CA2556789A1 - Use of beta-lapachone for treating hematologic tumors - Google Patents

Use of beta-lapachone for treating hematologic tumors Download PDF

Info

Publication number
CA2556789A1
CA2556789A1 CA002556789A CA2556789A CA2556789A1 CA 2556789 A1 CA2556789 A1 CA 2556789A1 CA 002556789 A CA002556789 A CA 002556789A CA 2556789 A CA2556789 A CA 2556789A CA 2556789 A1 CA2556789 A1 CA 2556789A1
Authority
CA
Canada
Prior art keywords
lapachone
pharmaceutically acceptable
acceptable salt
cell
cancer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002556789A
Other languages
French (fr)
Inventor
Chiang J. Li
Youzhi Li
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Arqule Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2556789A1 publication Critical patent/CA2556789A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Pyrane Compounds (AREA)

Abstract

The present invention provides for methods that utilize agents effective in the treatment of hematologic cancers and pre-cancerous hematologic cancer conditions. Moreover, the present invention provides agents capable of acting as an inhibitor of cell proliferation in hematologic cells.

Description

USE OF BETA-LAPACHONE FOR TREATING HEMATOLOGIC TUMORS
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims the benefit under 35 U.S.C. ~ 119(e) of U.S.
Application No. 60/545,914, filed February 20, 2004.
BACKGROUND OF THE INVENTION
Hematologic cancers, including lymphomas and leukemias, are a leading cause of cancer mortality in the United States ("Cancer Facts and Figures 2003," American Cancer Society).
For example, non-Hodgkin's lymphoma is the sixth leading cause of cancer mortality in the U.S.
and is particularly insidious because symptoms are nonspecific and are frequently attributed to the effects of infectious etiologies. Non-Hodgkin's lymphoma can be cured in only about one third of the patients. ("Neoplasms of the Hematopoitic System: Non-Hodgkin's Lymphomas,"
in Cancer Medicine, 5th Edition, Bast et al. eds., B.C. Decker Inc., Hamilton, Ontario). In addition, various leukemias are estimated to have resulted in about 22,000 deaths in 2003 ("Cancer Facts and Figures 2003," American Cancer Society).
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 the new chemotherapies entering the market, improvement in patient survival is measured in months rather than in years, and the need continues for new drugs effective both 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).
(3-lapachone 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. 6,245,07 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, or a combination of these, in xenotransplant mouse models of human ovarian cancer (Li, C.J. et al., (1999) Pr~oc. Natl. Acad.
Sci. USA, 96(23): 13369-13374), human prostate cancer (Li, C.J. et al., (1999) Proc. Natl. 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).
While [3-lapachone, alone or in combination with other agents, has been reported to reduce tumor size in a limited number of tumor models it has not been reported to be an effective agent for the treatment of certain human hematologic cancers.
SUMMARY OF THE INVENTION
The present invention provides a method of treating a cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm, comprising administering to a subject in need thereof a therapeutically effective amount of (3-lapachone, or a pharmaceutically acceptable salt thereof, in combination with a pharmaceutically acceptable Garner, where the cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm is treated.
The present invention also provides a method of treating lymphoma comprising administering to a subject in need thereof a therapeutically effective amount of [3-lapachone, or pharmaceutically acceptable salt thereof, in combination with a pharmaceutically acceptable Garner, where the lymphoma is treated.
The present invention also provides a method of treating leukemia comprising administering to a subject in need thereof a therapeutically effective amount of (3-lapachone, or pharmaceutically acceptable salt thereof, in combination with a pharmaceutically acceptable Garner, where the leukemia is treated.
The present invention also provides a method of treating myeloid neoplasms comprising administering to a subject in need thereof a therapeutically effective amount of (3-lapachone, or pharmaceutically acceptable salt thereof, in combination with a pharmaceutically acceptable carrier, where the myeloid neoplasms are treated.
The present invention also provides a method for inducing cell death in a cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm, comprising contacting the cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm with an effective amount of (3-lapachone, or a pharmaceutically acceptable salt thereof, where the contacting induces the cell death in the a cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm.
The present invention also provides a method of treating a cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm, comprising administering to a subject in need thereof a therapeutically effective amount of ~3-lapachone, or a pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative thereof, in combination with a pharmaceutically acceptable carrier, where the cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm is treated.
The present invention also provides a method of treating lymphoma comprising administering to a subject in need thereof a therapeutically effective amount of (3-lapachone, or a pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative thereof, in combination with a pharmaceutically acceptable carrier, where the lymphoma is treated.
The present invention also provides a method of treating leukemia comprising administering to a subject in need thereof a therapeutically effective amount of (3-lapachone, or a pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative thereof, in combination with a pharmaceutically acceptable carrier, where the leukemia is treated.
The present invention also provides a method of treating myeloid neoplasms comprising administering to a subject in need thereof a therapeutically effective amount of (3-lapachone, or a pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative thereof, in combination with a pharmaceutically acceptable carrier, where the myeloid neoplasms are treated.
The present invention also provides a method for inducing cell death in a cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm, comprising contacting the cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm with an effective amount of (3-lapachone, or a pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative thereof, where the contacting induces the cell death in the a cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm.
The present invention also provides a method of treating a cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm, 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 carrier, where the cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm is treated.
The present invention also provides a method for inducing cell death in a cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm, comprising contacting the lung 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 selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm.
The present invention also provides a method of treating a cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm, comprising achninistering 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 cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm is treated.
The present invention also provides a method for inducing cell death in a cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm comprising contacting the lung cancer cell with an effective amount of a) (3-lapachone, or a pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative thereof, and b) gemcitabine, or a pharmaceutically acceptable salt, prodrug, metabolite, analog or. derivative thereof, where the contacting induces the cell death in the cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm.
The present invention provides a method of treating a hematologic cancer selected from the group consisting of a 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, and mast cell leukemia), myeloid neoplasms, and mast cell neoplasms (collectively "a hematologic cancer of the present invention"), comprising administering to a subject in need thereof a therapeutically effective amount of (3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, in combination with a pharmaceutically acceptable Garner, where the [3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, treats the cancer selected from the group consisting of a hematologic cancer of the present invention.
The present invention also provides a method of treating a cancer selected from the group consisting of a hematologic cancer of the present invention, comprising administering to a subject in need thereof a therapeutically effective amount of (3-lapachone or pharmaceutically acceptable salt thereof, or a metabolite thereof, in combination with a pharmaceutically acceptable Garner, and activating one or more cell cycle checkpoints in cells of a hematologic cancer of the present invention, where the (3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, treats the cancer selected from the group consisting of a hematologic cancer of the present invention.
The present invention further provides a method of treating a cancer selected from the group consisting of a hematologic cancer of the present invention, comprising administering to a subject in need thereof a therapeutically effective amount of [3-lapachone or pharmaceutically acceptable salt thereof, or a metabolite thereof, in combination with a pharmaceutically acceptable carrier, and activating one or more cell cycle checkpoint pathways in one or more cells of a cancer selected from the group consisting of a hematologic cancer of the present invention, where the (3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, treats the cancer selected from the group consisting of a hematologic cancer of the present invention The present invention also provides a method of treating a cancer selected from the group consisting of a hematologic cancer of the present invention, comprising administering to a subject in need thereof a therapeutically effective amount of [3-lapachone or pharmaceutically acceptable salt thereof, or a metabolite thereof, in combination with a pharmaceutically acceptable carrier, and activating one or more cell cycle checkpoint regulators in one or more cells of a cancer selected from the group consisting of a hematologic cancer of the present invention, where the (3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, treats the cancer selected from the group consisting of a hematologic cancer of the present invention.
The present invention further provides a method of treating a cancer selected from the group consisting of a hematologic cancer of the present invention, comprising administering to a subject in need thereof a therapeutically effective amount of (3-lapachone or pharmaceutically acceptable salt thereof, or a metabolite thereof, in combination with a pharmaceutically acceptable carrier, and activating cell death selectively in one or more cells of a cancer selected from the group consisting of a hematologic cancer of the present invention, where the (3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, treats the cancer selected from the group consisting of a hematologic cancer of the present invention.
The present invention also provides a method of treating or preventing a cell proliferative disorder selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, mast cell neoplasm, myelodysplasia, benign monoclonal gammopathy, lymphomatoid granulomatosis, lymphomatoid papulosis, polycythemia vera, chronic myelocytic leukemia, agnogenic myeloid metaplasia, and essential thrombocythemia (collectively, "a hematologic cell proliferative disorder of the present invention"), comprising administering to a subj ect in need thereof a therapeutically effective amount of (3-lapachone or pharmaceutically acceptable salt thereof, or a metabolite thereof, in combination with a pharmaceutically acceptable Garner, where the (3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, treats or prevents the cell proliferative disorder selected from the group consisting of a hematologic cell proliferative disorder of the present invention.
The present invention provides a method of treating a cancer selected from the group consisting of a hematologic cancer of the present invention, comprising administering to a subject in need thereof a therapeutically effective amount of a derivative or analog of (3-lapachone or pharmaceutically acceptable salt thereof, or a metabolite thereof, in combination with a pharmaceutically acceptable carrier, where the derivative or analog of (3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, treats the cancer selected from the group consisting of a hematologic cancer of the present invention.
The present invention also provides a method of treating a cancer selected from the group consisting of a hematologic cancer of the present invention, comprising administering to a subject in need thereof a therapeutically effective amount of a derivative or analog of ~3-lapachone or pharmaceutically acceptable salt thereof, or a metabolite thereof, in combination with a pharmaceutically acceptable Garner, and activating one or more cell cycle checkpoints in one or more cells of a cancer selected from the group consisting of a hematologic cancer of the present invention, where the derivative or analog of (3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, treats the cancer selected from the group consisting of a hematologic cancer of the present invention.
The present invention further provides a method of treating a cancer selected from the group consisting of a hematologic cancer of the present invention, comprising administering to a subject in need thereof a therapeutically effective amount of a derivative or analog (3-lapachone or pharmaceutically acceptable salt thereof, or a metabolite thereof, in combination with a pharmaceutically acceptable Garner, and activating one or more cell cycle checkpoint pathways in one or more cells of a cancer selected from the group consisting of a hematologic cancer of the present invention, where the [3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, treats the cancer selected from the group consisting of a hematologic cancer of the present invention.
The present invention also provides a method of treating a cancer selected from the group consisting of a hematologic cancer of the present invention, comprising administering to a subject in need thereof a therapeutically effective amount of a derivative or analog of ~3-lapachone or pharmaceutically acceptable salt thereof, or a metabolite thereof, iri combination with a pharmaceutically acceptable carrier, and activating one or more cell cycle checkpoint regulators in one or more cells of a cancer selected from the group consisting of a hematologic cancer of the present invention, where the derivative or analog of [3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, treats the cancer selected from the group consisting of a hematologic cancer of the present invention.
The present invention further provides a method of treating a cancer selected from the group consisting of a hematologic cancer of the present invention, comprising administering to a subject in need thereof a therapeutically effective amount of a derivative or analog of (3-lapachone or pharmaceutically acceptable salt thereof, or a metabolite thereof, in combination with a pharmaceutically acceptable Garner, and activating cell death selectively in one or more cells of a cancer selected from the group consisting of a hematologic cancer of the present invention, where the derivative or analog of (3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, treats the cancer selected from the group consisting of a hematologic cancer of the present invention.
The present invention also provides a method of treating or preventing a cell proliferative disorder selected from the group consisting of a hematologic cell proliferative disorder of the present invention, comprising administering to a subject in need thereof a therapeutically effective amount of a derivative or analog of (3-lapachone or pharmaceutically acceptable salt thereof, or a metabolite thereof, in combination with a pharmaceutically acceptable Garner, where the derivative or analog of (3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, treats or prevents the cell proliferative disorder selected from the group consisting of a hematologic cell proliferative disorder of the present invention.
The present invention also provides a method of treating or preventing a hematologic cell proliferative disorder of the present invention, comprising administering to a subject in need thereof a therapeutically effective amount of (3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, in combination with a pharmaceutically acceptable carrier, and activating one or more cell cycle checkpoints in a hematologic cell, where the ~i-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, treats or prevents a hematologic cell proliferative disorder of the present invention.
The present invention also provides a method of treating or preventing a hematologic cell proliferative disorder of the present invention, comprising administering to a subj ect in need thereof a therapeutically effective amount of (3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, in combination with a pharmaceutically acceptable carrier, and activating one or more cell cycle checkpoint pathways in a hematologic cell, where the [3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, treats or prevents a hematologic cell proliferative disorder of the present invention.
The present invention also provides a method of treating or preventing a hematologic cell proliferative disorder of the present invention, comprising administering to a subj ect in need thereof a therapeutically effective amount of (3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, in combination with a pharmaceutically acceptable carrier, and activating one or more cell cycle checkpoint regulators in a hematologic cell, where the (3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, treats or prevents a hematologic cell proliferative disorder of the present invention.
The present invention also provides a method of treating or preventing a hematologic cell proliferative disorder of the present invention, comprising administering to a subject in need thereof a therapeutically effective amount of (3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, in combination with a pharmaceutically acceptable carrier, and activating cell death selectively in hematologic cell, where the [3-lapachone or pharmaceutically acceptable salts thereof, or a metabolite thereof, treats or prevents a hematologic cell proliferative disorder of the present invention.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 sets forth a schematic of the proposed points of action of (3-Lapachone and Taxol on the cell cycle.
Figure 2 sets forth an effect of ~i-Lapachone on survival of human cancer cell lines in the NCI60 assay in vitro.

DETAILED DESCRIPTION OF THE INVENTION
The present invention provides a method of treating a cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm comprising administering to a subject in need thereof a therapeutically effective amount of (3-lapachone, or a pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative thereof, in combination with a pharmaceutically acceptable Garner, where the a cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm is treated.
The present invention also provides a method of treating or preventing a cell proliferative disorder of the hematologic system, comprising administering to a subj ect in need thereof a therapeutically effective amount of [3-lapachone, or a pharmaceutically acceptable salt, prodrug, metabolite, malog or derivative thereof, in combination with a pharmaceutically acceptable carrier, where the cell proliferative disorder of the hematologic system is treated or prevented.
The invention also provides the use of (3-lapachone for the preparation of a medicament useful for the treatment of a cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm. The invention also provides the use of (3-lapachone for the preparation of a medicament useful for the treatment or prevention of a cell proliferative disorder of the hematologic system. The invention also provides the use of an analog or derivative of (3-lapachone for the preparation of a medicament useful for the treatment of hematologic cancer. The invention also provides the use of an analog or derivative of (3-lapachone for the preparation of a medicament useful for the treatment or prevention of a cell proliferative disorder of the hematologic system. .
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 can be delayed at checkpoints in a regulated fashion, resulting in no cell death-prone collisions.
Third, normal cells with intact Gl checkpoint control are expected to arrest in Gl. Cancer cells, on the other hand, are expected to be delayed in S-, G2-, and M-phases, since most cancer cells harbor G1 checkpoint defects, making cancer cells more sensitive to drugs imposing S and M
phase checkpoints. ~3-lapachone is a Gl and S phase compound, and contacting a cell with [3-lapachone results in activation of a Gl or S cell cycle checkpoint. Figure 1 sets forth a schematic of the proposed points of action of (3-lapachone and Taxol on the cell cycle.
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 Gl 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. Proc. Natl. Acad. Sci. USA (1999), 96(23), 13369-13374, and Y. Li et al.
Pr°oc. 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 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 Chk1 respectively, which in turn activate a variety of substrates involved in arresting cells at the Gl/S 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 BRCAl, 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, EZF1 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 E2F1/Rb complex following phosphorylation of Rb. E2F1 levels rise, promoting progression through G1.
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 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].
I. Compositions As used herein, the phrase "[3-lapachone" refers to 3,4-dihydro-2,2-dimethyl-naphtho[1,2-b]pyran-5,6-dione and derivatives and analogs thereof, and has the chemical structure:
O
Formula I
Preferred derivatives and analogs are discussed below.

(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. Ana. 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. Ana. Chem. Soc. 58:1181-1190). [3-Lapachone can, for example, be obtained by simple sulfuric acid treatment of the naturally occurring lapachol, which is readily isolated from Tabebuia avellenedae growing mainly in Brazil, or is easily synthesized from seeds of lomatia growing in Australia (Li, CJ, et al., (1993) J. Biol. Chem. 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-A1.
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-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.S. 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) Bulletin. of Tlae Chemical Society ofJapan 43:1454-1459;
Tapia, R.A et al., (2000) Heterocycles 53(3):585-598; Tapia, RA et al., (1997) Tetrahedron Lettet~s 38(1):153-154; Chuang, CP et al., (1996) Heterocycles 40(10):2215-2221; Suginome H
et al., (1993) Journal of the Chemical Society, Chemical Communications 9:807-809; Tonholo J
et al., (1988) Journal of the Brazilian Cherraical Society 9(2):163-169; and Krapcho AP et al., (1990) Journal ofMedicinal Chemistry 33(9):2651-2655).
Further, derivatives or analogs of (3-lapachone 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, Cl-C6 alkyl, C1-C6 alkylcarbonyl, or a pharmaceutically acceptable salt).
)R
Formula Ia While (3-lapachone is the preferred Gl/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/IJS93/07878 (WO 94/04145), which discloses compounds of the formula:
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. 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 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:
O
R
Formula III
where R and Rl are each independently selected from hydrogen, hydroxy, sulfliydryl, 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/LTS00/10169 (WO00/61142), which disclose compounds of the structure:

O
Formula IV
where R5 and R6 may be independently selected from hydroxy, C1-C~ alkyl, C1-C6 alkenyl, C1-C6 alkoxy, C1-C6 alkoxycaxbonyl, --(CH2)ri phenyl; and R7 is hydrogen, hydroxyl, C1-C6 alkyl, C1-C6 alkenyl, C1-C6 alkoxy, C1-C6 alkoxycarbonyl, --(CH2)"-amino, --(CH2)"-aryl, --(CH2)n-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 Chem 27:990-994 (1984), wluch discloses (3-lapachone with substitutions at one or more of the following positions: 2-, 8- and/or 9- positions. See also Portela et al., BioclZem PlZaf°m 51:275-283 (1996) (substituents at the 2- and 9-positions); Maruyama et al., Cherrl 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., IfZdian Jours2al 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:
O O

R R

Formula V Formula VI
where the dotted double bond between the ring carbons represents an optional ring double bond and where R and Rt 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 (i.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-C~ alkyl, Ci-C6 alkenyl, C1-C6 alkoxy, C1-C6 alkoxycarbonyl, --(CH2)"-aryl, --(CH2)ri heteroaryl, --(CH2)"-heterocycle, or --(CH2)"-phenyl; and R7 is hydrogen, hydroxyl, C1-C6 alkyl, C1-C6 alkenyl, C1-C6 alkoxy, C1-C6 alkoxycarbonyl, --(CH2)ri amino, --(CH2)"-aryl, --(CHz)"-heteroaryl, --(CH2)"-heterocycle, or --(CHZ)ri 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:
Formula VII

where Rl is (CH2)n R2, where n is an integer from 0-10 and R2 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-lapachone, 4-acetoxy-3-bromo-[3-lapachone, 4-keto-(3-lapachone, 7-hydroxy-(3-lapachone, 7-methoxy-(3-lapachone, 8-hydroxy-(3-lapachone, 8-methoxy-(3-lapachone, 8-chloro-(3-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
t1 Formula VIII
where Rl-R4 are each, independently, selected from the group consisting of H, C1-C6 alkyl, C1-CG allcenyl, C1-C~ alkoxy, C1-C6 alkoxycarbonyl, --(CHZ)ri aryl, --(CH2)"-heteroaryl, --(CHZ)n heterocycle, or --(CH2)"-phenyl; or Rl and RZ combined are a single substituent selected from 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 NOZ.
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/IJS2003/037219, 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-C6 alkyl, substituted and unsubstituted C1-C6 alkenyl, substituted and unsubstituted C1-C6 alkoxy, substituted and unsubstituted C1-C~
alkoxycarbonyl, substituted and unsubstituted C1-C~ acyl, -(CH2)"-amino, -(CH2)"-aryl, -(CHZ)"-heterocycle, and -(CHZ)"-phenyl;
or one of Rl 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, nitro, 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):

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 CI-C6 alkyl, substituted and unsubstituted C1-C6 alkenyl, substituted and unsubstituted C1-C6 alkoxy, substituted and unsubstituted C1-C6 alkoxycarbonyl, substituted and unsubstituted C1-C~ acyl, -(CH2)"-amino, -(CH2)n-aryl, -(CHZ)n heterocycle, and -(CHZ)n 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, nitro, cyano or amide; and n is an integer from 0 to 10. In certain embodiments of Formula XI, Rl, R2, R3, R4, R5, R6, R7 and R8 are not each simultaneously H.
All stereoisomers of the compounds of the instant invention are contemplated, either in admixture or in pure or substantially pure form. 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 or separation by chiral column 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.
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 (3-lapachone, or a pharmaceutically acceptable salt, analog or derivative thereof, that exhibits a similar activity in vivo to (3-lapachone.
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 of the hematologic system, or a subject having an increased risk of developing a cell proliferative disorder of the hematologic system relative to the population at large. In one aspect, a subj ect in need thereof has a precancerous condition of the hematologic system. In a preferred aspect, a subject in need thereof has hematologic cancer. In an aspect, the subject may be suffering from a known (i.e., diagnosed) condition characterized by cell hyperproliferation (e.g., cancer) of the hematologic system.

As used herein, an "immune cell" is any cell that functions in an immune response or is a direct precursor to a cell that functions in an immune response, including but not limited to hematopoietic cells, lymphoid cells, myeloid cells, lymphocyte precursors, B
cell precursors, T
cell precursors, lymphocytes, B cells, T cells, plasma cells, monocytes, macrophages, neutrophils, eosinophils, basophils, natural killer cells (i.e., NK cells), mast cells, and dendritic cells. As used herein, a "white blood cell" (i.e., a leukocyte) is a blood cells that lacks . hemoglobin, and includes but is not limited to lymphocytes, B cells, T
cells, monocytes, macrophages, natural killer cells, neutrophils, eosinophils, and basophils. As used herein, a "lymphocyte" is type of white blood cell that is continuously made in the bone marrow, may be present in blood, lymph nodes, spleen, thymus, gut wall and bone marrow, and includes but is not limited to B lymphocytes and T lymphocytes. As used herein, a "monocyte"
is a large white blood cell that is capable of phagocytosis and which, when it enters tissue, develops into a macrophage. As used herein a "neutrophil" is a white blood cell that is capable of phagocytosis and is distinguished by a lobed nucleus and granular cytoplasm. As used herein, a "natural killer cell" is a subset of bone marrow-derived lymphocytes, distinct from B or T
cells, that function in innate immune responses through lytic mechanisms and by secreting IFN-y.
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, for example, hematologic cancer and precancerous conditions of the hematologic system. 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 includes a pre-cancer or precancerous condition of the hematologic system. In one aspect, a cell proliferative disorder of the hematologic system includes a non-cancerous cell proliferative disorder of the hematologic system. In another aspect, a cell proliferative disorder includes hematologic cancer, including metastatic lesions in other tissues or organs distant from the 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.

In one aspect, cell proliferative disorders of the hematologic system include all forms of cell proliferative disorders affecting hematologic cells, e.g., benign and premalignant hematologic conditions of the hematologic system, as well as cancers of the hematologic system.
In one aspect, cell proliferative disorders of the hematologic system include hematologic cancer and precancerous conditions of the hematologic system. In one aspect, cell proliferative disorders of the hematologic system include hyperplasia, metaplasia, and dysplasia of the hematologic system. In one aspect, cell proliferative disorders of the hematologic system include sporadic and hereditary cell proliferative disorders of the hematologic system. In one aspect, cell proliferative disorders of the hematologic system include benign tumors of the hematologic system. In an aspect, a cell proliferative disorder of the hematologic system that is to be treated includes a cell proliferative disorder of immune cells. In another aspect, a cell proliferative disorder of the hematologic system includes a cell proliferative disorder of white blood cells. In another aspect, a cell proliferative disorder of the hematologic system includes a cell proliferative disorder of lymphocytes, monocytes, macrophages, natural killer cells, neutrophils, eosinophils, or basophils. In another aspect, a cell proliferative disorder of the hematologic system includes a cell proliferative disorder of T cells or B cells. In another aspect, a cell proliferative disorder of the hematologic system includes a cell proliferative disorder of myeloblasts or monoblasts. In another aspect, a cell proliferative disorder of the hematologic system includes a cell proliferative disorder of mononuclear cells (e.g., lymphocytes and monocytes) or granulocytes (e.g., neutrophils, eosinophils, and basophils). In one aspect, a cell proliferative disorder of the hematologic system includes a cell proliferative disorder of plasma cells (e.g., multiple myeloma). In another aspect, a cell proliferative disorder of the hematologic system does not include a cell proliferative disorder of plasma cells (e.g., multiple myeloma).
In an aspect, a cell proliferative disorder of the hematologic system that is to be treated includes lymphoma (including, for example, Hodgkin's lymphoma, non-Hodgkin's lymphoma, childhood lymphomas, and lymphomas of lymphocytic and cutaneous origin), leukemia (including, for example, childhood leukemia, hairy-cell leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, chronic lymphocytic leukemia, chronic myelocytic leukemia, and mast cell leukemia), myeloid neoplasms, and mast cell neoplasm. In another aspect, a cell proliferative disorder of the hematologic system that is to be treated includes 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 that is to be treated is a myelodyplastic syndrome (e.g., refractory anemia, refractory anemia with ringed sideroblasts, refractory cytopenia with multilineage dysplasia, refractory cytopenia with multilineage dysplasia and ringed sideroblasts, refractory anemia with excess blasts-1 (RAEB-1), refractory anemia with excess blasts-2 (R.AEB-2), myelodysplastic syndrome unclassified (MDS-L~, and MDS associated with isolated del(Sq)). Other exemplary cell proliferative disorder of the hematologic system are known to those of ordinary skill in the art.
In a preferred aspect, the cell proliferative disorder of the hematologic system is selected from the group consisting of a hematologic cancer of the present invention or a hematologic cell proliferative disorder of the present invention. In a preferred aspect, compositions of the present invention may be used to treat a hematologic cancer of the present invention or a hematologic cell proliferative disorder of the present invention.
In one aspect, a hematologic cancer that is to be treated includes lymphoma (including, for example, Hodgkin's lymphoma, non-Hodgkin's lymphoma, childhood lymphomas, and lymphomas of lymphocytic and cutaneous origin), leukemia (including, for example, childhood leukemia, hairy-cell leukemia, acute lyrnphocytic leukemia, acute myelocytic leukemia, chronic lymphocytic leukemia, chronic myelocytic leukemia, and mast cell leukemia), myeloid neoplasms and mast cell neoplasms. Cancers to be treated may include but are not limited to sarcoma, carcinoma, and adenocarcinoma. In an aspect, a hematologic cancer that is to be treated includes a cancer of immune cells. In another aspect, a hematologic cancer includes a cancer of white blood cells. In another aspect, a hematologic cancer includes a cancer of lymphocytes, monocytes, macrophages, natural killer cells, neutrophils, eosinophils, or basophils. In another aspect, a hematologic cancer includes a cancer of T
cells or B cells. In .
another aspect, a hematologic cancer includes a cancer of myeloblasts or monoblasts. In another aspect, a hematologic cancer includes a cancer of mononuclear cells (e.g., lymphocytes and monocytes) or granulocytes (e.g., neutrophils, eosinophils, and basophils). In another aspect, a hematologic cancer to be treated is selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm. Tn one aspect, a hematologic cancer to be treated includes a cancer of plasma cells (e.g., multiple myeloma). In another aspect, a hematologic cancer to be treated does not include a cancer of plasma cells (e.g., multiple myeloma).
In one aspect, a hematologic cancer that is to be treated is a leukemia. As used herein, the term "leulcernia" does not encompass multiple myeloma. In one aspect, a leukemia to be treated includes acute leukemias and chronic leukemias. In an aspect, a leukemia to be treated includes lymphocytic leukemias derived from lymphocytes. In another aspect, a leukemia to be treated includes myeloid leukemias (e.g., myeloid neoplasms). In another aspect, a leukemia to be treated includes childhood leukemia, hairy-cell leukemia, acute lymphocytic leukemia (ALL), acute myelocytic leukemia (AML), acute promyelocytic leukemia, chronic lymphocytic leukemia (CLL), chronic myelocytic leukemia (CML), mast cell leukemia, prolylnphocytic leukemia and T-cell chronic lymphocytic leukemia.
In one aspect, a hematologic cancer that is to be treated is myeloid neoplasms. As used herein, the term "myeloid neoplasms" does not encompass multiple myeloma. As used herein, the term "myeloid neoplasms" refers to cancers of cells of the myeloid lineage, e.g., myeloid (myelocytic or myelogenous) leukemias derived from granulocytes (e.g., neutrophils, eosinophils, and basophils) or monocytes; for example, chronic myelocytic leukemia, acute myelocytic leukemia, chronic neutrophilic leukemia, chronic eosinophilic leukemia, and myelodyplastic syndromes.
In one aspect, a hematologic cancer that is to be treated is a lymphoma. As used herein, the term "lymphoma" does not encompass multiple myeloma. In one aspect, a a hematologic cancer that is to be treated is a B cell lymphoma or a T cell lymphoma. In one aspect, a hematologic cancer that is to be treated is non-Hodgkin's lymphoma, Hodgkin's disease (e.g., Hodgkin's lymphoma), childhood lymphomas, or lymphomas of lymphocytic and cutaneous origin. In another aspect, a hematologic cancer that is to be treated is a diffuse large B-cell lymphoma (DLBCL); follicular lymphoma; small lymphocytic lymphoma (SLL);
mantle cell lymphoma; extranodal marginal zone B cell lymphoma; mucosa-associated lyphoid tissue (MALT) lymphoma; nodal marginal zone B cell lymphoma; splenic marginal zone B
cell lymphoma; primary mediastinal B cell lymphoma; Burkitt's lymphoma;
lymphoplamocytic lymphoma (Waldenstrom macroglobulinemia); primary central nervous system (CNS) lymphoma. In one aspect, a hematologic cancer that is to be treated is a peripheral T-cell lymphoma, such as cutaneous T cell lymphoma, angioimmunoblastic T-cell lymphoma, extranodal natural killer/T cell lymphoma, enteropathy type T cell lymphoma, subcutaneous panniculitis-like T cell lymphoma, anaplastic large cell lymphoma, anaplastic large T/null-cell lymphoma, or null cell lymphoma. In another aspect, a hematologic cancer that is to be treated is precursor T-lymphoblastic lymphoma/leukemia. In an aspect, a hematologic cancer that is to be treated is granulocytic sarcoma.
In one aspect, a hematologic cancer that is to be treated is a mast cell neoplasm. As used herein, the term "mast cell neoplasm" does not encompass multiple myeloma. As used herein, the term "mast cell neoplasm" refers to cancers of mast cells (e.g., mast cell leukemia, malignant mastocytosis).
In one aspect, a hematologic 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 hematologic 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 hematologic 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 hematologic cancer that is to be treated has been typed to identify a familial or spontaneous mutation in p53, Rb, CDKN2A (P16INK4A), FHIT, ATM myc, ras, bcr, abl, or bax. In another aspect, a hematologic cancer that is to be treated is associated with the Philadelphia chromosome translocation [t(9;22)] to form a bcr-abl gene fusion.
In one aspect, a hematologic cancer that is to be treated is associated with a chromosomal translocation selected from the group consisting of t(9;22), t(10;11), t(11;19), t(5;12), t(3;11), t(15;17), t(11;17), and t(5;17). In another aspect, a hematologic cancer that is to be treated is associated with an increased level of a marker selected from the group consisting of beta-2-microglobulin, Zap-70 and CD3~.
In one aspect, a hematologic cancer that is to be treated includes a localized tumor of the hematologic system. In one aspect, a hematologic cancer that is to be treated includes a tumor of the hematologic system that is associated with a negative regional lymph node biopsy. In one aspect, a hematologic cancer that is to be treated includes a tumor of the hematologic system that is associated with a positive regional lymph node biopsy. In another aspect, a hematologic cancer that is to be treated includes a tumor of the hematologic system 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 hematologic cancer that is to be treated includes a tumor of the hematologic system that has metastasized to other locations in the body. In one aspect, a hematologic cancer that is to be treated is classified as having metastasized to a location selected from the group consisting of lymph nodes, liver, lungs, bone marrow, brain, spinal cord, and spleen. In one aspect, a a hematologic cancer that is to be treated is classified as occurnng in a patient with a high white blood cell count (e.g., above 15,000), or a low blood lymphocyte count (e.g., below 600). W another aspect a hematologic 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 hematologic cancer that is to be treated has been staged as Stage 0, I, II, III, or IV. In one aspect, a hematologic cancer that is to be treated has been staged according to the Rai classification system as Rai Stage 0, Rai Stage I, Rai Stage II, Rai Stage III, or Rai Stage IV. In one aspect, a hematologic cancer that is to be treated has been staged according to the Binet classification system as Binet Stage A, Binet Stage B, or Binet Stage C.
In one aspect, a hematologic cancer that is to be treated has been staged as Stage I, IB, IE, II, IIB, IIE, III, IIIB, IIIE, IIIS, IIISE, IV or IVB. In one aspect, In one aspect, a hematologic cancer that is to be treated has been staged as chronic phase, accelerated phase, or blast phase.
In one aspect, a hematologic 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, Tl, T2, T3,T4; and where the regional lymph nodes (N) 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 hematologic 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 hematologic cancer. In another aspect, a hematologic 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 l, Grade 2, Grade 3 or Grade 4.
In one aspect, a hematologic 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 hematologic cancer that is to be treated includes a tumor that has been determined to be from about 2 to about 5 centimeters in diameter. hl another aspect, a hematologic 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. hl another aspect, a hematologic 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 hematologic cancer that is to be treated is classified by microscopic appearance as well differentiated, moderately differentiated, poorly differentiated, or undifferentiated. In another aspect, a hematologic 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 hematologic 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 hematologic 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 hematologic cancer that is to be treated is classified as being aneuploid, triploid, tetraploid, or as having an altered ploidy. In one aspect, a hematologic 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 hematologic cancer that is to be treated has been typed as having less than 5% blast cells. In another aspect, a hematologic cancer that is to be treated has been typed as having more than 5%, 10%, 20%, 30% or 40% blast cells. In another aspect, a hematologic cancer that is to be treated has been typed as having more than 5%, 10%, 20%, 30% or 40%
blast cells and promyelocytes.
In one aspect, a hematologic cancer that is to be treated is evaluated by DNA
cytometry, flow cytometry, or image cytometry. In one aspect, a hematologic cancer that is to be treated has been typed as having 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of cells in the synthesis stage of cell division (e.g., in S phase of cell division). In one aspect, a hematologic 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, "monotherapy" refers to administration of a single active or therapeutic compound to a subj ect in need thereof. Preferably, monotherapy will involve administration of a therapeutically effective amount of an active compound. For example, (3-lapachone monotherapy for cancer comprises administration of a therapeutically effective amount of (3-lapachone, or a pharmaceutically acceptable salt, prodrug, metabolite, analog or derivative thereof, to a subject in need of treatment of cancer. Monotherapy may be contrasted with combination therapy, in which a combination of multiple active compounds is administered, preferably with each component of the combination present in a therapeutically effective amount. In one aspect, (3-lapachone montherapy is more effective than combination therapy in inducing a desired biological effect.
In one aspect, combination therapy includes (3-lapachone with taxol; (3-lapachone with docetaxel; [3-lapachone with vincristin; (3-lapachone with vinblastin; ~3-lapachone with nocodazole; [3-lapachone with teniposide; [3-lapachone with etoposide; (3-lapachone with adriamycin; (3-lapachone with epothilone; (3-lapachone with navelbine; (3-lapachone with camptothecin; (3-lapachone with daunorubicin; (3-lapachone with dactinomycin;
(3-lapachone with mitoxantrone; (3-lapachone with amsacrine; (3-lapachone with epirubicin;
or (3-lapachone with idarubicin. In a preferred aspect, combination therapy includes (3-lapachone with gemcitabine. In another aspect, combination therapy includes reduced (3-lapachone with taxol;
reduced (3-lapachone with docetaxel; reduced (3-lapachone with vincristin;
reduced (3-lapachone with vinblastin; reduced (3-lapachone with nocodazole; reduced [3-lapachone with teniposide;
reduced (3-lapachone with etoposide; reduced (3-lapachone with adriamycin;
reduced (3-lapachone with epothilone; reduced [3-lapachone with navelbine; reduced (3-lapachone with camptothecin; reduced (3-lapachone with daunorubicin; reduced (3-lapachone with dactinomycin;
reduced (3-lapachone with mitoxantrone; reduced (3-lapachone with amsacrine;
reduced (3-lapachone with epirubicin; or reduced (3-lapachone with idarubicin. In a preferred aspect, combination therapy includes reduced [3-lapachone with gemcitabine.
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 compound 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 hematologic 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, 10x, 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 immunofluorescence microscopy.
In another aspect, treating a hematologic 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.
In another aspect, treating a hematologic 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 hematologic 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.
In another aspect, treating a hematologic 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 hematologic 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 survival following initiation of treatment with an active compound. In an 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 hematologic 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 an 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 hematologic cancer of the present invention results in increase in average survival time of a population of treated subj ects in comparison to a population receiving monotherapy with a drug that is not ~3-lapachone, or a pharmaceutically acceptable salt, metabolite, analog or derivative thereof. 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 an 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 hematologic cancer of the present invention results in a decrease in the mortality rate of a population of treated subj ects in comparison to a population receiving carrier alone. In another aspect, treating hematologic 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 hematologic cancer results a decrease in the mortality rate of a population of treated subjects in comparison to a population receiving monotherapy with a drug that is not (3-lapachone, or a pharmaceutically acceptable salt, 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 hematologic 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 tumor diameter per unit time.
In another aspect, treating a hematologic cancer of the present invention results in a decrease in tumor regrowth. Preferably, after treatment, tumor regrowth is less than 5%; 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, 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 SO%;
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.
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.

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, ~i-lapachone, or a pharmaceutically acceptable salt, metabolite, analog or derivative thereof, acts selectively on a cancer or precancer 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 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 50%, 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 isozyrne 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 flftyy 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.
W 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
family of transcription factors (e.g., E2F-l, 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 hematologic cancer or a cell proliferative disorder results in cell death, and preferably, cell death results in a decrease of at least 10% in number 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 (FACS). 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 U S A.
100(5): 2674-~. In a preferred aspect, cell death results from apoptosis.
In a preferred aspect, an effective amount of (3-lapachone, or a pharmaceutically acceptable salt, 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 at a therapeutically effective amount does not induce . apoptosis 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 at a therapeutically effective amount does not induce cell death in greater than 10%
of normal cells.
In one aspect, activating refers to placing one or more compositions 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.
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 (i-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 another 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-~.
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.
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.
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 selectively in hematologic 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 hematologic cancer cells but not in normal cells.
In one aspect, contacting a cell with (3-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 (3-lapachone, or a pharmaceutically acceptable salt, metabolite, analog or derivative thereof, stimulates unscheduled activation of an E2F
transcription factor selectively in hematologic 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 hematologic 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 (i-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 (3-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 /3-lapachone, or a pharmaceutically acceptable salt, metabolite, analog or derivative thereof, results in activation of one or more cell cycle checkpoint pathways.
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 hematologic 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 hematologic 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 of the hematologic system. 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 of the hematologic system.
In a preferred aspect, the present invention relates to a method of treating or preventing cancer by administering (3-lapachone, or a pharmaceutically acceptable salt, metabolite, analog or derivative thereof to a subject in need thereof, where administration of the (3-lapachone, or a pharmaceutically acceptable salt, 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 but not in normal cells, antitumor activity in animals with a therapeutic index of at least 2, and acitvation of a cell cycle checkpoint (e.g., activation or elevation of a member of the E2F family of transcription factors). As used herein, "therapeutic index" is the maximum tolerated dose divided by the efficacious dose.
In additional aspects, (3-lapachone, or a pharmaceutically acceptable salt, metabolite, analog or derivative thereof, can be administered in combination with a chemotherapeutic agent.
Exemplary chemotheraputics with activity against cell proliferative disorders, such as hematologic 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-F~, methotrexate (MTX), TAXOTERE~ (docetaxel), ZOLADEX~ (goserelin), AVASTINTM (bevacizuxnab), vincristin, vinblastin, nocodazole, teniposide, etoposide, epothilone, navelbine, camptothecin, daunonibicin, dactinomycin, 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 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).
In a preferred aspect, a cell proliferative disorder of the hematologic system, such as hematologic cancer, is treated by administering to a patient in need thereof a therapeutically effective amount of (3-lapachone, or a pharmaceutically acceptable salt, metabolite, analog or derivative thereof, in combination with a therapeutically effective amount of gemcitabine.
GEMZAR~ (gemcitabine HCl) is 2'-deoxy-2',2'-difluorocytidine monohydrochloride ((3-isomer), 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 hematologic 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, 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).
As used herein, "leukopenia" is a condition in which the number of white blood cells circulating in the blood is reduced, e.g., a condition in which the white blood cell count (WBC) is below the normal range. In one aspect, in an adult human subject, a normal white blood cell count is between about 5,000 and 11,000 cu per microliter of whole blood. In one aspect, in an adult human subject, leukopenia exists when the white blood cell~count is below about 5,000 cu per microliter. In another aspect, in an adult human subject, mild leukopenia exists when the white blood cell count is between about 3,000 and 5,000 cu per microliter, moderate leukopenia exists when the white blood cell count is between about 1,500 and 3,000 cu per microliter, and severe leukopenia exists when the white blood cell count is less than about 1,500 cu per microliter. White blood cell count may be measured by any reproducible means.
In one aspect, a white blood cell count is measured by a medical diagnostic instrument capable of performing an automated white blood count.
As used herein, "neutropenia" is a condition in which the number of neutrophils circulating in the blood is reduced. In one aspect, in an adult human subject, a normal neutrophil count is between about 1,000 and 1,500 cells per cc3 of whole blood. In one aspect, in an adult human subject, neutropenia exists when the neutrophil count is below about 1,000 cells per cc3.
In another aspect, in an adult human subject, mild neutropenia exists when the neutrophil count is between about 500 and 1,000 cells per cc3, moderate neutropenia exists when the neutrophil count is between about 200 and500 cells per cc3, and severe neutropenia exists when the neutrophil count is less than about 200 cells per cc3. Neutropenia may be assessed by any reproducible means. In one aspect, neutropenia is assessed by performing a white blood count and differential. In one aspect, a differential is performed by counting one hundred white blood cells and identifying them by shape and appearance of nucleus, color and granularity as either neutrophils, bands, lymphocytes, monocytes, eosinophils, basophils, or atypical or immature cells. In one aspect, a white blood cell count is measured by a medical diagnostic instrument capable of performing an automated white blood count. In one aspect, a differential is performed manually using a light microscope. In another aspect, a differential is performed automatically by a medical diagnostic instrument.

As used herein, "monocytopenia" is a condition in which the number of monocytes circulating in the blood is reduced. In one aspect, in an adult human subject, a normal monocyte count is between about 40 and 180 cells per cc3 of whole blood. In one aspect, in an adult human subject, monocytopenia exists when the monocyte count is below about 35 cells per cc3.
In another aspect, monocytopenia exists when the percentage of monocytes is less than about 2% using the differential assay method. Monocytopenia may be assessed by any reproducible means. In one aspect, monocytopenia is assessed by performing a white blood count and differential. In one aspect, a differential is performed by counting one hundred white blood cells and identifying them by shape and appearance of nucleus, color and granularity as either neutrophils, bands, lymphocytes, monocytes, eosinophils, basophils, or atypical or immature cells. In one aspect, a white blood cell count is measured by a medical diagnostic instrument capable of performing an automated white blood count. In one aspect, a differential is performed manually using a light microscope. In another aspect, a differential is performed automatically by a medical diagnostic instrument.
As used herein, "lymphocytopenia" is a condition in which the number of lymphocytes circulating in the blood is reduced. W one aspect, in an adult human subject, a normal lymphocyte count is between about 400 and 1,200 cells per cc3 of whole blood.
In one aspect, in an adult human subject, lymphocytopenia exists when the lymphocyte count is below about 350 cells per cc3. lil another aspect, lylnphocytopenia exists when the percentage of lymphocytes is less than about 20% using the differential assay method. Lymphocytopenia may be assessed by any reproducible means. In one aspect, lymphocytopenia is assessed by performing a white blood count and differential. In one aspect, a differential is performed by counting one hundred white blood cells and identifying them by shape and appearance of nucleus, color and granularity as either neutrophils, bands, lymphocytes, monocytes, eosinophils, basophils, or atypical or immature cells. In one aspect, a white blood cell count is measured by a medical diagnostic instrument capable of performing an automated white blood count. In one aspect, a differential is performed manually using a light microscope. In another aspect, a differential is performed automatically by a medical diagnostic instrument.
W one aspect, an altered profile of immune cells may be assessed by performing a white blood count and differential. In one aspect, in an adult human subject, a normal profile of immune cells includes a total white cell count of between about 5,000 and 11,000 cu per microliter of whole blood; a neutrophil count of between about 1,000 and 1,500 cells per cc3 of whole blood; neutrophils 50-60%; lymphocytes 20-40%; monocytes 2-6%;
eosinophils 1-4%;
and basophils 0.5-1%. In one aspect, an altered profile of immune cells is a profile that differs from a normal profile in one of the stated parameters, or two, three, four, five, six or seven of the stated parameters. In one aspect, a white blood cell count is measured by a medical diagnostic instrument capable of performing an automated white blood count. In one aspect, a differential is performed manually using a light microscope. In another aspect, a differential is performed automatically by a medical diagnostic instrument.
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., Current Protocols in Molecular Biology, John Wiley and Sons, Inc. (2005);
Sambrook et al., Molecular' Cloning, A Laboratory Manual (3d ed.), Cold Spring Harbor Press, Cold Spring Harbor, New York (2000); Coligan et al., Current Protocols in Immunology, John Wiley &
Sons, N.Y.; Enna et al., Current Ps°otocols in Plaarmacology, John Wiley & Sons, N.Y.; Fingl et al., The Plaa~macological Basis of Tlaenapeutics (1975), Remingtofa's Pharmaceutical Sciences, 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 earner.
As used herein, "pharmaceutically acceptable excipient" or "pharmaceutically acceptable earner" 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 earners 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 solution, and 5% human serum albumin. Pharmaceutically acceptable carriers include solid earners such as lactose, terra alba, 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, methylinethacrylate 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 carriers 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 Garner 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 Garner will be those amounts which are effective to obtain a pharmaceutically acceptable formulation in terns 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.
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 G1/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 serum albumin, polyalginic acid or a combination thereof. Examples of water-soluble chelating agents are, but not limited to: D1PA (diethylenetriaminepentaacetic acid), EDTA, DTTP, DOTA or their water-soluble salts, etc. Examples of radionuclides include, but not limited to: 111In, 9oY, 166H0' 68Ga, g9»'Tc, and the like.
Due to the water insolubility of (3-lapachone, pharmaceutical carriers or solubilizing agents may be used to provide sufficient quantities of (3-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 Bootlnnan 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 Gl/S
phase drug, or an analog or derivative 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, 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 (i. 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 adaninistration 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.
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 ethylenediaxninetetraacetic 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, injected 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, 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 clincian. 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 account 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 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 Garner 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 Garner 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 Garners. 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 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 m2, 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.
A compound of the present invention may be administered in combination with an S
phase compound, such as an gemcitabine, in any manner found appropriate by a clinician in generally accepted efficacious dose ranges, such as those described in the Physician's Desk Reference, 59th Edition, Thomson PDR (2005)("PDR"). In general, gemcitabine is administered intravenously at dosages from about 10 mg/m2 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/mz. In an embodiment, gemcitabine is administered intravenously at a dosage from approximately 100 mg/m2 to about 2000 mg/ma. In an embodiment, gemcitabine is administered intravenously at a dosage of approximately 1000 mg/ma. 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.
The S phase drug, such as an gemcitabine, will be administered in a similar regimen with a Gl/S phase drug, such as (3-lapachone or an analog or derivative thereof, although the amounts will preferably be reduced from that normally administered. It is preferred, for example, that the gemcitabine be administered at the same time or after the (3-lapachone has administered to the patient. When the gemcitabine is administered after the (3-lapachone, the gemcitabine 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 Gl/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 G1/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 (i-lapachone or an analog or derivative thereof.
(3-lapachone has been shown to have a variety of pharmacological effects. (3-lapachone has been shown to be a DNA repair inhibitor which sensitizes cells to DNA
damaging agents (Boorstein, R.J., et al., (1984) Biocherra. Biophys. Res. Conarraun., 118:828-834; Boothman, D.A., et al., (1989) J. Cancer 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 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 p,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 ~,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/ma to 5000 mg/m2 per day, more preferably from about 20 mg/m2 to 2000 mg/m2 per day, more preferably from about 20 mg/m2 to 500 mg/m2 per day, most preferably from about 30 to 300 mg/m2 per day. Preferably, 2 mg/m2 to 5000 mg/m2 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 ~.g 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/m2 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 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.
The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration.
EXAMPLES
Example 1 Proliferating human leukemia or lymphoma cells are seeded at 1000 per well in six-well plates and incubated 48 hours. (3-lapachone is added to dishes in less than 5 ~1 of concentrated solution (corresponding to a final DMSO concentration of less that 0.1%). (3-lapachone is dissolved at a concentration of 20 mM in DMSO and diluted in complete media.
Control plates receive the same volume of DMSO alone. After 1-4 hours exposure, cells are rinsed and drug-free medium may be added. Cultures are left undisturbed for 10-20 days to allow for colony formation and then may be fixed and stained with modified Wright-Giemsa stain (Sigma).
Colonies of greater than 30 cells are scored as survivors. Cells are maintained at 37 °C in 5%
C02 in complete humidity.
Alternatively, cell death of human leukemia or lymphoma cells cultured in the absence or presence of (3-Lapachone (e.g., at 2, 4, 8, and 20 ~M) for one to 24 hours is measured by MTT
assay. Briefly, the MTT assay is performed by plating in a 96-well plate at 10,000 cells per well, culturing for 48 hours in complete growth medium, treating with (3-Lapachone for one to 24 hours, and cultured with drug-free medium for 24 hours. MTT solution is added to the culture medium, and after 2 hours, optical density may be read with an ELISA
reader.
Alternatively, cell death of human leukemia or lymphoma cells cultured in the absence or presence of 13-Lapachone is measured by fluorescence activated cell sorting (FACS) analysis.
Alternatively, cell death of human leukemia or lymphoma cells cultured in the absence or presence of (i-Lapachone is measured by methods described in Li et al., (2003) Proc Natl Acad Sci USA. 100(5): 2674-8.

Example 2 (3-lapachone is tested in the NCI in vitro screen of 60 cancer cell lines, which allows comparison with other anti-tumor agents under standardized conditions. The NCI
assays are performed under standardized conditions not designed to mimic the conditions of dosing and use the sulforhodamine B assay as the endpoint. (i-lapachone is broadly active against many cell types, with LCSO (1og10 molar concentration causing 50% lethality) between -4.5 and -5.3, and mean of -5.07 across all cells. The NCI set of 60 lines includes at least four leukemia cell lines, CCRF-CEM (acute lymphoblastic leukemia); HL-60 (acute myeloid leukemia); K562 (chronic myelogenous leukemia); MOLT-4 (acute lymphoblastic leukemia); and one lymphoma cell line SR (lymphoid lymphoma). The NCI set of 60 lines also includes cell line RPMI-8226 (multiple myeloma). When compared to many FDA approved chemotherapeutic agents for common cancer types with publicly available data, no approved drug exceeds the mean of (3-lapachone across all cells and only mitoxantrone equals it. See, e.g., Figure 2.

Claims (39)

What is claimed is:
1. A method of treating a cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm, comprising administering to a subject in need thereof a therapeutically effective amount of .beta.-lapachone, or a pharmaceutically acceptable salt thereof, in combination with a pharmaceutically acceptable carrier, wherein said cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm is treated.
2. The method according to claim 1, wherein said cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm is mast cell neoplasm.
3. The method according to claim 1, wherein said administering to a subject in need thereof a therapeutically effective amount of said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, results in activation of a cell cycle checkpoint.
4. The method according to claim 1, wherein said administering to a subject in need thereof a therapeutically effective amount of said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, results in modulation of an activity of E2F.
5. The method according to claim 1, wherein said administering to a subject in need thereof a therapeutically effective amount of said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, induces cell death in said cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm.
6. The method according to claim 5, wherein said cell death is apoptosis.
7. The method according to claim 1, wherein said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, is administered parenterally.
8. The method according to claim 1, wherein said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, is administered by injection.
9. The method according to claim 1, wherein said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, is administered intravenously.
10. The method according to claim 1, wherein said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, is administered orally.
11. A method of treating lymphoma comprising administering to a subject in need thereof a therapeutically effective amount of .beta.-lapachone, or pharmaceutically acceptable salt thereof, in combination with a pharmaceutically acceptable carrier, wherein said lymphoma is treated.
12. The method according to claim 11, wherein said administering to a subject in need thereof a therapeutically effective amount of said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, results in activation of a cell cycle checkpoint.
13. The method according to claim 11, wherein said administering to a subject in need thereof a therapeutically effective amount of said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, results in modulation of an activity of E2F.
14. The method according to claim 11, wherein said administering to a subject in need thereof a therapeutically effective amount of said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, induces cell death in said lymphoma.
15. The method according to claim 14, wherein said cell death is apoptosis.
16. The method according to claim 11, wherein said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, is administered parenterally.
17. The method according to claim 11, wherein said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, is administered by injection.
18. The method according to claim 11, wherein said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, is administered intravenously.
19. The method according to claim 11, wherein said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, is administered orally.
20. A method of treating leukemia comprising administering to a subject in need thereof a therapeutically effective amount of .beta.-lapachone, or pharmaceutically acceptable salt thereof, in combination with a pharmaceutically acceptable carrier, wherein said leukemia is treated.
21. The method according to claim 20, wherein said administering to a subject in need thereof a therapeutically effective amount of said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, results in activation of a cell cycle checkpoint.
22. The method according to claim 20, wherein said administering to a subject in need thereof a therapeutically effective amount of said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, results in modulation of an activity of E2F.
23. The method according to claim 20, wherein said administering to a subject in need thereof a therapeutically effective amount of said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, induces cell death in said leukemia.
24. The method according to claim 23, wherein said cell death is apoptosis.
25. The method according to claim 20, wherein said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, is administered parenterally.
26. The method according to claim 20, wherein said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, is administered by injection.
27. The method according to claim 20, wherein said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, is administered intravenously.
28. The method according to claim 20, wherein said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, is administered orally.
29. A method of treating myeloid neoplasms comprising administering to a subject in need thereof a therapeutically effective amount of .beta.-lapachone, or pharmaceutically acceptable salt thereof, in combination with a pharmaceutically acceptable carrier, wherein said myeloid neoplasms are treated.
30. The method according to claim 29, wherein said administering to a subject in need thereof a therapeutically effective amount of said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, results in activation of a cell cycle checkpoint.
31. The method according to claim 29, wherein said administering to a subject in need thereof a therapeutically effective amount of said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, results in modulation of an activity of E2F.
32. The method according to claim 29, wherein said administering to a subject in need thereof a therapeutically effective amount of said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, induces cell death in said myeloid neoplasms.
33. The method according to claim 32, wherein said cell death is apoptosis.
34. The method according to claim 29, wherein said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, is administered parenterally.
35. The method according to claim 29, wherein said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, is administered by injection.
36. The method according to claim 29, wherein said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, is administered intravenously.
37. The method according to claim 29, wherein said .beta.-lapachone, or a pharmaceutically acceptable salt thereof, is administered orally.
38. A method for inducing cell death in a cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm, comprising contacting said cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm with an effective amount of .beta.-lapachone, or a pharmaceutically acceptable salt thereof, wherein said contacting induces said cell death in said a cancer selected from the group consisting of lymphoma, leukemia, myeloid neoplasms, and mast cell neoplasm.
39. The method according to claim 38, wherein said cell death is apoptosis.
CA002556789A 2004-02-20 2005-02-18 Use of beta-lapachone for treating hematologic tumors Abandoned CA2556789A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US54591404P 2004-02-20 2004-02-20
US60/545,914 2004-02-20
PCT/US2005/005646 WO2005082357A1 (en) 2004-02-20 2005-02-18 Use of beta-lapachone for treating hematologic tumors

Publications (1)

Publication Number Publication Date
CA2556789A1 true CA2556789A1 (en) 2005-09-09

Family

ID=36168365

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002556789A Abandoned CA2556789A1 (en) 2004-02-20 2005-02-18 Use of beta-lapachone for treating hematologic tumors

Country Status (3)

Country Link
JP (1) JP2007523191A (en)
CA (1) CA2556789A1 (en)
WO (1) WO2005082357A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050187288A1 (en) * 2004-02-20 2005-08-25 Chiang Li Beta-lapachone and methods of treating cancer
CA2583700A1 (en) 2004-08-11 2006-02-23 Arqule, Inc. Quinone prodrug compositions and methods of use
US8614228B2 (en) 2004-08-11 2013-12-24 Arqule, Inc. Quinone prodrug compositions and methods of use
EP2152686B1 (en) 2007-04-30 2014-12-17 ArQule, Inc. Hydroxy sulfonate of quinone compounds and their uses
US20120142650A1 (en) * 2010-12-07 2012-06-07 John Hollingsworth Method for treating lung disease

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4244141B2 (en) * 2000-11-07 2009-03-25 デイナ ファーバー キャンサー インスティチュート,インコーポレイテッド Methods for treating blood tumors and blood cancers
TW200510367A (en) * 2002-11-18 2005-03-16 Arqule Inc Novel lapachone compounds, their preparation, and the use thereof
AU2003293333A1 (en) * 2002-12-02 2004-06-23 Arqule, Inc. Method of treating cancers

Also Published As

Publication number Publication date
WO2005082357A8 (en) 2006-04-20
JP2007523191A (en) 2007-08-16
WO2005082357A1 (en) 2005-09-09

Similar Documents

Publication Publication Date Title
US20080146584A1 (en) Method of treating cancers using beta-lapachone or analogs or derivatives thereof
US6214821B1 (en) Methods and composition for the inhibition of cancer cells
Bashash et al. Anti-tumor activity of PI3K-δ inhibitor in hematologic malignant cells: shedding new light on resistance to Idelalisib
CA2556789A1 (en) Use of beta-lapachone for treating hematologic tumors
Ku et al. Phase II trial of sequential paclitaxel and 1 h infusion of bryostatin-1 in patients with advanced esophageal cancer
An et al. PDE3A inhibitor anagrelide activates death signaling pathway genes and synergizes with cell death-inducing cytokines to selectively inhibit cancer cell growth
EP1722776A2 (en) Use of beta-lapachone as a broad spectrum anti-cancer agent
EP1722775A1 (en) Use of beta-lapachone for treating hematologic tumors
US20050197405A1 (en) Treatment of hematologic tumors and cancers with beta-lapachone, a broad spectrum anti-cancer agent
EP2033638A2 (en) Beta-lapachone for the treatment of pancreatic cancer
CA2556759A1 (en) Beta-lapachone and s-phase drug combinations for cancer treatment
US20050192360A1 (en) Method of treatment of pancreatic cancer
Gupta et al. In vitro synergism between LFA-1 targeting leukotoxin (Leukothera™) and standard chemotherapeutic agents in leukemia cells
Parekh et al. Circumvention of adriamycin resistance: effect of 2-methyl-1, 4-naphthoquinone (vitamin K3) on drug cytotoxicity in sensitive and MDR P388 leukemia cells
US20050197406A1 (en) Method of treatment of lung cancer
Liu et al. TCH-1030 targeting on topoisomerase I induces S-phase arrest, DNA fragmentation, and cell death of breast cancer cells
US20050192247A1 (en) Method of treating cancers
EP2033639A2 (en) Beta-lapachone for the treatment of colon cancer
US20050222246A1 (en) Beta-lapachone is a broad spectrum anti-cancer agent
US20050192361A1 (en) Method of treatment of colon cancer
US10550130B2 (en) Benzo-thiazolo-imidazole compounds and uses thereof
Viladkar et al. In vitro Effects of Pentoxifylline and Doxorabicin on Cell Survival and DNA Damage in Sensitive and MDRP388 Leukemia Cells
ITRM20130312A1 (en) EXTRACT OF CYNARA SPP. AND ITS USES.

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued
FZDE Discontinued

Effective date: 20091229