorder of the breast is breast
cancer. In a
preferred aspect, compositions of the present invention may be used to treat
breast cancer. In
one aspect, breast cancer includes all forms of cancer of the breast. In one
aspect, breast cancer
includes primary epithelial breast cancers. In another aspect, breast cancer
includes cancers in
which the breast is involved by other tumors such as lymphoma, sarcoma or
melanoma. In
another aspect, breast cancer includes carcinoma of the breast, ductal
carcinoma of the breast,
lobular carcinoma of the breast, undifferentiated carcinoma of the breast,
cystosarcoma
phyllodes of the breast, angiosarcoma of the breast, and primary lymphoma of
the breast. In one
aspect, breast cancer includes Stage I, II, IIIA, IIIB, IIIC and IV breast
cancer. In one aspect,
ductal carcinoma of the breast includes invasive carcinoma, invasive carcinoma
in situ with
predominant intraductal component, inflammatory breast cancer, and a ductal
carcinoma of the
breast with a histologic type selected from the group consisting of comedo,
mucinous (colloid),
medullary, medullary with lymphcytic infiltrate, papillary, scirrhous, and
tubular. In one aspect,
lobular carcinoma of the breast includes invasive lobular carcinoma with
predominant in situ
component, invasive lobular carcinoma, and infiltrating lobular carcinoma. In
one aspect, breast
cancer includes Paget's disease, Paget's disease with intraductal carcinoma,
and Paget's disease
with invasive ductal carcinoma. In another aspect, breast cancer includes
breast neoplasms
having histologic and ultrastructual heterogeneity (e.~., mixed cell types).
In one aspect, a breast cancer that is to be treated has been typed to
identify a familial or
spontaneous mutation in BRCA1, BRCA2, or p53. Iii one aspect, a breast cancer
that is to be
treated has been typed as having a HER2/neu gene amplification, as
overexpressing HER2/neu,
or as having a low, intermediate or high level of HER2/neu expression. In
another aspect, a
breast cancer that is to be treated has been typed for a marker selected from
the group consisting
of estrogen receptor (ER), progesterone receptor (PR), human epidermal growth
factor receptor-
2, Ki-67, CA15-3, CA 27-29, and c-Met. In one aspect, a breast cancer that is
to be treated has
been typed as ER-unknown, ER-rich or ER-poor. In another aspect, a breast
cancer that is to be
treated has been typed as ER-negative or ER-positive. ER-typing of a breast
cancer may be
performed by any reproducible means. In a preferred aspect, ER-typing of a
breast cancer may
be performed as set forth in Onkologie 27: 175-179 (2004). In one aspect, a
breast cancer that is
to be treated has been typed as PR-unknown, PR-rich or PR-poor. In another
aspect, a breast
34


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
cancer that is to be treated has been typed as PR-negative or PR-positive. In
another aspect, a
breast cancer that is to be treated has been typed as receptor positive or
receptor negative. In one
aspect, a breast cancer that is to be treated has been typed as being
associated with elevated
blood levels of CA 15-3, or CA 27-29, or both.
A "cell proliferative disorder of the pancreas" is a cell proliferative
disorder involving
cells of the pancreas. In one aspect, a cell proliferative disorder includes a
pre-cancer or
precancerous condition of the pancreas. In one aspect, a cell proliferative
disorder of the
pancreas includes a non-cancerous cell proliferative condition of the
pancreas. In another aspect,
a cell proliferative disorder includes pancreatic cancer, including metastatic
lesions in other
tissues or organs distant from the primary tumor site. In one aspect, a
"precancer cell" or
"precancerous cell" is a cell manifesting a cell proliferative disorder that
is a precancer or a
precancerous condition. In another aspect, a "cancer cell" or "cancerous cell"
is a cell
manifesting a cell proliferative disorder that is a cancer. Any reproducible
means of
measurement may be used to identify cancer cells or precancerous cells. In a
preferred aspect,
cancer cells or precancerous cells are identified by histological typing or
grading of a tissue
sample (e.g., a biopsy sample). W another aspect, cancer cells or precancerous
cells are
identified through the use of appropriate molecular markers.
In a preferred aspect, the cell proliferative disorder of the pancreas is
pancreatic cancer.
In a preferred aspect, compositions of the present invention may be used to
treat pancreatic
cancer or cell proliferative disorders of the pancreas. In one aspect,
pancreatic cancer includes
all forms of cancer of the pancreas. In one aspect, a pancreatic cancer to be
treated includes
carcinoma, sarcoma, and adenocarcinoma. In another aspect, a pancreatic cancer
to be treated
includes sporadic and hereditary pancreatic cancers. In another aspect, a
pancreatic cancer to be
treated includes duct cell carcinoma, acinar cell carcinoma, papillary
mucinous carcinoma,
signet ring carcinoma, adenosquamous carcinoma, undifferentiated carcinoma,
mucinous
carcinoma, giant cell carcinoma, small cell carcinoma, cystadenocarcinoma,
serous
cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified pancreatic
cancer, and
pancreatoblastoma. In another aspect, a pancreatic cancer to be treated
includes mixed type
pancreatic cancers (e.g., ductal-endocrine or acinar-endocrine). In one
aspect, a pancreatic
cancer to be treated includes ductal adenocarcinoma, adenosquamous carcinoma,
pleiomorphic
giant cell carcinoma, mucinous adenocarcinoma, osteoclast-like giant cell
carcinoma, mucinous
cystadenocarcinoma, acinar carcinoma, unclassified large cell carcinoma, and
small cell
carcinoma.


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
In one aspect, cell proliferative disorders of the pancreas to be treated
include all forms
of cell proliferative disorders affecting pancreas cells. In one aspect, cell
proliferative disorders
of the pancreas to be treated include pancreatic cancer, a precancer or
precancerous condition of
the pancreas, benign growths or lesions of the pancreas, and malignant growths
or lesions of the
pancreas, and metastatic lesions in tissue and organs in the body other than
the pancreas. In
another aspect, cell proliferative disorders of the pancreas to be treated
include hyperplasia,
metaplasia, and dysplasia of the pancreas. In another aspect, cell
proliferative disorders of the
pancreas to be treated include mucinous cystadenoma, intraductal papillary
neoplasm, serous
cystadenoma, papillary-cystic neoplam, mucinous cystic tumor with dysplasia,
intraductal
papillary mucinous tumor with dysplasia, and pseudopapillary solid tumor. In
one aspect,
current disease that may predispose individuals to development of cell
proliferative disorders of
the pancreas include diabetes mellitus or pancreatitis. In another aspect,
individuals are at an
increased risk of developing a cell proliferative disorder of the pancreas,
such as pancreatic
cancer, due to a hereditary syndrome selected from the group consisting of
hereditary
nonpolyposis colorectal cancer (HNPCC) and familial adenomatous polyposis
(FAP). In another
aspect, individuals are at an increased risk of developing a cell
proliferative disorder of the
pancreas, such as pancreatic cancer, due to a mutation in a gene selected from
the group
consisting of MSH2, MSH6, MLH1, and APC.
In one aspect, a pancreatic cancer that is to be treated has arisen in a
subject equal to or
older than 30 years old, or a subject younger than 30 years old. In one
aspect, a pancreatic
cancer that is to be treated has arisen in a subject equal to or older than 50
years old, or a subject
younger than 50 years old. In one aspect, a pancreatic cancer that is to be
treated has arisen in a
subj ect equal to or older than 70 years old, or a subj ect younger than 70
years old. In one aspect,
a pancreatic cancer that is to be treated has been typed to identify a
familial or spontaneous
mutation in p53, Rb, myc or ras. In one aspect, a pancreatic cancer that is to
be treated is
associated with a mutation in a gene selected from the group consisting of K-
Ras, p53, BRCA2,
p16 (CDKN2A), MADH4 (DPC4), STKl l, MSH2, MSH6, MLH1, and APC. In one aspect,
a
pancreatic cancer that is to be treated is associated with elevated levels of
expression of a growth
factor selected from the group consisting of EGF, TGF alpha, TGF beta 1-3,
aFGF, and bTGF.
In one aspect, a pancreatic cancer that is to be treated is associated with
elevated levels in blood
of CEA (carcinoembryonic antigen). In one aspect, a pancreatic cancer that is
to be treated is
associated with increased levels in blood or increased cellular expression of
tumor marker
carbohydrate antigen 19-9 (CA 19-9).
36


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
In one aspect, a pancreatic cancer that is to be treated includes a localized
tumor of the
pancreas. In one aspect, a pancreatic cancer that is to be treated includes a
tumor of the pancreas
that is associated with a negative regional lymph node biopsy. In one aspect,
a pancreatic cancer
that is to be treated includes a tumor of the pancreas that is associated with
a positive regional
lymph node biopsy. In another aspect, a pancreatic cancer that is to be
treated includes a tumor
of the pancreas that has been typed as having nodal negative status (e.g.,
node-negative) or nodal
positive status (e.g., node-positive). In another aspect, a pancreatic cancer
that is to be treated
includes a tumor of the pancreas that has metastasized to other locations in
the body. In one
aspect, a pancreatic cancer that is to be treated is classified as having
metastasized to a location
selected from the group consisting of lymph node, stomach, bile duct, liver,
bone, ovary,
peritoneum and brain. In another aspect a pancreatic cancer that is to be
treated is classified
according to a characteristic selected from the group consisting of
metastatic, limited stage,
extensive stage, unresectable, resectable, locally advanced, localized,
regional, local-regional,
locally advanced, distant, multicentric, bilateral, ipsilateral,
contralateral, newly diagnosed,
recurrent, and inoperable.
In one aspect, a pancreatic cancer that is to be treated has been staged
according to the
American Joint Committee on Cancer (AJCC) TNM classification system, where the
tumor (T)
has been assigned a stage of Tx, T1, T2, T3, T4; and where the regional lymph
nodes (N] have
been assigned a stage of NX, N0, N1; and where distant metastasis (M) has been
assigned a
stage of MX, M0, or M1. In another aspect, a pancreatic cancer that is to be
treated has been
staged according to an American Joint Committee on Cancer (AJCC)
classification as Stage 0, I,
IA, IB, II, IIA, IIB, III, and IV pancreatic cancer. In another aspect, a
pancreatic cancer that is to
be treated has been assigned a grade according to an AJCC classification as
Grade GX (e.g.,
grade cannot be assessed), Grade 1, Grade 2, Grade 3 or Grade 4.
In one aspect, a pancreatic cancer that is to be treated includes a tumor that
has been
determined to be less than or equal to about 2 centimeters in diameter. In
another aspect, a
pancreatic cancer that is to be treated includes a tumor that has been
determined to be from
about 2 to about 5 centimeters in diameter. In another aspect, a pancreatic
cancer that is to be
treated includes a tumor that has been determined to be greater than or equal
to about 2
centimeters in diameter. In another aspect, a pancreatic cancer that is to be
treated includes a
tumor that has been determined to be greater than 5 centimeters in diameter.
In another aspect, a
pancreatic cancer that is to be treated is classified by microscopic
appearance as well
differentiated, moderately differentiated, poorly differentiated, or
undifferentiated. In another
aspect, a pancreatic cancer that is to be treated is classified by microscopic
appearance with
37


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
respect to mitosis count (e.g., amount of cell division) or nuclear
pleiomorphism (e.g., change in
cells). hi another aspect, a pancreatic cancer that is to be treated is
classified by microscopic
appearance as being associated with areas of necrosis (e.g., areas of dying or
degenerating cells).
In one aspect, a pancreatic cancer that is to be treated is classified as
having an abnormal
karyotype, having an abnormal number of chromosomes, or having one or more
chromosomes
that are abnormal in appearance. In one aspect, a pancreatic cancer that is to
be treated is
classified as being aneuploid, triploid, tetraploid, or as having an altered
ploidy. In one aspect, a
pancreatic cancer that is to be treated is classified as having a chromosomal
translocation, or a
deletion or duplication of an entire chromosome, or a region of deletion,
duplication or
amplification of a portion of a chromosome.
A "cell proliferative disorder of the prostate" is a cell proliferative
disorder involving
cells of the prostate. In one aspect, cell proliferative disorders of the
prostate include all forms
of cell proliferative disorders affecting prostate cells. In one aspect, cell
proliferative disorders
of the prostate include prostate cancer, a precancer or precancerous condition
of the prostate,
benign growths or lesions of the prostate, and malignant growths or lesions of
the prostate, and
metastatic lesions in tissue and organs in the body other than the prostate.
In another aspect, cell
proliferative disorders of the prostate include hyperplasia, metaplasia, and
dysplasia of the
prostate.
A "cell proliferative disorder of the skin" is a cell proliferative disorder
involving cells of
the skin. In one aspect, cell proliferative disorders of the skin include all
forms of cell
proliferative disorders affecting skin cells. In one aspect, cell
proliferative disorders of the skin
include a precancer or precancerous condition of the skin, benign growths or
lesions of the skin,
melanoma, malignant melanoma and other malignant growths or lesions of the
skin, and
metastatic lesions in tissue and organs in the body other than the skin. In
another aspect, cell
proliferative disorders of the skin include hyperplasia, metaplasia, and
dysplasia of the skin.
A "cell proliferative disorder of the ovary" is a cell proliferative disorder
involving cells
of the ovary. In one aspect, cell proliferative disorders of the ovary include
all forms of cell
proliferative disorders affecting cells of the ovary. In one aspect, cell
proliferative disorders of
the ovary include a precancer or precancerous condition of the ovary, benign
growths or lesions
of the ovary, ovarian cancer, malignant growths or lesions of the ovary, and
metastatic lesions in
tissue and organs in the body other than the ovary. In another aspect, cell
proliferative disorders
of the skin include hyperplasia, metaplasia, and dysplasia of cells of the
ovary.
In one aspect, a cancer that is to be treated has been staged according to the
American
Joint Committee on Cancer (AJCC) TNM classification system, where the tumor
(T) has been
38


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
assigned a stage of TX, T1, Tlmic, Tis, Tla, Tlb, Tlc, T2, T3, T4, T4a, T4b,
T4c, or T4d; and
where the regional lymph nodes (I~ have been assigned a stage of NX, N0, Nl,
N2, N2a, N2b,
N3, N3a, N3b, or N3c; and where distant metastasis (M) has been assigned a
stage of MX, M0,
or M1. In another aspect, a cancer that is to be treated has been staged
according to an American
Joint Committee on Cancer (AJCC) classification as Stage I, Stage II, Stage
IIA, Stage IIB,
Stage III, Stage IIIA, Stage IIIB, Stage IIIC, or Stage IV. In another aspect,
a cancer that is to be
treated has been assigned a grade according to an AJCC classification as Grade
GX (e.g., grade
cannot be assessed), Grade 1, Grade 2, Grade 3 or Grade 4. In another aspect,
a cancer that is to
be treated has been staged according to an AJCC pathologic classification (pN)
of pNX, pNO,
PNO (I-), PNO (I+), PNO (mol-), PNO (mol+), PNl, PN1(mi), PNla, PNIb, PNlc,
pN2, pN2a,
pN2b, pN3, pN3a, pN3b, or pN3c.
In one aspect, a cancer that is to be treated includes a tumor that has been
determined to
be less than or equal to about 2 centimeters in diameter. In another aspect, a
cancer that is to be
treated includes a tumor that has been determined to be from about 2 to about
5 centimeters in
diameter. In another aspect, a cancer that is to be treated includes a tumor
that has been
determined to be greater than or equal to about 3 centimeters in diameter. In
another aspect, a
cancer that is to be treated includes a tumor that has been determined to be
greater than 5
centimeters in diameter. In another aspect, a cancer that is to be treated is
classified by
microscopic appearance as well differentiated, moderately differentiated,
poorly differentiated,
or undifferentiated. In another aspect, a cancer that is to be treated is
classified by microscopic
appearance with respect to mitosis count (e.g., amount of cell division) or
nuclear
pleiomorphism (e.g., change in cells). In another aspect, a cancer that is to
be treated is
classified by microscopic appearance as being associated with areas of
necrosis (e.g., areas of
dying or degenerating cells). In one aspect, a cancer that is to be treated is
classified as having
an abnormal karyotype, having an abnormal number of chromosomes, or having one
or more
chromosomes that are abnormal in appearance. In one aspect, a cancer that is
to be treated is
classified as being aneuploid, triploid, tetraploid, or as having an altered
ploidy. In one aspect, a
cancer that is to be treated is classified as having a chromosomal
translocation, or a deletion or
duplication of an entire chromosome, or a region of deletion, duplication or
amplification of a
portion of a chromosome.
In one aspect, a cancer that is to be treated is evaluated by DNA cytometry,
flow
cytometry, or image cytometry. In one aspect, a cancer that is to be treated
has been typed as
having 10%, 20%, 30%, 40%, 50%, 60%, 70%, SO%, or 90% of cells in the
synthesis stage of
39


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
cell division (e.g., in S phase of cell division). In one aspect, a cancer
that is to be treated has
been typed as having a low S-phase fraction or a high S-phase fraction.
As used herein, a "normal cell" is a cell that cannot be classified as part of
a "cell
proliferative disorder." In one aspect, a normal cell lacks unregulated or
abnormal growth, or
both, that can lead to the development of an unwanted condition or disease.
Preferably, a
normal cell possesses normally functioning cell cycle checkpoint control
mechanisms.
As used herein, "contacting a cell" refers to a condition in which a compound
or other
composition of matter is in direct contact with a cell, or is close enough to
induce a desired
biological effect in a cell.
As used herein, "treating" describes the management and care of a patient for
the
purpose of combating a disease, condition, or disorder and includes the
administration of a
combination of the present invention to prevent the onset of the symptoms or
complications,
alleviating the symptoms or complications, or eliminating the disease,
condition or disorder.
In one aspect, treating a cancer of the present invention results in a
decrease in number of
cancerous cells. Preferably, after treatment, number of cancerous cells is
reduced by 5% or
greater relative to number prior to treatment; more preferably, number of
cancerous cells is
reduced by 10% or greater; more preferably, reduced by 20% or greater; more
preferably,
reduced by 30% or greater; more preferably, reduced by 40% or greater; even
more preferably,
reduced by 50% or greater; and most preferably, reduced by greater than 75%.
Number of
cancerous cells may be measured by any reproducible means of measurement. In a
preferred
aspect, number of cancerous cells may be measured by counting cancerous cells
at a specified
magnification. In a preferred aspect, the specified magnification is 2x, 3x,
4x, Sx, l Ox, or 50x.
In another aspect, number of cancerous cells is measured by fluorescence
activated cell sorting
(FACS). In another aspect, number of cancerous cells is measured by
irmnunofluorescence
microscopy.
In one aspect, treating a cancer of the present invention results in a
reduction in size of a
tumor. A reduction in size of a tumor may also be referred to as "tumor
regression." Preferably,
after treatment, tumor size is reduced by 5% or greater relative to its size
prior to treatment;
more preferably, tumor size is reduced by 10% or greater; more preferably,
reduced by 20% or
greater; more preferably, reduced by 30% or greater; more preferably, reduced
by 40% or
greater; even more preferably, reduced by 50% or greater; and most preferably,
reduced by
greater than 75% or greater. Size of a tumor may be measured by any
reproducible means of
measurement. In a preferred aspect, size of a tumor may be measured as a
diameter of the
tumor.


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
In another aspect, treating a cancer of the present invention results in a
reduction in
tumor volume. Preferably, after treatment, tumor volume is reduced by 5% or
greater relative to
its size prior to treatment; more preferably, tumor volume is reduced by 10%
or greater; more
preferably, reduced by 20% or greater; more preferably, reduced by 30% or
greater; more
preferably, reduced by 40% or greater; even more preferably, reduced by 50% or
greater; and
most preferably, reduced by greater than 75% or greater. Tumor volume may be
measured by
any reproducible means of measurement.
In another aspect, treating a cancer of the present invention results in a
decrease in
number of tumors. Preferably, after treatment, tumor number is reduced by 5%
or greater
relative to number prior to treatment; more preferably, tumor number is
reduced by 10% or
greater; more preferably, reduced by 20% or greater; more preferably, reduced
by 30% or
greater; more preferably, reduced by 40% or greater; even more preferably,
reduced by 50% or
greater; and most preferably, reduced by greater than 75%. Number of tumors
may be measured
by any reproducible means of measurement. In a preferred aspect, number of
tumors may be
measured by counting tumors visible to the naked eye or at a specified
magnification. In a
preferred aspect, the specified magnification is 2x, 3x, 4x, Sx, 10x, or 50x.
hi another aspect, treating a cancer of the present invention results in a
decrease in
number of metastatic lesions in other tissues or organs distant from the
primary tumor site.
Preferably, after treatment, the number of metastatic lesions is reduced by 5%
or greater relative
to number prior to treatment; more preferably, the number of metastatic
lesions is reduced by
10% or greater; more preferably, reduced by 20% or greater; more preferably,
reduced by 30%
or greater; more preferably, reduced by 40% or greater; even more preferably,
reduced by 50%
or greater; and most preferably, reduced by greater than 75%. The number of
metastatic lesions
may be measured by any reproducible means of measurement. In a preferred
aspect, the number
of metastatic lesions may be measured by counting metastatic lesions visible
to the naked eye or
at a specified magnification. In a preferred aspect, the specified
magnification is 2x, 3x, 4x, Sx,
10x, or 50x.
In another aspect, treating a cancer of the present invention results in an
increase in
average survival time of a population of treated subjects in comparison to a
population receiving
Garner alone. Preferably, the average survival time is increased by more than
30 days; more
preferably, by more than 60 days; more preferably, by more than 90 days; and
most preferably,
by more than 120 days. An increase in average survival time of a population
may be measured
by any reproducible means. In a preferred aspect, an increase in average
survival time of a
population may be measured, for example, by calculating for a population the
average length of
41


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
surmval tollowmg initiation of treatment with an active compound. In another
preferred aspect,
an increase in average survival time of a population may also be measured, for
example, by
calculating for a population the average length of survival following
completion of a first round
of treatment with an active compound.
In another aspect, treating a cancer of the present invention results in an
increase in
average survival time of a population of treated subjects in comparison to a
population of
untreated subjects. Preferably, the average survival time is increased by more
than 30 days;
more preferably, by more than 60 days; more preferably, by more than 90 days;
and most
preferably, by more than 120 days. An increase in average survival time of a
population may be
measured by any reproducible means. In a preferred aspect, an increase in
average survival time
of a population may be measured, for example, by calculating for a population
the average
length of survival following initiation of treatment with an active compound.
In another
preferred aspect, an increase in average survival time of a population may
also be measured, for
example, by calculating for a population the average length of survival
following completion of
a first round of treatment with an active compound.
In another aspect, treating a cancer of the present invention results in
increase in average
survival time of a population of treated subjects in comparison to a
population receiving
monotherapy with a drug that is not a compound of the present invention, or a
pharmaceutically
acceptable salt, prodrug, metabolite, analog or derivative thereof.
Preferably, the average
survival tune is increased by more than 30 days; more preferably, by more than
60 days; more
preferably, by more than 90 days; and most preferably, by more than 120 days.
An increase in
average survival time of a population may be measured by any reproducible
means. In a
preferred aspect, an increase in average survival time of a population may be
measured, for
example, by calculating for a population the average length of survival
following initiation of
treatment with an active compound. In another preferred aspect, an increase in
average survival
time of a population may also be measured, for example, by calculating for a
population the
average length of survival following completion of a first round of treatment
with an active
compound.
In another aspect, treating a cancer of the present invention results in a
decrease in the
mortality rate of a population of treated subjects in comparison to a
population receiving carrier
alone. In another aspect, treating cancer results in a decrease in the
mortality rate of a
population of treated subjects in comparison to an untreated population. In a
further aspect,
treating cancer results a decrease in the mortality rate of a population of
treated subj ects in
comparison to a population receiving monotherapy with a drug that is not a
compound of the
42


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
present invention, or a pharmaceutically acceptable salt, prodrug, metabolite,
analog or
derivative thereof. Preferably, the mortality rate is decreased by more than
2%; more preferably,
by more than 5%; more preferably, by more than 10%; and most preferably, by
more than 25%.
In a preferred aspect, a decrease in the mortality rate of a population of
treated subjects may be
measured by any reproducible means. In another preferred aspect, a decrease in
the mortality
rate of a population may be measured, for example, by calculating for a
population the average
number of disease-related deaths per unit time following initiation of
treatment with an active
compound. In another preferred aspect, a decrease in the mortality rate of a
population may also
be measured, for example, by calculating for a population the average number
of disease-related
deaths per unit time following completion of a first round of treatment with
an active compound.
In another aspect, treating a cancer of the present invention results in a
decrease in tumor
growth rate. Preferably, after treatment, tumor growth rate is reduced by at
least 5% relative to
number prior to treatment; more preferably, tumor growth rate is reduced by at
least 10%; more
preferably, reduced by at least 20%; more preferably, reduced by at least 30%;
more preferably,
reduced by at least 40%; more preferably, reduced by at least 50%; even more
preferably,
reduced by at least 50%; and most preferably, reduced by at least 75%. Tumor
growth rate may
be measured by any reproducible means of measurement. In a preferred aspect,
tumor growth
rate is measured according to a change in ttunor diameter per unit time.
W another aspect, treating a cancer of the present invention results in a
decrease in tumor
regrowth. Preferably, after treatment, tumor regrowth is less than S%; more
preferably, tumor
regrowth is less than 10%; more preferably, less than 20%; more preferably,
less than 30%;
more preferably, less than 40%; more preferably, less than 50%; even more
preferably, less than
50%; and most preferably, less than 75%. Tumor regrowth may be measured by any
reproducible means of measurement. In a preferred aspect, tumor regrowth is
measured, for
example, by measuring an increase in the diameter of a tumor after a prior
tumor shrinkage that
followed treatment. In another preferred aspect, a decrease in tumor regrowth
is indicated by
failure of tumors to reoccur after treatment has stopped.
In another aspect, preventing cancer metastases results in a decrease in
number of
metastatic lesions in other tissues or organs distant from the primary tumor
site. Preferably, after
treatment, the number of metastatic lesions is reduced by 5% or greater
relative to number prior
to treatment; more preferably, the number of metastatic lesions is reduced by
10% or greater;
more preferably, reduced by 20% or greater; more preferably, reduced by 30% or
greater; more
preferably, reduced by 40% or greater; even more preferably, reduced by 50% or
greater; and
most preferably, reduced by greater than 75%. The number of metastatic lesions
may be
43


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
measured by any reproducible means of measurement. In a preferred aspect, the
number of
metastatic lesions may be measured by counting metastatic lesions visible to
the naked eye or at
a specified magnification. In a preferred aspect, the specified magnification
is 2x, 3x, 4x, Sx,
10x, or 50x.
In another aspect, treating or preventing a cell proliferative disorder of the
present
invention results in a reduction in the rate of cellular proliferation.
Preferably, after treatment,
the rate of cellular proliferation is reduced by at least 5%; more preferably,
by at least 10%;
more preferably, by at least 20%; more preferably, by at least 30%; more
preferably, by at least
40%; more preferably, by at least 50%; even more preferably, by at least 50%;
and most
preferably, by at least 75%. The rate of cellular proliferation may be
measured by any
reproducible means of measurement. In a preferred aspect, the rate of cellular
proliferation is
measured, for example, by measuring the number of dividing cells in a tissue
sample per unit
time.
In another aspect, treating or preventing a cell proliferative disorder of the
present
invention results in a reduction in the proportion of proliferating cells.
Preferably, after
treatment, the proportion of proliferating cells is reduced by at least 5%;
more preferably, by at
least 10%; more preferably, by at least 20%; more preferably, by at least 30%;
more preferably,
by at least 40%; more preferably, by at least 50%; even more preferably, by at
least 50%; and
most preferably, by at least 75%. The proportion of proliferating cells may be
measured by any
reproducible means of measurement. In a preferred aspect, the proportion of
proliferating cells
is measured, for example, by quantifying the number of dividing cells relative
to the number of
nondividing cells in a tissue sample. In another preferred aspect, the
proportion of proliferating
cells is equivalent to the mitotic index.
In another aspect, treating or preventing a cell proliferative disorder of the
present
invention results in a decrease in size of an area or zone of cellular
proliferation. Preferably,
after treatment, size of an area or zone of cellular proliferation is reduced
by at least 5% relative
to its size prior to treatment; more preferably, reduced by at least 10%; more
preferably, reduced
by at least 20%; more preferably, reduced by at least 30%; more preferably,
reduced by at least
40%; more preferably, reduced by at least 50%; even more preferably, reduced
by at least 50%;
and most preferably, reduced by at least 75%. Size of an area or zone of
cellular proliferation
may be measured by any reproducible means of measurement. In a preferred
aspect, size of an
area or zone of cellular proliferation may be measured as a diameter or width
of an area or zone
of cellular proliferation.
44


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
In another aspect, treating or preventing a cell proliferative disorder of the
present
invention results in a decrease in the number or proportion of cells having an
abnormal
appearance or morphology. Preferably, after treatment, the number of cells
having an abnormal
morphology is reduced by at least 5% relative to its size prior to treatment;
more preferably,
reduced by at least 10%; more preferably, reduced by at least 20%; more
preferably, reduced by
at least 30%; more preferably, reduced by at least 40%; more preferably,
reduced by at least
50%; even more preferably, reduced by at least 50%; and most preferably,
reduced by at least
75%. An abnormal cellular appearance or morphology may be measured by any
reproducible
means of measurement. In one aspect, an abnormal cellular morphology is
measured by
microscopy, e.g., using an inverted tissue culture microscope. In one aspect,
an abnormal
cellular morphology takes the form of nuclear pleiomorphism.
In one aspect, activating refers to placing a composition of matter (e.g.,
protein or nucleic
acid) in a state suitable for carrying out a desired biological function. In
one aspect, a
composition of matter capable of being activated also has an unactivated
state. In one aspect, an
activated composition of matter may have an inhibitory or stimulatory
biological function, or
both.
In one aspect, elevation refers to an increase in a desired biological
activity of a
composition of matter (e.g., a protein or a nucleic acid). In one aspect,
elevation may occur
through an increase in concentration of a composition of matter.
As used herein, the term "selectively" means tending to occur at a higher
frequency in
one population than in another population. In one aspect, the compared
populations are cell
populations. In a preferred aspect, a compound of the present invention, or a
pharmaceutically
acceptable salt, prodrug, metabolite, analog or derivative thereof, acts
selectively on a cancer or
precancerous cell but not on a normal cell. In another preferred aspect, a
compound of the
present invention, or a pharmaceutically acceptable salt, prodrug, metabolite,
analog or
derivative thereof, acts selectively to modulate one molecular target (e.g.,
E2F-1) but does not
significantly modulate another molecular target (e.g., Protein Kinase C). In
another preferred
aspect, the invention provides a method for selectively inhibiting the
activity of an enzyme, such
as a kinase. Preferably, an event occurs selectively in population A relative
to population B if it
occurs greater than two times more frequently in population A as compared to
population B.
More preferably, an event occurs selectively if it occurs greater than five
times more frequently
in population A. More preferably, an event occurs selectively if it occurs
greater than ten times
more frequently in population A; more preferably, greater than fifty times;
even more
preferably, greater than 100 times; and most preferably, greater than 1000
times more frequently


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
in population A as compared to population B. For example, cell death would be
said to occur
selectively in cancer cells if it occurred greater than twice as frequently in
cancer cells as
compared to normal cells.
In a preferred aspect, a compound of the present invention or a
pharmaceutically
acceptable salt, prodrug, metabolite, analog or derivative thereof, modulates
the activity of a
molecular target (e.g., E2F-1). In one aspect, modulating refers to
stimulating or inhibiting an
activity of a molecular target. Preferably, a compound of the present
invention modulates the
activity of a molecular target if it stimulates or inhibits the activity of
the molecular target by at
least 10% relative to the activity of the molecular target under the same
conditions but lacking
only the presence of said compound. More preferably, a compound of the present
invention
modulates the activity of a molecular target if it stimulates or inhibits the
activity of the
molecular target by at least 25%, at least 2-fold, at least 5-fold, at least
10-fold, at least 20-fold,
at least 50-fold, at least 100-fold relative to the activity of the molecular
target under the same
conditions but lacking only the presence of said compound. The activity of a
molecular target
may be measured by any reproducible means. The activity of a molecular target
may be
measured in vitro or in vivo. For example, the activity of a molecular target
may be measured in
vitro by an enzymatic activity assay or a DNA binding assay, or the activity
of a molecular
target may be measured in vivo by assaying for expression of a reporter gene.
In one aspect, a compound of the present invention, or a pharmaceutically
acceptable
salt, prodrug, metabolite, analog or derivative thereof, does not
significantly modulate the
activity of a molecular target if the addition of the compound stimulates or
inhibits the activity
of the molecular target by less than 10% relative to the activity of the
molecular target under the
same conditions but lacking only the presence of said compound.
As used herein, the term "isozyme selective" means preferential inhibition or
stimulation
of a first isoform of an enzyme in comparison to a second isoform of an enzyme
(e.g.,
preferential inhibition or stimulation of a kinase isozyme alpha in comparison
to a kinase
isozyme beta). Preferably, a compound of the present invention demonstrates a
minimum of a
four fold differential, preferably a ten fold differential, more preferably a
fifty fold differential,
in the dosage required to achieve a biological effect. Preferably, a compound
of the present
invention demonstrates this differential across the range of inhibition, and
the differential is
exemplified at the ICSO, i.e., a 50% inhibition, for a molecular target of
interest.
In a preferred embodiment, administering (3-lapachone, or a pharmaceutically
acceptable
salt, prodrug, metabolite, analog or derivative thereof, to a cell or a
subject in need thereof
results in modulation (i.e., stimulation or inhibition) of an activity of a
member of the E2F
46


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
family of transcription factors (e.g., E2F-1, E2F-2, or E2F-3). As used
herein, an activity of a
member of the E2F family of transcription factors refers to any biological
function or activity
that is carried out by an E2F family member. For example, a function of E2F-1
includes binding
of E2F-1 to its cognate DNA sequences. Other functions of E2F-1 include
migrating to the cell
nucleus and activating transcription.
In one aspect, treating cancer or a cell proliferative disorder results in
cell death, and
preferably, cell death results in a decrease of at least 10% in ntunber of
cells in a population.
More preferably, cell death means a decrease of at least 20%; more preferably,
a decrease of at
least 30%; more preferably, a decrease of at least 40%; more preferably, a
decrease of at least
50%; most preferably, a decrease of at least 75%. Number of cells in a
population may be
measured by any reproducible means. In one aspect, number of cells in a
population is
measured by fluorescence activated cell sorting (FACE). In another aspect,
number of cells in a
population is measured by immunofluorescence microscopy. In another aspect,
number of cells
in a population is measured by light microscopy. In another aspect, methods of
measuring cell
death are as shown in Li et al., (2003) Proc Natl Acad Sci USA. 100(5): 2674-
8. In an aspect,
cell death occurs by apoptosis.
In a preferred aspect, an effective amount of a compound of the present
invention, or a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof is not
significantly cytotoxic to normal cells. A therapeutically effective amount of
a compound is not
significantly cytotoxic to normal cells if administration of the compound in a
therapeutically
effective amount does not induce cell death in greater than 10% of normal
cells. A
therapeutically effective amount of a compound does not significantly affect
the viability of
normal cells if administration of the compound in a therapeutically effective
amount does not
induce cell death in greater than 10% of normal cells. In an aspect, cell
death occurs by
apoptosis.
In one aspect, contacting a cell with a combination of the present invention,
or a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof, induces or
activates cell death selectively in cancer cells. Preferably, administering to
a subject in need
thereof a combination of the present invention, or a pharmaceutically
acceptable salt, prodrug,
metabolite, analog or derivative thereof, induces or activates cell death
selectively in cancer
cells. In another aspect, contacting a cell with a combination of the present
invention, or a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof, induces cell
death selectively in one or more cells affected by a cell proliferative
disorder. Preferably,
administering to a subject in need thereof a combination of the present
invention, or a
47


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof, induces cell
death selectively in one or more cells affected by a cell proliferative
disorder. In a preferred
aspect, the present invention relates to a method of treating or preventing
cancer by
administering a combinationof the present invention, or a pharmaceutically
acceptable salt,
prodrug, metabolite, analog or derivative thereof to a subject in need
thereof, where
administration of the combination of the present invention, or a
pharmaceutically acceptable salt,
prodrug, metabolite, analog or derivative thereof results in one or more of
the following:
accumulation of cells in G1 and/or S phase of the cell cycle, cytotoxicity via
cell death in cancer
cells without a significant amount of cell death in normal cells, antitumor
activity in animals
with a therapeutic index of at least 2, and activation of a cell cycle
checkpoint. As used herein,
"therapeutic index" is the maximum tolerated dose divided by the efficacious
dose.
In one aspect, stimulation of unscheduled expression of a checkpoint molecule
by ~3-
lapachone, or a pharmaceutically acceptable salt, metabolite, analog or
derivative thereof,
triggers cell death in cells with defective checkpoints, a hallmark of cancer
and pre-cancer cells.
In one aspect, contacting a cell with (3-lapachone, or a pharmaceutically
acceptable salt,
metabolite, analog or derivative thereof, stimulates unscheduled expression of
the checkpoint
molecule E2F.
In one aspect, contacting a cell with (3-lapachone, or a pharmaceutically
acceptable salt,
metabolite, analog or derivative thereof, results in activation of an E2F
checkpoint pathway.
Preferably, administering to a subject in need thereof (3-lapachone, or a
pharmaceutically
acceptable salt, metabolite, analog or derivative thereof, results in
activation of an E2F
checkpoint pathway. In a preferred aspect, E2F pathway activity is increased
by more than
10%; more than 25%; more than 50%; more than 2-fold; more than 5-fold; and
most preferably,
by more than 10-fold. In a preferred aspect, E2F activity is increased by more
than 10%; more
than 25%; more than 50%; more than 2-fold; more than 5-fold; and most
preferably, by more
than 10-fold. Methods of measuring induction of E2F activity and elevation of
E2F levels are as
shown in Li et al., (2003) Proc Natl Acad Sci USA. 100(5): 2674-8.
In one aspect, contacting a cell with ~-lapachone, or a pharmaceutically
acceptable salt,
metabolite, analog or derivative thereof, results in elevation of an E2F
transcription factor.
Preferably, administering to a subject in need thereof ~i-lapachone, or a
pharmaceutically
acceptable salt, metabolite, analog or derivative thereof, results in
elevation of an E2F
transcription factor.
In one aspect, contacting a cell with (3-lapachone, or a pharmaceutically
acceptable salt,
metabolite, analog or derivative thereof, results in elevation of an E2F
transcription factor
48


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
selectively in cancer cells but not in normal cells. Preferably, administering
to a subject in need
thereof (3-lapachone, or a pharmaceutically acceptable salt, metabolite,
analog or derivative
thereof, results in elevation of an E2F transcription factor selectively in
cancer cells but not in
normal cells.
In one aspect, contacting a cell with ~i-lapachone, or a pharmaceutically
acceptable salt,
metabolite, analog or derivative thereof, stimulates unscheduled activation of
an E2F
transcription factor. Preferably, administering to a subject in need thereof
(3-lapachone, or a
pharmaceutically acceptable salt, metabolite, analog or derivative thereof,
stimulates
unscheduled activation of an E2F transcription factor.
In one aspect, contacting a cell with (i-lapachone, or a pharmaceutically
acceptable salt,
metabolite, analog or derivative thereof, stimulates unscheduled activation of
an E2F
transcription factor selectively in cancer cells but not in normal cells.
Preferably, administering
to a subject in need thereof (3-lapachone, or a pharmaceutically acceptable
salt, metabolite,
analog or derivative thereof, stimulates unscheduled activation of an E2F
transcription factor
selectively in cancer cells but not in normal cells.
In normal cells with their intact regulatory mechanisms, imposed expression of
a
checkpoint molecule (e.g., as induced by contacting a cell with (3-lapachone,
or a
pharmaceutically acceptable salt, metabolite, analog or derivative thereof )
results in an
expression pattern that is not reported to be of substantial consequence. In
contrast, cancer and
pre-cancer cells have defective mechanisms, which result in unchecked or
persistent expression,
or both, of unscheduled checkpoint molecules, e.g., E2F, leading to selective
cell death in cancer
and pre-cancer cells. The present invention includes and provides for the
unchecked or
persistent expression, or both, of unscheduled checkpoint molecules by the
administration of (3-
lapachone, or a pharmaceutically acceptable salt, metabolite, analog or
derivative thereof.
In one aspect, contacting a cell with ~-lapachone, or a pharmaceutically
acceptable salt,
metabolite, analog or derivative thereof, results in activation of one or more
cell cycle
checkpoints. Preferably, administering to a subject in need thereof (3-
lapachone, or a
pharmaceutically acceptable salt, metabolite, analog or derivative thereof,
results in activation of
one or more cell cycle checkpoints.
In one aspect, contacting a cell with ~i-lapachone, or a pharmaceutically
acceptable salt,
metabolite, analog or derivative thereof, results in activation of one or more
cell cycle
checkpoint pathways. Preferably, administering to a subject in need thereof ~-
lapachone, or a
pharmaceutically acceptable salt, metabolite, analog or derivative thereof,
results in activation of
one or more cell cycle checkpoint pathways.
49


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
In one aspect, contacting a cell with (3-lapachone, or a pharmaceutically
acceptable salt,
metabolite, analog or derivative thereof, results in activation of one or more
cell cycle
checkpoint regulators. Preferably, administering to a subject in need thereof
(3-lapachone, or a
pharmaceutically acceptable salt, metabolite, analog or derivative thereof,
results in activation of
one or more cell cycle checkpoint regulators.
In one aspect, contacting a cell with (3-lapachone, or a pharmaceutically
acceptable salt,
metabolite, analog or derivative thereof, induces or activates cell death
selectively in cancer
cells. Preferably, administering to a subject in need thereof ~3-lapachone, or
a pharmaceutically
acceptable salt, metabolite, analog or derivative thereof, induces or
activates cell death
selectively in cancer cells. In another aspect, contacting a cell with (3-
lapachone, or a
pharmaceutically acceptable salt, metabolite, analog or derivative thereof,
induces cell death
selectively in one or more cells affected by a cell proliferative disorder.
Preferably,
administering to a subject in need thereof (3-lapachone, or a pharmaceutically
acceptable salt,
metabolite, analog or derivative thereof, induces cell death selectively in
one or more cells
affected by a cell proliferative disorder.
One skilled in the art may refer to general reference texts for detailed
descriptions of
known techniques discussed herein or equivalent techniques. These texts
include Ausubel et al.,
Curs°ent Pf°otocols in Molecular Biology, John Wiley and Sons,
Inc. (2005); Sambrook et al.,
Molecular Clonif2g, A Laboratory Mafaual (3d ed.), Cold Spring Harbor Press,
Cold Spring
Harbor, New York (2000); Coligan et al., Currefat Protocols in Immuyaology,
John Wiley &
Sons, N.Y.; Enna et al., Cur3°ent Protocols ih Pharmacology, John Wiley
& Sons, N.Y.; Fingl et
al., The Pharmacological Basis of Therapeutics (1975), Remifagton's
Pharmaceutical Sciefaces,
Mack Publishing Co., Easton, PA, 18th edition (1990). These texts can, of
course, also be
referred to in making or using an aspect of the invention.
A compound of the present invention, or a pharmaceutically acceptable salt,
prodrug,
metabolite, analog or derivative thereof, can be incorporated into
pharmaceutical compositions
suitable for administration. Such compositions typically comprise the compound
(i.e. including
the active compound), and a pharmaceutically acceptable excipient or carrier.
As used herein,
"pharmaceutically acceptable excipient" or "pharmaceutically acceptable
Garner" is intended to
include any and all solvents, dispersion media, coatings, antibacterial and
antifungal agents,
isotonic and absorption delaying agents, and the like, compatible with
pharmaceutical
administration. Suitable carriers are described in the most recent edition of
Remington's
Pharmaceutical Sciences, a standard reference text in the field. Preferred
examples of such
carriers or diluents include, but are not limited to, water, saline, ringer's
solutions, dextrose


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
solution, and 5% human serum albumin. Pharmaceutically acceptable carriers
include solid
carriers such as lactose, terra albs, sucrose, talc, gelatin, agar, pectin,
acacia, magnesium
stearate, stearic acid and the like. Exemplary liquid earners include syrup,
peanut oil, olive oil,
water and the like. Similarly, the carrier or diluent may include time-delay
material known in the
art, such as glyceryl monostearate or glyceryl distearate, alone or with a
wax, ethylcellulose,
hydroxypropylmethylcellulose, methylmethacrylate or the like. Other fillers,
excipients,
flavorants, and other additives such as are known in the art may also be
included in a
pharmaceutical composition according to this invention. Liposomes and non-
aqueous vehicles
such as fixed oils may also be used. The use of such media and agents for
pharmaceutically
active substances is well known in the art. Except insofar as any conventional
media or agent is
incompatible with the active compound, use thereof in the compositions is
contemplated.
Supplementary active compounds can also be incorporated into the compositions.
The pharmaceutical compositions of this invention which are provided as part
of the
combination therapies may exist in the dosage form as a solid, semi-solid, or
liquid such as, e.g.,
suspensions, aerosols or the like. Preferably the compositions are
administered in unit dosage
forms suitable for single administration of precise dosage amounts. The
compositions may also
include, depending on the formulation desired, pharmaceutically-acceptable,
nontoxic earners or
diluents, which are defined as vehicles commonly used to formulate
pharmaceutical
compositions for animal or human administration. The diluent is selected so as
not to affect the
biological activity of the combination. Examples of such diluents are
distilled water,
physiological saline, Ringer's solution, dextrose solution, and Hank's
solution. A preferred
carrier for the solubilization of (3-lapachone is hydroxypropyl beta
cyclodextrin, a water
solubilizing carrier molecule. Other water-solubilizing agents for combining
with (3-lapachone
and/or an S-phase compound, such as Poloxamer, Povidone K17, Povidone K12,
Tween 80,
ethanol, Cremophor/ethanol, polyethylene glycol 400, propylene glycol and
Trappsol, are
contemplated. Furthermore, the invention is not limited to water-solubilizing
agents, and oil-
based solubilizing agents such as lipiodol and peanut oil, may also be used.
In addition, the pharmaceutical composition or formulation may also include
other
carriers, adjuvants, or nontoxic, nontherapeutic, nonimmunogenic stabilizers
and the like.
Effective amounts of such diluent or carrier will be those amounts which are
effective to obtain a
pharmaceutically acceptable formulation in terms of solubility of components,
or biological
activity, and the like. Liposome formulations, are also contemplated by the
present invention,
and have been described See, e.g., U.S. Pat. No. 5,424,073.
51


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
For the purposes of the present invention, the Gl/S phase drugs or compounds,
or
derivatives or analogs thereof, and the S phase drugs or compounds, or
derivatives or analogs
thereof, described herein include their pharmacologically acceptable salts,
preferably sodium;
analogs containing halogen substitutions, preferably chlorine or fluorine;
analogs containing
ammonium or substituted ammonium salts, preferably secondary or tertiary
ammonium salts;
analogs containing alkyl, alkenyl, aryl or their alkyl, alkenyl, aryl, halo,
alkoxy, alkenyloxy
substituted derivatives, preferably methyl, methoxy, ethoxy, or phenylacetate;
and natural
analogs such as naphthyl acetate. Further, the Gl/S phase compounds or
derivatives or analogs
thereof, and the S phase compounds or derivatives or analogs thereof,
described herein may be
conjugated to a water-soluble polymer or may be derivatized with water-soluble
chelating agents
or radionuclides. Examples of water soluble polymers are, but not limited to:
polyglutamic acid
polymer, copolymers with polycaprolactone, polyglycolic acid, polyactic acid,
polyacrylic acid,
poly (2-hydroxyethyl 1-glutamine), carboxymethyl dextran, hyaluronic acid,
human senun
albumin, polyalginic acid or a combination thereof. Examples of water-soluble
chelating agents
are, but not limited to: DIPA (diethylenetriaminepentaacetic acid), EDTA,
DTTP, DOTA or
their water-soluble salts, etc. Examples of radionuclides include, but not
limited to: 11 lIn, 9°Y,
166H~' 6aGa~ 99m.j,C, and the like.
Due to the water insolubility of [3-lapachone, pharmaceutical Garners or
solubilizing
agents may be used to provide sufficient quantities of ~i-lapachone for use in
the treatment
methods of the present invention. See, e.g., U.S. Patent Publication
20030091639 to Jiang et al.,
and U.S. Patent Publication 20040001871 to Boothman et al. This publication
describes the use
of complexing agents such as cyclodextrins, including hydroxypropyl beta-
cyclodextrin
(HPBCD), to permit the solubilization of (3-lapachone at levels sufficient for
administration. See
also U.S. Patent Publication 20040001871 to Boothman et al. In an embodiment,
the G1/S
phase drug, or a derivative or analog thereof, is administered with a
pharmaceutically acceptable
water solubilizing carrier molecule selected from the group consisting of
Poloxamer, Povidone
K17, Povidone K12, Tween 80, ethanol, Cremophor/ethanol, polyethylene glycol
(PEG) 400,
propylene glycol, Trappsol, alpha-cyclodextrin or derivatives or analogs
thereof, beta-
cyclodextrin or derivatives or analogs thereof, and gamma-cyclodextrin or
derivatives or analogs
thereof.
In one aspect, a compound of the present invention, or a pharmaceutically
acceptable
salt, prodrug, metabolite, analog or derivative thereof, is administered in a
suitable dosage form
prepared by combining a therapeutically effective amount (e.g., an efficacious
level sufficient to
achieve the desired therapeutic effect through inhibition of tumor growth,
killing of tumor cells,
52


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
treatment or prevention of cell proliferative disorders, etc.) of a compound
of the present
invention, or a pharmaceutically acceptable salt, prodrug, metabolite, analog
or derivative
thereof, (as an active ingredient) with standard pharmaceutical carriers or
diluents according to
conventional procedures (a. e., by producing a pharmaceutical composition of
the invention).
These procedures may involve mixing, granulating, and compressing or
dissolving the
ingredients as appropriate to attain the desired preparation.
A pharmaceutical composition of the invention is formulated to be compatible
with its
intended route of administration. Examples of routes of administration include
parenteral, e.g.,
intravenous, intradermal, subcutaneous, oral (e.g." inhalation), transdermal
(topical), and
transmucosal administration. Although intravenous administration is preferred
as discussed
above, the invention is not intended to be limited in this respect, and the
compounds can be
administered by any means known in the art. Such modes include oral, rectal,
nasal, topical
(including buccal and sublingual) or parenteral (including subcutaneous,
intramuscular,
intravenous and intradermal) administration. For ease of administration and
comfort to the
patient, oral administration is generally preferred. However, oral
administration may require the
administration of a higher dose than intravenous administration. The skilled
artisan can
determine which form of administration is best in a particular case, balancing
dose needed
versus the number of times per month administration is necessary.
Solutions or suspensions used for parenteral, intradermal, or subcutaneous
application
can include the following components: a sterile diluent such as water for
injection, saline
solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or
other synthetic solvents;
antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants
such as ascorbic
acid or sodium bisulfate; chelating agents such as ethylenediaminetetraacetic
acid; buffers such
as acetates, citrates or phosphates, and agents for the adjustment of tonicity
such as sodium
chloride or dextrose. The pH can be adjusted with acids or bases, such as
hydrochloric acid or
sodium hydroxide. The parenteral preparation can be enclosed in ampoules,
disposable syringes
or multiple dose vials made of glass or plastic.
A compound or pharmaceutical composition of the invention can be administered
to a
subject in many of the well-known methods currently used for chemotherapeutic
treatment. For
example, for treatment of cancers, a compound of the invention may be injected
directly into
tumors, inj ected into the blood stream or body cavities or taken orally or
applied through the
skin with patches. The dose chosen should be sufficient to constitute
effective treatment but not
so high as to cause unacceptable side effects. The state of the disease
condition (e.g., cancer,
53


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
precancer, and the like) and the health of the patient should preferably be
closely monitored
during and for a reasonable period after treatment.
The term "therapeutically effective amount," as used herein, refers to an
amount of a
pharmaceutical agent to treat, ameliorate, or prevent an identified disease or
condition, or to
exhibit a detectable therapeutic or inhibitory effect. The effect can be
detected by any assay
method known in the art. The precise effective amount for a subject will
depend upon the
subject's body weight, size, and health; the nature and extent of the
condition; and the
therapeutic or combination of therapeutics selected for administration.
Therapeutically effective
amounts for a given situation can be determined by routine experimentation
that is within the
skill and judgment of the clinician. In a preferred aspect, the disease or
condition to be treated is
cancer. In another aspect, the disease or condition to be treated is a cell
proliferative disorder.
For any compound, the therapeutically effective amount can be estimated
initially either
in cell culture assays, e.g., of neoplastic cells, or in animal models,
usually rats, mice, rabbits,
dogs, or pigs. The animal model may also be used to determine the appropriate
concentration
range and route of administration. Such information can then be used to
determine useful doses
and routes for administration in humans. Therapeutic/prophylactic efficacy and
toxicity may be
determined by standard pharmaceutical procedures in cell cultures or
experimental animals, e.g.,
EDSO (the dose therapeutically effective in 50% of the population) and LDso
(the dose lethal to
50% of the population). The dose ratio between therapeutic and toxic effects
is the therapeutic
index, and it can be expressed as the ratio, EDSO/LDSO. Pharmaceutical
compositions that exhibit
large therapeutic indices are preferred. The dosage may vary within this range
depending upon
the dosage form employed, sensitivity of the patient, and the route of
administration.
Dosage and administration are adjusted to provide sufficient levels of the
active agents)
or to maintain the desired effect. Factors which may be taken into accomit
include the severity
of the disease state, general health of the subject, age, weight, and gender
of the subject, diet,
time and frequency of administration, drug combination(s), reaction
sensitivities, and
tolerance/response to therapy. Long-acting pharmaceutical compositions may be
administered
every 3 to 4 days, every week, or once every two weeks depending on half life
and clearance
rate of the particular formulation.
The pharmaceutical compositions containing active compounds of the present
invention
may be manufactured in a manner that is generally known, e.g., by means of
conventional
mixing, dissolving, granulating, dragee-making, levigating, emulsifying,
encapsulating,
entrapping, or lyophilizing processes. Pharmaceutical compositions may be
formulated in a
conventional manner using one or more pharmaceutically acceptable carriers
comprising
54


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
excipients and/or auxiliaries that facilitate processing of the active
compounds into preparations
that can be used pharmaceutically. Of course, the appropriate formulation is
dependent upon the
route of administration chosen.
Pharmaceutical compositions suitable for injectable use include sterile
aqueous solutions
(where water soluble) or dispersions and sterile powders for the
extemporaneous preparation of
sterile injectable solutions or dispersion. For intravenous administration,
suitable carriers
include physiological saline, bacteriostatic water, Cremophor ELTM (BASF,
Parsippany, N.J.) or
phosphate buffered saline (PBS). In all cases, the composition must be sterile
and should be
fluid to the extent that easy syringeability exists. It must be stable under
the conditions of
manufacture and storage and must be preserved against the contaminating action
of
microorganisms such as bacteria and fungi. The carrier can be a solvent or
dispersion medium
containing, for example, water, ethanol, polyol (for example, glycerol,
propylene glycol, and
liquid polyethylene glycol, and the like), and suitable mixtures thereof. The
proper fluidity can
be maintained, for example, by the use of a coating such as lecithin, by the
maintenance of the
required particle size in the case of dispersion and by the use of
surfactants. Prevention of the
action of microorganisms can be achieved by various antibacterial and
antifungal agents, for
example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the
like. In many
cases, it will be preferable to include isotonic agents, for example, sugars,
polyalcohols such as
manitol, sorbitol, sodium chloride in the composition. Prolonged absorption of
the injectable
compositions can be brought about by including in the composition an agent
which delays
absorption, for example, aluminum monostearate and gelatin.
Sterile injectable solutions can be prepared by incorporating the active
compound in the
required amount in an appropriate solvent with one or a combination of
ingredients enumerated
above, as required, followed by filtered sterilization. Generally, dispersions
are prepared by
incorporating the active compound into a sterile vehicle that contains a basic
dispersion medium
and the required other ingredients from those enumerated above. In the case of
sterile powders
for the preparation of sterile injectable solutions, methods of preparation
are vacuum drying and
freeze-drying that yields a powder of the active ingredient plus any
additional desired ingredient
from a previously sterile-filtered solution thereof.
Oral compositions generally include an inert diluent or an edible
pharmaceutically
acceptable Garner. They can be enclosed in gelatin capsules or compressed into
tablets. For the
purpose of oral therapeutic administration, the active compound can be
incorporated with
excipients and used in the form of tablets, troches, or capsules. Oral
compositions can also be
prepared using a fluid carrier for use as a mouthwash, wherein the compound in
the fluid carrier


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
is applied orally and swished and expectorated or swallowed. Pharmaceutically
compatible
binding agents, and/or adjuvant materials can be included as part of the
composition. The
tablets, pills, capsules, troches and the like can contain any of the
following ingredients, or
compounds of a similar nature: a binder such as microcrystalline cellulose,
gum tragacanth or
gelatin; an excipient such as starch or lactose, a disintegrating agent such
as alginic acid,
Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes;
a glidant such as
colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or
a flavoring agent
such as peppermint, methyl salicylate, or orange flavoring.
For administration by inhalation, the compounds are delivered in the form of
an aerosol
spray from pressured container or dispenser, which contains a suitable
propellant, e.g., a gas
such as carbon dioxide, or a nebulizer.
Systemic administration can also be by transmucosal or transdermal means. For
transmucosal or transdermal administration, penetrants appropriate to the
barrier to be permeated
are used in the formulation. Such penetrants are generally known in the art,
and include, for
example, for transmucosal administration, detergents, bile salts, and fusidic
acid derivatives.
Transmucosal administration can be accomplished through the use of nasal
sprays or
suppositories. For transdermal administration, the active compounds are
formulated into
ointments, salves, gels, or creams as generally known in the art.
In one aspect, the active compounds are prepared with pharmaceutically
acceptable
carriers that will protect the compound against rapid elimination from the
body, such as a
controlled release formulation, including implants and microencapsulated
delivery systems.
Biodegradable, biocompatible polymers can be used, such as ethylene vinyl
acetate,
polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic
acid. Methods for
preparation of such formulations will be apparent to those skilled in the art.
The materials can
also be obtained commercially from Alza Corporation and Nova Pharmaceuticals,
Inc.
Liposomal suspensions (including liposomes targeted to infected cells with
monoclonal
antibodies to viral antigens) can also be used as pharmaceutically acceptable
carriers. These can
be prepared according to methods known to those skilled in the art, for
example, as described in
U.S. Pat. No. 4,522,811.
It is especially advantageous to formulate oral or parenteral compositions in
dosage unit
form for ease of administration and uniformity of dosage. Dosage unit form as
used herein
refers to physically discrete units suited as unitary dosages for the subject
to be treated; each unit
containing a predetermined quantity of active compound calculated to produce
the desired
therapeutic effect in association with the required pharmaceutical carrier.
The specification for
56


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
the dosage unit forms of the invention are dictated by and directly dependent
on the unique
characteristics of the active compound and the particular therapeutic effect
to be achieved.
In therapeutic applications, the dosages of the pharmaceutical compositions
used in
accordance with the invention vary depending on the agent, the age, weight,
and clinical
condition of the recipient patient, and the experience and judgment of the
clinician or
practitioner administering the therapy, among other factors affecting the
selected dosage.
Generally, the dose should be sufficient to result in slowing, and preferably
regressing, the
growth of the tumors and also preferably causing complete regression of the
cancer. Dosages
can range from about 0.01 mg/kg per day to about 3000 mg/kg per day. In
preferred aspects,
dosages can range from about 1 mg/kg per day to about 1000 mg/kg per day. In
an aspect, the
dose will be in the range of about 0.1 mg/day to about 70 g/day; about 0.1
mg/day to about 25
g/day; about 0.1 mg/day to about 10 g/day; about 0.1 mg to about 3g/day; or
about 0.1 mg to
about 1 g/day, in single, divided, or continuous doses (which dose may be
adjusted for the
patient's weight in kg, body surface area in ma, and age in years). An
effective amount of a
pharmaceutical agent is that which provides an objectively identifiable
improvement as noted by
the clinician or other qualified observer. For example, regression of a tumor
in a patient may be
measured with reference to the diameter of a tumor. Decrease in the diameter
of a tumor
indicates regression. Regression is also indicated by failure of tumors to
reoccur after treatment
has stopped. As used herein, the term "dosage effective manner" refers to
amount of an active
compound to produce the desired biological effect in a subject or cell.
The pharmaceutical compositions can be included in a container, pack, or
dispenser
together with instructions for administration.
The S phase compound, such as an antimetabolite drug, may be administered in
any
manner found appropriate by a clinician in generally accepted efficacious dose
ranges, such as
those described in the Physician's DeskRefe~eface, 59th Edition, Thomson PDR
(2005)("PDR").
In general, the S phase drug or compound, such as gemcitabine, is administered
intravenously at
dosages from about 10 mg/m~ to about 10,000 mg/ma, preferably from about 100
mg/m2to about
2000 mg/m2, and most preferably about 500 to about 1500 mg/m2. In an
embodiment, the S
phase drug is administered intravenously at a dosage from approximately 100
mg/m~' to about
2000 mg/m2. In an embodiment, the S phase drug is administered intravenously
at a dosage of
approximately 1000 mg/m2. Dosage can be repeated, e.g., once weekly,
preferably for about 1
to 6 weeks. It is preferred that dosages be administered over a time period of
about 30 minutes
to about 6 hours, and typically over a period of about 3 hours.
57


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
The S phase drug, such as an antimetabolite drug, will be administered in a
similar
regimen with a Gl/S phase drug, such as (3-lapachone or a derivative or analog
thereof, although
the amounts will preferably be reduced from that normally administered. It is
preferred, for
example, that the S-phase drug be administered at the same time or after the
[3-lapachone has
administered to the patient. When the S-phase drug is administered after the
(3-lapachone, the S-
phase drug is advantageously administered about 24 hours after the (3-
lapachone has been
administered.
The combination therapy agents described herein may be administered singly and
sequentially, or in a cocktail or combination containing both agents or one of
the agents with
other therapeutic agents, including but not limited to, immunosuppressive
agents, potentiators
and side-effect relieving agents. As aforesaid, the therapeutic combination,
if administered
sequentially, may be more effective when the G1/S phase drug component (e.g.,
(3-lapachone) is
administered prior to the S phase drug, e.g., gemcitabine. For example, a dose
of the Gl/S phase
drug component (e.g., (3-lapachone) is administered at least one hour (more
preferably at least 2
hours, 4 hours, 8 hours, 12 hours, or 24 hours) prior to administration of a
dose of the S phase
drug, e.g., gemcitabine. In another embodiment, a dose of the Gl/S phase drug
component (e.g.,
(3-lapachone) is administered at least one hour (more preferably at least 2
hours, 4 hours, 8
hours, 12 hours, or 24 hours) following administration of a dose of the S
phase drug, e.g.,
gemcitabine. The therapeutic agents will preferably be administered
intravenously or otherwise
systemically by injection intramuscularly, subcutaneously, intrathecally or
intraperitoneally. In
an embodiment, the S phase drug is administered simultaneously with or
following
administration of the Gl/S phase drug. In another embodiment, the S phase drug
is administered
following administration of the Gl/S phase drug. In another embodiment, the S
drug is
administered within 24 hours after the G1/S phase drug is administered.
The other component of the combination therapy for combination with the S
phase drug
or compound is the G1/S phase drug, which is preferably (3-lapachone or a
derivative or analog
thereof.
(3-lapachone has been shown to have a variety of pharmacological effects. ~-
lapachone
has been shown to be a DNA repair inhibitor which sensitizes cells to DNA
damaging agents
(Boorstein, R.J., et al., (1984) Biochem. Bioplays. Res. Commun., 118:828-834;
Boothtnan, D.A.,
et al., (1989) J. Cancef° Res., 49:605-612). (3-lapachone is generally
well-tolerated in dogs, rats,
and mice.
The present invention provides a method of treating cancer or a precancerous
condition
or preventing cancer in a subject, the method comprising administering to the
subject a
58


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
therapeutically effective amount of a pharmaceutical composition comprising (3-
lapachone, or a
derivative or analog thereof, or pharmaceutically acceptable salt thereof, or
a metabolite thereof,
and a pharmaceutically acceptable carrier such that the composition maintains
a plasma
concentration of about 0.15 ~,M to about 50 ~,M and treats the cancer or
precancerous condition
or prevents the cancer. In one aspect, the plasma concentration can be about
0.1 ~,M to about
100 ~M, about 0.125 ~M to about 75 ~.M; about 0.15 ~.M to about 50 ~,M; about
0.175 ~M to
about 30 ~M; and about 0.2 ~M to about 20 ~M. In another aspect, the
pharmaceutical
composition can maintain a suitable plasma concentration for at least a month,
at least a week, at
least 24 hours, at least 12 hrs, at least 6 hrs, at least 1 hour. In a further
aspect, a suitable plasma
concentration of the pharmaceutical composition can be maintained
indefinitely. In yet another
aspect, the subject can be exposed to the pharmaceutical composition in a AUC
(area under the
curve) range of about 0.5 p,M-hr to about 100 ~,M-hr, about 0.5 ~,M-hr to
about 50 ~.M-hr, about
1 ~M-hr to about 25 ~.M-hr, about 1 ~,M-hr to about 10 ~.M-hr; about 1'.25 ~,M-
hr to about 6.75
~M-hr, about 1.5 ~,M-hr to about 6.5 ~M-hr. The pharmaceutical composition can
be
administered at a dosage from about 2 mg/mz to 5000 mg/mz per day, more
preferably from
about 20 mg/m2 to 2000 mg/m2 per day, more preferably from about 20 mg/ma to
500 mg/m2 per
day, most preferably from about 30 to 300 mg/m2 per day. Preferably, 2 mg/m2
to 5000 mg/mz
per day is the administered dosage for a human. In another aspect, the
pharmaceutical
composition can be administered at a dosage from about 10 to 1,000,000 ~,g per
kilogram body
weight of recipient per day; preferably about 100 to 500,000 ~.g per kilogram
body weight of
recipient per day, more preferably from about 1000 to 250,000 dug per kilogram
body weight of
recipient per day, most preferably from about 10,000 to 150,000 ~g per
kilogram body weight of
recipient per day. One of ordinary skill in the art can determine the
appropriate dosage amount
in mg/mz per day or ~.g per kilogram body weight of recipient per day
depending on subject to
which the pharmaceutical composition is to be administered.
As with the use of other chemotherapeutic drugs, the individual patient will
be monitored
in a manner deemed appropriate by the treating physician. Dosages can also be
reduced if
severe neutropenia or severe peripheral neuropathy occurs, or if a grade 2 or
higher level of
mucositis is observed, using the Common Toxicity Criteria of the National
Cancer Institute.
In administering a G1/S phase compound such as (3-lapachone, the normal dose
of such
compound individually is utilized as set forth above. However, when
combination therapies are
used, it is preferable to use a lower dosage -- preferably 75% or less of the
individual amount,
more preferably 50% or less, still more preferably 40% or less. The term
"effective amount," as
59


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
used herein, refers to an amount effective to treat the disease condition in
combination with any
other active agent in a combination regimen according to the invention.
In therapeutic applications, the dosages of the agents used in accordance with
the
invention vary depending on the agent, the age, weight, and clinical condition
of the recipient
patient, and the experience and judgment of the clinician or practitioner
administering the
therapy, among other factors affecting the selected dosage. Generally, the
dose should be
sufficient to result in slowing, and preferably regressing, the growth of the
tumors and also
preferably causing complete regression of the cancer. An effective amount of a
pharmaceutical
agent is that which provides an obj ectively identifiable improvement as noted
by the clinician or
other qualified observer. Regression of a tumor in a patient is typically
measured with reference
to the diameter of a tumor. Decrease in the diameter of a tumor indicates
regression. Regression
is also indicated by failure of tumors to reoccur after treatment has stopped.
In preferred
embodiments, a decrease in tumor size or burden of at least 20%, more
preferably 50%, 80%,
90%, 95% or 99% is preferred.
This invention further includes compositions comprising a dose of a Gl/S-phase
drug
(such as (3-lapachone or a derivative or analog thereof as provided above) and
a dose of a Gl/S-
phase drug (such as [3-lapachone or a derivative or analog thereof as provided
above). The
compositions may optionally include a pharmceutically acceptable solvent or
carrier. The Gl/S-
phase drug and S-phase drug are present in an effective amount in combination.
In an embodiment, a kit of the present invention comprises a) a first
container
comprising a therapeutically effective amount of (3-lapachone, or a
pharmaceutically acceptable
salt, prodrug, metabolite, analog or derivative thereof, b) a second container
comprising a
therapeutically effective amount of an S phase drug, (e.g., gemcitabine) or a
pharmaceutically
acceptable salt, prodrug, metabolite, analog or derivative thereof, and c)
instructions for using
the (3-lapachone, or a pharmaceutically acceptable salt, prodrug, metabolite,
analog or derivative
thereof, and the S phase drug, or a pharmaceutically acceptable salt, prodrug,
metabolite, analog
or derivative thereof, to treat a subject. In an embodiment, a kit of the
present invention
comprises instructions for using the (3-lapachone, or a pharmaceutically
acceptable salt, prodrug,
metabolite, analog or derivative thereof, and the S phase drug, or a
pharmaceutically acceptable
salt, prodrug, metabolite, analog or derivative thereof, to treat a cell
proliferative disorder, such
as cancer. In an embodiment, a kit of the present invention further comprises
one or more
additional doses of a therapeutically effective amount of (3-lapachone, or a
pharmaceutically
acceptable salt, prodrug, metabolite, analog or derivative thereof. In another
embodiment, a kit
of the present invention further comprises one or more additional doses of a
therapeutically


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
effective amount of an S phase drug, or a pharmaceutically acceptable salt,
prodrug, metabolite,
analog or derivative thereof.
In one embodiment, a kit of the present invention comprises (3-lapachone, or a
pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative
thereof, and the S
phase drug, or a pharmaceutically acceptable salt, prodrug, metabolite, analog
or derivative
thereof, in separate containers. In another embodiment, a kit of the present
invention comprises
[3-lapachone, or a pharmaceutically acceptable salt, prodrug, metabolite,
analog or derivative
thereof, and the S phase drug, or a pharmaceutically acceptable salt, prodrug,
metabolite, analog
or derivative thereof, in a single container. In another embodiment, a kit of
the present invention
comprises [3-lapachone, or a pharmaceutically acceptable salt, prodrug,
metabolite, analog or
derivative thereof, and the S phase drug, or a pharmaceutically acceptable
salt, prodrug,
metabolite, analog or derivative thereof, in combination with a
pharmaceutically acceptable
carrier (e.g., for co-administration via a single pill or single intravenous
formulation).
In an embodiment, a kit of the present invention comprises instructions for
administering
(3-lapachone, or a pharmaceutically acceptable salt, prodrug, metabolite,
analog or derivative
thereof, by a route of administration selected from the group consisting of
orally, intravenously,
intramuscularly, and by injection. In another embodiment, a kit of the present
invention
comprises instructions for administering the S phase drug, or a
pharmaceutically acceptable salt,
prodrug, metabolite, analog or derivative thereof, by a route of
administration selected from the
group consisting of orally, intravenously, intramuscularly, and by injection.
In another embodiment, a kit of the present invention comprises a dose of a
G1/S-phase
drug (such as ~-lapachone or a derivative or analog thereof as provided above)
and a dose of a
Gl/S-phase drug (such as (3-lapachone or a derivative or analog thereof as
provided above), e.g.,
at the doses provided above. Each dose of drug may be contained in an
individual vial.
Preferably, the kit contains instructions describing the use of the drugs in
combination for the
treatment of cancer. In an embodiment, the therapeutically effective amount of
the Gl/S phase
drug, or a derivative or analog thereof, is contained in a first vial, and the
S phase drug is
contained in a second vial, the contents of the first and second vials being
administered to the
patient simultaneously or sequentially.
The invention is further defined by reference to the following examples. It is
understood
that the foregoing detailed description and the following examples are
illustrative only and are
not to be taken as limitations upon the scope of the invention. It will be
apparent to those skilled
in the art that many modifications, both to the materials and methods, may be
practiced without
departing from the purpose and interest of the invention.
61


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
EXAMPLES
Example 1. [3-lapachone administered in monotherapy, or in combination with
gemcitabine (GEMZAR~), potently reduces mean tumor volume in a human
pancreatic
cancer xenograft mouse model.
(i-lapachone alone, and in combination with gemcitabine, shows striking
efficacy in a
pancreatic cancer xenograft model. Human pancreatic cancer Panc-1 cells
(2x106) are implanted
subcutaneously into male athymic nude (Ncr) mice. Following establishment of
tumor nodules
(about 50 mm3), the animals are randomized into four groups of five animals
per group. The
animals are treated intraperitoneally with one of four treatments: (3-
lapachone at 40 mg/kg in
40% hydroxypropy-(3-cyclodextran; gemcitabine (Gemzar0) at 120 mg/kg (in PBS);
(3-
lapachone (40mg/kg) + gemcitabine (120mglkg); or vehicle control. The mice
receive a total of
ten treatments, administered every three days (on study day 5, 8, 11, 14, 17,
20, 31, 34, 37, and
40). The combination therapy group receives (3-lapachone and gemcitabine on
the same day in
each treatment. Following treatment, the animals are observed for an
additional 19 days.
Tumors are measured throughout the treatment and post-treatment periods.
As shown in Figure 1 and Table 2, treatment with ~-lapachone alone induced
tumor
regression by 30-40% at termination of treatment. In contrast, tumors in
vehicle-treated control
group increased to 300%. There was no significant re-growth after termination
of the (i-
lapachone treatment. Gemcitabine at 120 mglkg reduces tumor size by 30-40% at
termination of
treatment; however, tumors in animals treated with gemcitabine re-grew by 400%
after
termination of the treatment. When given in combination, [3-lapachone and
gemcitabine induced
nearly complete regression of formed tumors. No treatment-related signs of
toxicity were noted,
and weights of treated animals are comparable to those of control animals at
the end of the
study.
Table 2. Statistical Significance for Human Pancreatic Tumor Xenograft Model
at Day 60
Control(3-lapachone ~ [3-lapachone
40 mg/kg, Gemzar 120 +
mg/kg,


q3d q3d GemzarR,
q3d


Control P = 0.16577 P = 0.991695 P = 0.000655


~i-lapachone P = 0.248456 P = 0.018434
40 mg/kg,


3d


Gemzar~ 120 mg/kg, P=0.006442


3d


62


CA 02556759 2006-08-17
WO 2005/082354 PCT/US2005/005620
Example 2. [3-lapachone administered in monotherapy, or in combination with
gemcitabine (GEMZAR~), potently reduces mean tumor volume in a human lung
cancer
xenograft mouse model.
The anti-tumor activity of (3-lapachone is examined using a human lung cancer
xenograft
model. Briefly, athymic female nude mice (Ncr) are inoculated subcutaneously
with 4x106
A549 human lung cancer cells, and the tumors are allowed to grow to
approximately SOmm3 in
size. The animals are randomized into five groups of seven animals per group,
and treated
intraperitoneally every three days with one of the following five regimens: [3-
lapachone at
40mg/kg in 40% hydroxypropy-(3-cyclodextran ("HPBCD"); ~i-lapachone at 60mg/kg
in 40%
HPBCD; gemcitabine (GEMZAR~) at 120mg/kg in PBS; (3-lapachone (40mg/kg) +
gemcitabine
(120mg/kg); or vehicle control (40% HPBCD). The combination therapy group
receives
treatments with (3-lapachone and gemcitabine at the indicated concentrations
on the same day,
every three days. Mice receive a total of eight treatments. Mean tumor volume
is analyzed; data
points in Figure 2 represent the arithmetic mean +/- SEM of five tumors.
As shown in Figure 2, treatment with either ~i-lapachone (60mg/kg) or
gemcitabine
(120mg/kg) alone retarded tumor growth to a similar extent during treatment.
See, e.g., Figure
2, days 24 and 27 of treatment. Animals treated with (3-lapachone (40mg/kg) in
combination
with gemcitabine (120mg/kg) showed an unexpected synergistic retardation of
tumor growth. [3-
lapachone dosed at 60 mg/kg was shown to be more effective at retarding tumor
growth than (3-
lapachone dosed at 40 mg/kg. No significant toxicity was noted for any of the
treatment
regimens. We conclude from this study that (3-lapachone either alone, or in
combination with
gemcitabine, can be safely dosed in regimens that are effective for treating
lung cancer.
63

Representative Drawing

Sorry, the representative drawing for patent document number 2556759 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2005-02-23
(87) PCT Publication Date 2005-09-09
(85) National Entry 2006-08-17
Examination Requested 2006-08-17
Dead Application 2009-12-29

Abandonment History

Abandonment Date Reason Reinstatement Date
2008-12-29 R30(2) - Failure to Respond
2009-02-23 FAILURE TO PAY APPLICATION MAINTENANCE FEE

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Request for Examination $800.00 2006-08-17
Application Fee $400.00 2006-08-17
Registration of a document - section 124 $100.00 2006-11-29
Maintenance Fee - Application - New Act 2 2007-02-23 $100.00 2007-01-26
Maintenance Fee - Application - New Act 3 2008-02-25 $100.00 2008-01-31
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
ARQULE, INC.
Past Owners on Record
LI, CHIANG J.
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2006-08-17 2 105
Claims 2006-08-17 7 350
Drawings 2006-08-17 2 28
Description 2006-08-17 63 4,362
Description 2006-08-18 67 4,555
Claims 2006-08-18 10 335
Cover Page 2006-10-19 1 36
PCT 2006-08-17 4 121
Assignment 2006-08-17 2 107
Prosecution-Amendment 2006-08-17 23 977
Correspondence 2006-10-13 1 27
Assignment 2006-11-29 3 119
Fees 2007-01-26 1 49
PCT 2006-08-18 4 261
Fees 2008-01-31 1 49
Prosecution-Amendment 2008-06-26 4 191