WO2000018234A1 - Thiazolidenediones alone or in combination with other therapeutic agents for tumor therapy - Google Patents

Thiazolidenediones alone or in combination with other therapeutic agents for tumor therapy Download PDF

Info

Publication number
WO2000018234A1
WO2000018234A1 PCT/US1998/020611 US9820611W WO0018234A1 WO 2000018234 A1 WO2000018234 A1 WO 2000018234A1 US 9820611 W US9820611 W US 9820611W WO 0018234 A1 WO0018234 A1 WO 0018234A1
Authority
WO
WIPO (PCT)
Prior art keywords
troglitazone
cells
ppar
cell proliferation
treatment
Prior art date
Application number
PCT/US1998/020611
Other languages
French (fr)
Inventor
John Alton Copland, Iii
Slavisa Gasic
Randall Urban
Melvyn Soloff
Original Assignee
Board Of Regents, The University Of Texas System
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 Board Of Regents, The University Of Texas System filed Critical Board Of Regents, The University Of Texas System
Priority to PCT/US1998/020611 priority Critical patent/WO2000018234A1/en
Publication of WO2000018234A1 publication Critical patent/WO2000018234A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca

Definitions

  • Troglitazone is a member of the thiazolidinedione class of drugs which act as ligands to activate the transcriptional factor, PPAR- ⁇ .
  • Ligands of PPAR- ⁇ have previously been shown to cause terminal differentiation in human liposarcoma cells in vitro (Tontonoz et al., 1997).
  • Another example of differentiation therapy is the use of all-trans retinoic acid in the treatment of myelocytic lineage derived cancers such as acute promyelocytic leukemia (Warrel et al., 1993). Induction of terminal differentiation of malignant cells is an ideal mechanism of curing cancer as opposed to a cell death mediated mechanisms.
  • PPAR- ⁇ plays a central role in the process of adipocyte differentiation and is expressed in high levels in this tissue.
  • PPAR- ⁇ heterodimerizes with retinoid X receptor (RXR- ⁇ ) to form a DNA-binding complex (DR-1) that regulates expression of adipocyte- specific genes (Kliewer et al., 1992; Tontonoz et al, 1994a; Tontonoz et al, 1994b; Tontonoz et al, 1995; Sears et al, 1996).
  • troglitazone is an antioxidant due to the vitamin E moiety in its structure.
  • Vitamin E has recently been shown to induce apoptosis in human colorectal cancer cell lines (Chinery et al, 1997). However, the IC 50 dose of vitamin E for HCT 15 cells was 0.75 mM and for HCT 116 cells was 3 mM.
  • Combinatorial therapy with 5-FU and 3 mM vitamin E reduces the IC 50 of 5-FU alone by 2 and 8 fold respectively in HCT 116 and HCT15 cells.
  • the present inventors demonstrate an increase in p21 expression and conclude that vitamin E's inhibitory activity is due to the increase in the cell cycle inhibitor, p21.
  • Saos-2 cells are an osteogenic sarcoma characterized by a mutant nonfunctional retinoma blastoma (Rb) protein and no p53 protein expression. Both of these proteins play a critical role in regulating cell cycle progression. Loss of function of these two proteins is a partial explanation for the developed resistance to chemotherapeutic agents used to treat patients.
  • the retinoblastoma gene product (pRb) is a critical substrate of the evolutionarily conserved cyclin-dependent kinases (CDK's) which regulate progression through the cell cycle.
  • CDKs cyclin dependent kinases
  • the CDKs regulate biochemical pathways or checkpoints which integrate mitogenic and growth inhibitory signals, monitor chromosome integrity, and coordinate cell cycle transitions. Passage through Gl and S phase is regulated by the activities of cyclin
  • E/CDK2, cyclin D/CDK4, and cyclin D/CDK6 E/CDK2, cyclin D/CDK4, and cyclin D/CDK6.
  • Cyclin A/CDK2 promotes passage through the S phase and cyclin B/CDK2 kinase is essential for G2/mitosis transition.
  • Two families of CDK inhibitors (CKIs) mediate cell cycle arrest following growth inhibitory stimuli.
  • the INK4 family members pl5INK4b/MTS2, pl6LNK4/MTSl, pl8, and pl9 bind CDK4 and CDK6 specifically and inhibit cyclin D binding.
  • CKIs bind CDKs inhibiting progression through the cell cycle. Phosphorylation of CKIs lead to degradation of
  • FIG. 1 A DNA concentration of Saos-2 cells 3 days after a single treatment of troglitazone.
  • FIG. 1 A shows DNA content per well following a single treatment with troglitazone. * Denotes different from control (P ⁇ 0.05).
  • FIG. IB shows lactate dehydrogenase (LDH) levels as measured in the media after 3 days of treatment.
  • FIG. 2 - CAT activity in Saos-2 cells transfected with AOX-PPRE-tk-CAT construct.
  • FIG. 2 shows that troglitazone treatment increase CAT activity 4 fold in the absence of transfected PPAR- ⁇ (lane 2 vs lane 1) as well as the presence of transfected PPAR (lanes 3 and 4). This demonstrates the existence of functional
  • FIG. 3 3H-Thymidine incorporation into Saos-2 cells.
  • FIG. 3 shows that a dose of 5 ⁇ g/ml troglitazone caused a 16% reduction of 3H-uridine uptake while a 10 ⁇ g/ml dose caused a 43% reduction. * Denotes statistically different from control (P ⁇ 0.05).
  • FIG. 4 - mRNA levels in Saos-2 cells shows the effect upon mRNA levels for cell cycle inhibitor genes in response to a single treatment of 1 ⁇ M troglitazone for 24 hr. This revealed a 2.6 fold increase in p21 mRNA levels. Levels were normalized to GAPDH. Another control used is L32 which is seen to change very little, pi 8 is shown not change, as well.
  • FIG. 5 DNA content of Saos-2 cells 3 days after a single treatment of troglitazone and/or 5-fluorouracil.
  • FIG. 5 shows the effects of toxic chemotherapeutic agent, 5-fluorouracil (5-FU), revealing that combinatorial therapy with 10 ⁇ g/ml troglitazone and 1 ⁇ M 5-FU was equivalent to 100 ⁇ M dose of 5-FU alone. Combinatorial therapy reduces the dose of 5-FU needed by a factor of 100.
  • * Denotes different from control (P ⁇ 0.05); + Denotes different from corresponding 5-fluorouracil treatment (P ⁇ 0.05).
  • FIG. 6 DNA content of Saos-2 cells.
  • FIG.6 shows that troglitazone demonstrates a similar lowering of the dose of doxorubicin needed to inhibit cell proliferation. * Denotes P ⁇ 0.05 as compared to control; + denotes P ⁇ 0.05 as troglitazone/doxorubicin compared to equivalent doxorubicin dose of doxorubicin. DNA was isolated from cells 4 days after a single treatment with the indicated drug.
  • FIG. 7 DNA content of Saos-2 cells.
  • FIG. 7 shows that troglitazone may be acting or acting additionally through its vitamin E antioxidant properties in addition to its ability to bind to PPAR- ⁇ .
  • FIG. 8 DNA content of Saos-2 cells.
  • FIG.8 shows the results of combinatorial therapy with 5-flourouracil (5-FU) or doxorubicin. P ⁇ 0.05 as compared to match contol with/without 5-fluorouracil (5-FU).
  • FIG. 9 DNA content of Saos-2 cells.
  • FIG 9 shows that the thiazolidinediones had no additional inhibitory effect on cell proliferation when administered with methotrexate but did with doxorubicin. * Denotes P ⁇ 0.05 as compared to its doxorubicin control.
  • One purpose of the present invention is to identify and/or develop nontoxic or low toxic chemotherapeutic regimens to treat cancer.
  • the cells used are derived from an osteosarcoma from an eleven year girl who died after aggressive chemotherapeutic treatment with adriamycin, vincristine, cytoxan, and aramycin-C.
  • Troglitazone prototypical thiazolidinedione is used clinically to treat Type II diabetics and has been shown to have little toxic activity except in rare instances of idiopathic hepatic toxicity.
  • This invention demonstrates that troglitazone alone is not toxic and inhibits cell proliferation in Saos-2cells by inhibiting cell cycle progression.
  • Another important discovery described herein is that combinatorial therapy using troglitazone requires 100 fold less toxic chemotherapeutic agent 5- fluorouracil (5-FU) for equivalent inhibition of cell proliferation (as compared to 5- FU treatment alone).
  • 5-FU is toxic to cells because it is a DNA base analog which inhibits DNA synthesis.
  • This latter discovery could prove to be a powerful technique allowing the use of lower concentrations of toxic chemotherapeutic agents while providing successful therapeutic results in causing tumor regression and curing cancer.
  • troglitazone in inhibiting tumor cell growth in osteosarcoma cells. It is also novel to use the nontoxic troglitazone in combination with powerful and potentially toxic chemotherapeutic agents for the purpose of using lower doses of the toxic agent to effectively cause tumor regression.
  • Other thiazolidinediones usable in the practice of the present invention are seen in U.S. Patent No. 5,478,852, issued December 26, 1995 and assigned to Sankyo
  • troglitazone alone would inhibit cell proliferation of human osteosarcoma cells.
  • One known mechanism of action of troglitazone is its action through binding to the peroxisome proliferator activated receptor ⁇ (PPAR- ⁇ ).
  • PPAR- ⁇ is a transcriptional factor that can cause terminal differentiation of adipocytes.
  • PPAR expression has not been previously demonstrated in osteosarcomas. It is also not obvious that troglitazone would lower the IC 50 of toxic chemotherapeutic compounds when administered in combination with these toxic agents.
  • the inventive steps described herein include demonstrating that troglitazone inhibits cell proliferation in human osteosarcoma cells, the presence of functional PPAR- ⁇ in such cells, and the demonstration of the ability of troglitazone to lower the IC 50 of other chemotherapeutic agents when administered in combination therewith as regarding cell proliferation
  • FIG. 1A shows DNA content per well following a single treatment with troglitazone. Cells were exposed for 3 days and cell number determined by measuring DNA content. Cells were exposed for 3 days and cell number determined by measuring DNA content. The IC 50 for troglitazone is between about
  • FIG. IB shows lactate dehydrogenase (LDH) levels measured in the media after 3 days of treatment.
  • LDH levels are indicative of cell death either through apoptosis or necrosis.
  • LDH levels are not altered at any concentration of troglitazone used, as compared to controls, indicating that troglitazone, while not being toxic to cells, inhibits cell proliferation.
  • FIG.2 shows troglitazone treatment to increase CAT expression 4-fold (lane 2 vs lane 1) in the absence of transfected PPAR- ⁇ as well as the presence of 0.67ug transfected PPAR (lanes 3 and 4) thus first demonstrating the existence of functional PPAR in Saos-2 cells.
  • 3-H uridine incorporation was used to determine whether troglitazone inhibited cell cycle progression.
  • Cells 80% confluent were treated with troglitazone for 18 hours and examined for 3H-uridine uptake into cells lysates.
  • a dose of 5 ⁇ g/ml troglitazone caused a 16% inhibition of 3H- uridine uptake while a 10 ⁇ g/ml dose caused a 43% inhibition.
  • FIG. 4 shows the effect upon mRNA levels for cell cycle inhibitor genes in response to a single treatment of 1 ⁇ M troglitazone for 24 hr which revealed a 2.6 fold increase in p21 mRNA levels. Levels were normalized to GAPDH. Another control used is L32 which is seen to change very little, pi 8 is shown not change. Other cell cycle inhibitor genes demonstrating little change in response to troglitazone treatment include pi 30, retinoma blastoma, pi 07, p27, pi 6, and -15.
  • FIG. 5 shows the effects of toxic chemotherapeutic agent, 5-fluorouracil (5- FU), revealing that combinatorial therapy with 10 ⁇ g/ml troglitazone and 1 ⁇ M 5-
  • FU was equivalent to 100 ⁇ M dose of 5-FU alone. Combinatorial therapy reduces the dose of 5-FU needed by a factor of 100.

Landscapes

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

Abstract

A purpose of the present invention is to identify and/or develop nontoxic or low toxic chemotherapeutic regiments to treat cancer. Prototype cells tested are derived from an osteosarcoma from an eleven year girl who died after aggressive chemotherapeutic treatment with adriamycin, vincristine, cytoxan, and aramycin-C. The inventive steps described herein include demonstrating that troglitazone inhibits cell proliferation in human osteosarcoma cells, the presence of functional PPAR-η in such cells, and the demonstration of the ability of troglitazone to lower the IC50 of other chemotherapeutic agents such as 5-fluorouracil and doxorubicin when administered in combination therewith as regarding cell proliferation.

Description

DESCRIPTION
THIAZOLIDENEDIONES ALONE OR IN COMBINATION WITH OTHER THERAPEUTIC AGENTS FOR TUMOR THERAPY
BACKGROUND OF THE INVENTION
Troglitazone is a member of the thiazolidinedione class of drugs which act as ligands to activate the transcriptional factor, PPAR-γ. Ligands of PPAR-γ have previously been shown to cause terminal differentiation in human liposarcoma cells in vitro (Tontonoz et al., 1997). Another example of differentiation therapy is the use of all-trans retinoic acid in the treatment of myelocytic lineage derived cancers such as acute promyelocytic leukemia (Warrel et al., 1993). Induction of terminal differentiation of malignant cells is an ideal mechanism of curing cancer as opposed to a cell death mediated mechanisms. PPAR-γ plays a central role in the process of adipocyte differentiation and is expressed in high levels in this tissue. Mechanistically, PPAR-γ heterodimerizes with retinoid X receptor (RXR- α) to form a DNA-binding complex (DR-1) that regulates expression of adipocyte- specific genes (Kliewer et al., 1992; Tontonoz et al, 1994a; Tontonoz et al, 1994b; Tontonoz et al, 1995; Sears et al, 1996). The combination of PPAR-γ ligand (pioglitazone) and RXR ligand (LG268) treatment of liposarcoma cells in vitro result in an additive effect upon terminal differentiation as characterized by accumulation of intracellular lipid, induction of adipocyte-specific genes, and withdrawal from the cell cycle (Tontonoz, 1997). To date, other tissues expressing PPAR-γ include muscle, kidney, liver, and lung, (Tontonoz, 1997).
Besides its ability to bind to PPAR-γ, troglitazone is an antioxidant due to the vitamin E moiety in its structure. Vitamin E has recently been shown to induce apoptosis in human colorectal cancer cell lines (Chinery et al, 1997). However, the IC50 dose of vitamin E for HCT 15 cells was 0.75 mM and for HCT 116 cells was 3 mM. Combinatorial therapy with 5-FU and 3 mM vitamin E reduces the IC50 of 5-FU alone by 2 and 8 fold respectively in HCT 116 and HCT15 cells. The present inventors demonstrate an increase in p21 expression and conclude that vitamin E's inhibitory activity is due to the increase in the cell cycle inhibitor, p21.
Saos-2 cells are an osteogenic sarcoma characterized by a mutant nonfunctional retinoma blastoma (Rb) protein and no p53 protein expression. Both of these proteins play a critical role in regulating cell cycle progression. Loss of function of these two proteins is a partial explanation for the developed resistance to chemotherapeutic agents used to treat patients. The retinoblastoma gene product (pRb) is a critical substrate of the evolutionarily conserved cyclin-dependent kinases (CDK's) which regulate progression through the cell cycle. Progression through the cell cycle is governed by a family of cyclin dependent kinases (CDKs), whose activity is governed by phosphorylation activated by binding of cyclins, and inhibited by the cdk inhibitors (reviewed in Sherr). The CDKs regulate biochemical pathways or checkpoints which integrate mitogenic and growth inhibitory signals, monitor chromosome integrity, and coordinate cell cycle transitions. Passage through Gl and S phase is regulated by the activities of cyclin
E/CDK2, cyclin D/CDK4, and cyclin D/CDK6. Cyclin A/CDK2 promotes passage through the S phase and cyclin B/CDK2 kinase is essential for G2/mitosis transition. Two families of CDK inhibitors (CKIs) mediate cell cycle arrest following growth inhibitory stimuli. The INK4 family members pl5INK4b/MTS2, pl6LNK4/MTSl, pl8, and pl9 bind CDK4 and CDK6 specifically and inhibit cyclin D binding. Members of the KIP family are currently composed of p21KIPl/WAFl/SDII (transcriptional target of p53), p27KIPl, p57KIP2 (reviewed in Harper et al.). These molecules specifically target CDKs that are important to Gl-S transition: CDK2, CDK4, and CDK6. Thus, CKIs bind CDKs inhibiting progression through the cell cycle. Phosphorylation of CKIs lead to degradation of
CKI and to cell cycle progression. Conversely loss of expression of key CKI(s) can lead to continual cell cycle progression. Compounds that can increase CKI levels are potential chemotherapeutic agents against cancer. BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 A - DNA concentration of Saos-2 cells 3 days after a single treatment of troglitazone. FIG. 1 A shows DNA content per well following a single treatment with troglitazone. * Denotes different from control (P<0.05).
FIG. IB - LDH activity in Saos-2 Cells treated with troglitazone for 3 days.
FIG. IB shows lactate dehydrogenase (LDH) levels as measured in the media after 3 days of treatment.
FIG. 2 - CAT activity in Saos-2 cells transfected with AOX-PPRE-tk-CAT construct. FIG. 2 shows that troglitazone treatment increase CAT activity 4 fold in the absence of transfected PPAR-γ (lane 2 vs lane 1) as well as the presence of transfected PPAR (lanes 3 and 4). This demonstrates the existence of functional
PPAR-γ in Saos-2 cells. * Denotes P<0.05 compared to appropriate control.
FIG. 3 - 3H-Thymidine incorporation into Saos-2 cells. FIG. 3 shows that a dose of 5 μg/ml troglitazone caused a 16% reduction of 3H-uridine uptake while a 10 μg/ml dose caused a 43% reduction. * Denotes statistically different from control (P<0.05).
FIG. 4 - mRNA levels in Saos-2 cells. FIG. 4 shows the effect upon mRNA levels for cell cycle inhibitor genes in response to a single treatment of 1 μM troglitazone for 24 hr. This revealed a 2.6 fold increase in p21 mRNA levels. Levels were normalized to GAPDH. Another control used is L32 which is seen to change very little, pi 8 is shown not change, as well.
FIG. 5 - DNA content of Saos-2 cells 3 days after a single treatment of troglitazone and/or 5-fluorouracil. FIG. 5 shows the effects of toxic chemotherapeutic agent, 5-fluorouracil (5-FU), revealing that combinatorial therapy with 10 μg/ml troglitazone and 1 μM 5-FU was equivalent to 100 μM dose of 5-FU alone. Combinatorial therapy reduces the dose of 5-FU needed by a factor of 100. * Denotes different from control (P<0.05); + Denotes different from corresponding 5-fluorouracil treatment (P<0.05).
FIG. 6 - DNA content of Saos-2 cells. FIG.6 shows that troglitazone demonstrates a similar lowering of the dose of doxorubicin needed to inhibit cell proliferation. * Denotes P<0.05 as compared to control; + denotes P<0.05 as troglitazone/doxorubicin compared to equivalent doxorubicin dose of doxorubicin. DNA was isolated from cells 4 days after a single treatment with the indicated drug.
FIG. 7 - DNA content of Saos-2 cells. FIG. 7 shows that troglitazone may be acting or acting additionally through its vitamin E antioxidant properties in addition to its ability to bind to PPAR-γ.
FIG. 8 - DNA content of Saos-2 cells. FIG.8 shows the results of combinatorial therapy with 5-flourouracil (5-FU) or doxorubicin. P<0.05 as compared to match contol with/without 5-fluorouracil (5-FU).
FIG. 9 DNA content of Saos-2 cells. FIG 9 shows that the thiazolidinediones had no additional inhibitory effect on cell proliferation when administered with methotrexate but did with doxorubicin. * Denotes P<0.05 as compared to its doxorubicin control.
DESCRIPTION OF THE INVENTION
One purpose of the present invention is to identify and/or develop nontoxic or low toxic chemotherapeutic regimens to treat cancer. The cells used are derived from an osteosarcoma from an eleven year girl who died after aggressive chemotherapeutic treatment with adriamycin, vincristine, cytoxan, and aramycin-C.
Troglitazone (prototypical thiazolidinedione) is used clinically to treat Type II diabetics and has been shown to have little toxic activity except in rare instances of idiopathic hepatic toxicity. This invention demonstrates that troglitazone alone is not toxic and inhibits cell proliferation in Saos-2cells by inhibiting cell cycle progression. Another important discovery described herein is that combinatorial therapy using troglitazone requires 100 fold less toxic chemotherapeutic agent 5- fluorouracil (5-FU) for equivalent inhibition of cell proliferation (as compared to 5- FU treatment alone). 5-FU is toxic to cells because it is a DNA base analog which inhibits DNA synthesis. Thus, its mechanism of action differs from that of troglitazone. This latter discovery could prove to be a powerful technique allowing the use of lower concentrations of toxic chemotherapeutic agents while providing successful therapeutic results in causing tumor regression and curing cancer.
It is novel to use troglitazone in inhibiting tumor cell growth in osteosarcoma cells. It is also novel to use the nontoxic troglitazone in combination with powerful and potentially toxic chemotherapeutic agents for the purpose of using lower doses of the toxic agent to effectively cause tumor regression. Other thiazolidinediones usable in the practice of the present invention are seen in U.S. Patent No. 5,478,852, issued December 26, 1995 and assigned to Sankyo
Company, Limited, Tokyo, Japan.
It was not apparent that troglitazone alone would inhibit cell proliferation of human osteosarcoma cells. One known mechanism of action of troglitazone is its action through binding to the peroxisome proliferator activated receptor γ (PPAR-γ). PPAR-γ is a transcriptional factor that can cause terminal differentiation of adipocytes. PPAR expression has not been previously demonstrated in osteosarcomas. It is also not obvious that troglitazone would lower the IC50 of toxic chemotherapeutic compounds when administered in combination with these toxic agents.
The inventive steps described herein include demonstrating that troglitazone inhibits cell proliferation in human osteosarcoma cells, the presence of functional PPAR-γ in such cells, and the demonstration of the ability of troglitazone to lower the IC50 of other chemotherapeutic agents when administered in combination therewith as regarding cell proliferation
FIG. 1A shows DNA content per well following a single treatment with troglitazone. Cells were exposed for 3 days and cell number determined by measuring DNA content. Cells were exposed for 3 days and cell number determined by measuring DNA content. The IC50 for troglitazone is between about
1 μg/ml and about 5 μg/ml.
FIG. IB shows lactate dehydrogenase (LDH) levels measured in the media after 3 days of treatment. LDH levels are indicative of cell death either through apoptosis or necrosis. LDH levels are not altered at any concentration of troglitazone used, as compared to controls, indicating that troglitazone, while not being toxic to cells, inhibits cell proliferation.
Examination of the mechanism of action of troglitazone was performed by using the reporter construct AOX-PPRE-tk-CAT. This expression vector contains a DNA sequence which specifically binds PPAR-γ thereby activating chloramphenicol acetyltransferase (CAT) expression. FIG.2 shows troglitazone treatment to increase CAT expression 4-fold (lane 2 vs lane 1) in the absence of transfected PPAR-γ as well as the presence of 0.67ug transfected PPAR (lanes 3 and 4) thus first demonstrating the existence of functional PPAR in Saos-2 cells.
Northern analysis of PPARγ revealed high expression in rat adipocytes, moderate levels in fibroblasts and osteoblasts and low levels in other tissues. (Figure 2B)
3-H uridine incorporation was used to determine whether troglitazone inhibited cell cycle progression. Cells (80% confluent) were treated with troglitazone for 18 hours and examined for 3H-uridine uptake into cells lysates. As shown in figure 3, a dose of 5 μg/ml troglitazone caused a 16% inhibition of 3H- uridine uptake while a 10 μg/ml dose caused a 43% inhibition.
FIG. 4 shows the effect upon mRNA levels for cell cycle inhibitor genes in response to a single treatment of 1 μM troglitazone for 24 hr which revealed a 2.6 fold increase in p21 mRNA levels. Levels were normalized to GAPDH. Another control used is L32 which is seen to change very little, pi 8 is shown not change. Other cell cycle inhibitor genes demonstrating little change in response to troglitazone treatment include pi 30, retinoma blastoma, pi 07, p27, pi 6, and -15.
FIG. 5 shows the effects of toxic chemotherapeutic agent, 5-fluorouracil (5- FU), revealing that combinatorial therapy with 10 μg/ml troglitazone and 1 μM 5-
FU was equivalent to 100 μM dose of 5-FU alone. Combinatorial therapy reduces the dose of 5-FU needed by a factor of 100.
In FIG. 6, troglitazone demonstrates a similar lowering of the dose of doxorubicin needed to inhibit cell proliferation. Note that troglitazone is being administered in μM quantities as opposed to μg/ml (10 μg/ml = 22.7 μM).
Examining compounds similar to troglitazone for inhibitory activity upon cell proliferation should further delineate the mechanism of action of troglitazone. The thiazolidinediones, pioglitazone and BRL49653 have a 10-100 fold greater affinity for PPAR-γ but do not contain the vitamin E moiety in their structures. Comparing these compounds as well as vitamin E succinate revealed that troglitazone was much more potent in inhibiting cell proliferation than the other thiozolidinediones after a single treatment (FIG. 7). Vitamin E succinate demonstrates a dose responsive inhibition of cell proliferation somewhat less effective than troglitazone.. At 100 μM vitamin E, cells are being killed as evidenced by microscopic examination and LDH levels in the media. This effect appears specific since 100 μM succinic acid had no effect upon cell proliferation (last column of FIG. 7). The data from FIG. 7 are indicative that troglitazone may be acting or acting additionally more through its vitamin E antioxidant properties as opposed to its ability to bind to PPAR-γ. Further comparisons with the thiazolidinediones in combinatorial therapy reveal that troglitazone is superior to pioglitazone and BRL49653 in its ability to inhibit cell proliferation. The results of combinatorial therapy with 5-FU are depicted in Figure 8 and those of doxorubicin are depicted in FIG. 9. As also seen in FIG. 9, none of the thiazolidinediones had any additional inhibitory effect on cell proliferation when administered with methotrexate.
Taken together, this data demonstrate that functional PPAR-γ exist in human osteosarcoma, Saos-2, cells. This is also the first demonstration of PPAR-γ mRNA expression is normal osteoblasts. However, thiozolidinediones with a higher affinity for PPAR-γ were less effective than troglitazone in inhibiting cell proliferation suggesting that troglitazone may work as an antioxidant. However, higher doses of vitamin E were needed to cause similar growth inhibitory repsonses when compared to troglitazone. Thus, it is plausible that troglitazone utilizes both functions in inhibiting cell proliferation. Troglitazone because of its unique properties of causing terminal differentiation and low toxicity may be an excellent candidate for prophylactic therapy for individuals at high risk for cancer.
References
Chinery, Brockman, Peeler, Shyr, Beauchamp, Coffey, Nature Med., 3:1233-1241,
1997. Harper and Elledge, Curr. Opin. Genet. Dev., 6:54-56, 1996. Kliewer, Umesona, Noonan, Heyman, Evans, Nature, 358:771-774, 1992.
Sears, MacGinnitie, Kovacs, Graves, Mol Cell Biol, 16:3410-3418, 1996. Sherr, Science, 274:1672-1677, 1996.
Tontonoz, Graves, Budavari, Erdjument-Bromage, Lui, Hu, Tempst, Spiegelman, Nucleic Acids Res., 22:5628-5634, 1994B. Tontonoz, Graves, Hu, Budavari, Spiegelman, Genes Dev., 8:1224-1234, 1994 A.
Tontonoz, Hu, Devine, Beale, Spiegelman, Mol Cell Biol, 15:351-357, 1995. Tontonoz, Singer, Forman, Sarraf et al, Proc. Natl. Acad. Sci. USA, 94:237-241,
1997. Warrel, de The, Wang, Degos, N. Engl J. Med, 329:177-189, 1993.

Claims

Claims:
1. Use of troglitazone alone as a chemotherapeutic agent for the treatment of bone cancer.
2. Use of troglitazone alone as a chemotherapeutic agent for the treatment of tumor expressing PPAR-γ.
3. Use of troglitazone for the treatment of tumors derived from precursor cells to osteosarcomas.
4. Use troglitazone in combination with at least one other chemotherapeutic agent for the treatment of bone cancer, other cancer types expressing PPAR-γ, or cancer derived from the precursor cell type to osteosarcoma.
5. The use of claim 1 or 4 where the bone cancer is osteosarcoma.
6. The use of claim 4 where the other chemotherapeutic agent is 5-fluorouracil or doxorubicin.
PCT/US1998/020611 1998-09-29 1998-09-29 Thiazolidenediones alone or in combination with other therapeutic agents for tumor therapy WO2000018234A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/US1998/020611 WO2000018234A1 (en) 1998-09-29 1998-09-29 Thiazolidenediones alone or in combination with other therapeutic agents for tumor therapy

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US1998/020611 WO2000018234A1 (en) 1998-09-29 1998-09-29 Thiazolidenediones alone or in combination with other therapeutic agents for tumor therapy

Publications (1)

Publication Number Publication Date
WO2000018234A1 true WO2000018234A1 (en) 2000-04-06

Family

ID=22267993

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/020611 WO2000018234A1 (en) 1998-09-29 1998-09-29 Thiazolidenediones alone or in combination with other therapeutic agents for tumor therapy

Country Status (1)

Country Link
WO (1) WO2000018234A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004045611A1 (en) * 2002-11-18 2004-06-03 N-Gene Research Laboratories Inc. Use of a thiazolidinedione for the reduction of side effects of chemotherapy
US7045523B2 (en) 2001-10-18 2006-05-16 Novartis Ag Combination comprising N-{5-[4-(4-methyl-piperazino-methyl)-benzoylamido]-2-methylphenyl}-4-(3-pyridyl)-2-pyrimidine-amine and telomerase inhibitor
WO2007144679A2 (en) * 2006-06-14 2007-12-21 University Of Debrecen Compounds, kits and methods for conferring cytoprotection
US7323481B2 (en) 2001-04-06 2008-01-29 Hoffmann-La Roche Inc. Thiazolidinediones alone or in combination with other therapeutic agents for inhibiting or reducing tumour growth

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH0342088Y2 (en) * 1985-05-29 1991-09-04

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH0342088Y2 (en) * 1985-05-29 1991-09-04

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7323481B2 (en) 2001-04-06 2008-01-29 Hoffmann-La Roche Inc. Thiazolidinediones alone or in combination with other therapeutic agents for inhibiting or reducing tumour growth
US7045523B2 (en) 2001-10-18 2006-05-16 Novartis Ag Combination comprising N-{5-[4-(4-methyl-piperazino-methyl)-benzoylamido]-2-methylphenyl}-4-(3-pyridyl)-2-pyrimidine-amine and telomerase inhibitor
WO2004045611A1 (en) * 2002-11-18 2004-06-03 N-Gene Research Laboratories Inc. Use of a thiazolidinedione for the reduction of side effects of chemotherapy
WO2007144679A2 (en) * 2006-06-14 2007-12-21 University Of Debrecen Compounds, kits and methods for conferring cytoprotection
WO2007144679A3 (en) * 2006-06-14 2009-06-18 Univ Debrecen Compounds, kits and methods for conferring cytoprotection

Similar Documents

Publication Publication Date Title
Perkins et al. Good cop, bad cop: the different faces of NF-κB
Owa et al. Cell cycle regulation in the G1 phase: a promising target for the development of new chemotherapeutic anticancer agents
Rapisarda et al. Identification of small molecule inhibitors of hypoxia-inducible factor 1 transcriptional activation pathway
TW474810B (en) Pharmaceutical pyrazole compositions useful as inhibitors of protein kinases
Göke et al. Regulation of TRAIL-induced apoptosis by transcription factors
Jung et al. Hypoxia-inducible factor induction by tumour necrosis factor in normoxic cells requires receptor-interacting protein-dependent nuclear factor kappaB activation
Chen et al. Induction of G1 phase arrest in MCF human breast cancer cells by pentagalloylglucose through the down-regulation of CDK4 and CDK2 activities and up-regulation of the CDK inhibitors p27Kip and p21Cip
HRP20010521A2 (en) The use of 4-h-1-benzopyran-4-one derivatives as inhibitors of smooth muscle cell proliferation
Crews et al. Small-molecule inhibitors of the cell cycle
Chang et al. Extracellular signal-regulated kinase activation and Bcl-2 downregulation mediate apoptosis after gemcitabine treatment partly via a p53-independent pathway
Atmaca et al. Novel ferrocenyl pyrazoles inhibit breast cancer cell viability via induction of apoptosis and inhibition of PI3K/Akt and ERK1/2 signaling
Rifkind et al. Induced differentiation, the cell cycle, and the treatment of cancer
Hsu et al. YC-1 inhibits proliferation of human vascular endothelial cells through a cyclic GMP-independent pathway
Wang et al. YC-1 [3-(5′-Hydroxymethyl-2′-furyl)-1-benzyl Indazole] exhibits a novel antiproliferative effect and arrests the cell cycle in G0-G1 in human hepatocellular carcinoma cells
Lafanechère The microtubule cytoskeleton: An old validated target for novel therapeutic drugs
Qiu et al. Anticancer quinones induce pRb-preventable G2/M cell cycle arrest and apoptosis
Fedier et al. Loss of atm sensitises p53-deficient cells to topoisomerase poisons and antimetabolites
Li et al. Synthesis and evaluation of the HIF-1α inhibitory activity of 3 (5)-substituted-4-(quinolin-4-yl)-and 4-(2-phenylpyridin-4-yl) pyrazoles as inhibitors of ALK5
Nazreen Design, synthesis, and molecular docking studies of thiazolidinediones as PPAR‐γ agonists and thymidylate synthase inhibitors
US20040253730A1 (en) Activated checkpoint therapy and methods of use thereof
Kim et al. NF-κB activation is required for cisplatin-induced apoptosis in head and neck squamous carcinoma cells
Lv et al. Small-molecule inhibitor targeting protein kinase D: a potential therapeutic strategy
US20060025429A1 (en) Composition and methods for inhibiting expression of hypoxia-inducible genes
EP2023925A2 (en) Cdki pathway inhibitors as selective inhibitors of tumor cell growth
WO2000018234A1 (en) Thiazolidenediones alone or in combination with other therapeutic agents for tumor therapy

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): CA JP

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)