US20140288078A1 - Use of phosphodiesterase inhibitors for treating multidrug resistance - Google Patents

Use of phosphodiesterase inhibitors for treating multidrug resistance Download PDF

Info

Publication number
US20140288078A1
US20140288078A1 US14/215,982 US201414215982A US2014288078A1 US 20140288078 A1 US20140288078 A1 US 20140288078A1 US 201414215982 A US201414215982 A US 201414215982A US 2014288078 A1 US2014288078 A1 US 2014288078A1
Authority
US
United States
Prior art keywords
cells
abcb1
sildenafil
vardenafil
abcg2
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
US14/215,982
Inventor
Zhe-Sheng CHEN
Zhi SHI
Charles R. ASHBY
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.)
St Johns University USA
Original Assignee
St Johns University USA
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 St Johns University USA filed Critical St Johns University USA
Priority to US14/215,982 priority Critical patent/US20140288078A1/en
Publication of US20140288078A1 publication Critical patent/US20140288078A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • 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
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/275Nitriles; Isonitriles
    • A61K31/277Nitriles; Isonitriles having a ring, e.g. verapamil
    • 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
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • 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
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to a method of treating multidrug resistance with phosphodiesterase (PDE) inhibitors, e.g., PDE5 inhibitors. More specifically, the present invention relates to a method of treating multidrug resistance, e.g., multidrug resistance that arises, e.g., during administration of chemotherapeutic or antineoplastic agents for treatment of cancer, with a PDE5 inhibitor (e.g., vardenafil, sildenafil, and tadalafil).
  • PDE phosphodiesterase
  • the invention relates to a method of treating cancer, e.g., multidrug resistant cancer, using a PDE5 inhibitor in combination with an anticancer agent, i.e., a chemotherapeutic/antineoplastic agent.
  • a pharmaceutical composition for treating a multidrug resistant cancer comprising a PDE5 inhibitor, or a combination of a PDE5 inhibitor and an antineoplastic agent.
  • Multidrug resistance is a phenomenon in which disease-causing organisms or cells are able to evade treatment with drugs designed to target them.
  • Multidrug resistance to cancer chemotherapy a major hurdle to successfully treating cancer, commonly arises due to the ability of one or more cancer cells to resist treatment with a variety of agents, e.g., drugs that may be distinct in one or both of the following: structure and mechanism of action. Therefore, whereas a portion of cancer cells, i.e., drug-sensitive cancer cells, are killed with a chemotherapeutic agent, drug-resistant cancer cells survive, such that the resultant or remaining cancers consist primarily of drug-resistant (or medicine-tolerant) cells.
  • ABC ATP-binding cassette
  • Pgp/ABCB1/MDR1 P-glycoprotein
  • ABCG2/BCRP/ABCP breast cancer resistant protein
  • ABCC1/MRP1 multidrug resistant protein 1
  • ABCB1 transporter occurs in 40-50% of all cancers; therefore, there is a major ongoing effort to design a strategy for inhibiting this transporter.
  • Three generations of compounds that inhibit ABCB1 have been studied.
  • the first generation of ABCB1 inhibitors included such drugs as verapamil, quinine, and cyclosporine A; however, these first generation agents produced undesirable adverse effects at concentrations necessary to inhibit ABCB1 (Krishna et al. (2000) Eur. J. Pharm. Sci. 11:265-83).
  • the second generation of ABCB1 inhibitors e.g., valspodar and biricodar, produced unpredictable interactions with other transport proteins and inhibited cytochrome P450 3A4 (CYP3A4), an enzyme responsible for metabolizing many xenobiotics, including chemotherapeutic drugs. Therefore, the use of the second generation ABCB1 inhibitors resulted in decreased clearance and increased toxicity of many chemotherapeutic agents (Gottesman et al., supra).
  • the third generation of inhibitors e.g., LY335979 (zosuquidar), GF120918 (elacridar), and MS-209 (dofequidar), were derived from second generation agents, and had nanomolar affinity for the ABCB1 transporter.
  • Multidrug resistant protein 7 (MRP7; ABCC10), a member of MRP subfamily, is similar in topology to other MRPs (including MRP1) with two nucleotide-binding domains and three transmembrane domains (Kruh et al. (2007) Pflugers Arch. 453:675-84; Chen et al. (2003) Mol. Pharmacol. 63:351-58).
  • ABCC10 is able to confer resistance to several natural product chemotherapeutic drugs, including taxanes and vinca alkaloids, which are also substrates of Pgp (Hopper-Borge et al. (2004) Cancer Res. 64:4927-30).
  • ABCC10 has been reported to confer resistance to vinorelbine and paclitaxel in non-small cell lung cancer cells (Bessho et al. (2009) Oncol. Rep. 21:263-68; Oguri et al. (2008) Mol. Cancer Ther. 7:1150-55) and to vincristine in human salivary gland adenocarcinoma cells (Naramoto et al. (2007) Int. J. Oncol. 30:393-401). The in vivo functions of ABCC10 have recently been confirmed using an Mrp7 knockout mouse model (Hopper-Borge et al. (2011) Cancer Res. 71(10):3649-57).
  • Phosphodiesterase type 5 (PDE5) inhibitors are widely used in the treatment of male erectile dysfunction and in improving breathing efficiency in pulmonary hypertension. As agents already used in the clinic for other purposes, these drugs were investigated for their effects on MDR and ABC transporters.
  • the present invention relates to methods of treating multidrug resistance in a cancer cell in a subject in need thereof by inhibiting ABCB1 transporter activity, comprising administering to the subject a therapeutically effective amount of a phosphodiesterase 5 (PDE5) inhibitor.
  • PDE5 inhibitor is administered in combination with an antineoplastic agent.
  • the present invention further relates to methods of treating a cancer in a subject by inhibiting ABCB1 transporter activity, comprising administering to the subject a therapeutically effective amount of an antineoplastic agent and a therapeutically effective amount of a PDE5 inhibitor.
  • the cancer is a multidrug resistant cancer.
  • the subject is a mammal, e.g., a human.
  • the present invention relates to (1) methods of increasing sensitivity of a multidrug resistant cancer cell to an antineoplastic agent by inhibiting ABCB1 transporter activity, comprising contacting the cancer cell with a PDE5 inhibitor; (2) methods of inhibiting growth of a multidrug resistant cancer cell by inhibiting ABCB1 transporter activity, comprising contacting the cell with a combination of an antineoplastic agent and a PDE5 inhibitor; (3) methods of increasing accumulation of an antineoplastic agent in a cancer cell by inhibiting ABCB1 transporter activity, comprising contacting the cell with a PDE5 inhibitor; and (4) methods of decreasing efflux of an antineoplastic agent from a cancer cell by inhibiting ABCB1 transporter activity, comprising contacting the cell with a PDE5 inhibitor.
  • these methods are performed in vivo.
  • the methods are performed in a mammalian subject, e.g., a human subject.
  • both the antineoplastic agent and the PDE5 inhibitor are administered in therapeutically effective amounts.
  • the PDE5 inhibitor is selected from the group consisting of sildenafil, vardenafil, tadalafil, lodenafil, udenafil, benzamidenafil, mirodenafil, avanafil, zaprinast, SLX-2101, UK-371,800, UK-122764, icariin, DA-8159, 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]-pyrazin-2(1H)-one, and their analogs.
  • the antineoplastic agent is selected from the group consisting of antineoplastic agents listed in Table 1; as a nonlimiting example, the antineoplastic agent can be selected from the group consisting of paclitaxel, vinblastine, and vincristine.
  • the present invention also relates to methods of treating multidrug resistance in a cancer cell in a subject in need thereof by inhibiting ABCG2 transporter activity, comprising administering to the subject a therapeutically effective amount of a PDE5 inhibitor.
  • the PDE5 inhibitor is administered in combination with an antineoplastic agent.
  • the present invention further relates to methods of treating a cancer in a subject by inhibiting ABCG2 transporter activity, comprising administering to the subject a therapeutically effective amount of an antineoplastic agent and a therapeutically effective amount of a PDE5 inhibitor.
  • the cancer is a multidrug resistant cancer.
  • the subject is a mammal, e.g., a human.
  • the present invention relates to (1) methods of increasing sensitivity of a multidrug resistant cancer cell to an antineoplastic agent by inhibiting ABCG2 transporter activity, comprising contacting the cancer cell with a PDE5 inhibitor; (2) methods of inhibiting growth of a multidrug resistant cancer cell by inhibiting ABCG2 transporter activity, comprising contacting the cell with a combination of an antineoplastic agent and a PDE5 inhibitor; (3) methods of increasing accumulation of an antineoplastic agent in a cancer cell by inhibiting ABCG2 transporter activity, comprising contacting the cell with a PDE5 inhibitor; and (4) methods of decreasing efflux of an antineoplastic agent from a cancer cell by inhibiting ABCG2 transporter activity, comprising contacting the cell with a PDE5 inhibitor.
  • these methods are performed in vivo.
  • the methods are performed in a mammalian subject, e.g., a human subject.
  • both the antineoplastic agent and the PDE5 inhibitor are administered in therapeutically effective amounts.
  • the PDE5 inhibitor is selected from the group consisting of sildenafil, vardenafil, tadalafil, lodenafil, udenafil, benzamidenafil, mirodenafil, avanafil, zaprinast, SLX-2101, UK-371,800, UK-122764, icariin, DA-8159, 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]-pyrazin-2(1H)-one, and their analogs.
  • the antineoplastic agent is selected from the group of antineoplastic agents listed in Table 2; as a nonlimiting example, the antineoplastic agent can be selected from the group consisting of SN-38, flavopiridol, mitoxantrone, and methotrexate.
  • the present invention also relates to methods of treating multidrug resistance in a cancer cell in a subject in need thereof by inhibiting ABCC10 transporter activity, comprising administering to the subject a therapeutically effective amount of a PDE5 inhibitor.
  • the PDE5 inhibitor is administered in combination with an antineoplastic agent.
  • the present invention further relates to methods of treating a cancer in a subject by inhibiting ABCC10 transporter activity, comprising administering to the subject a therapeutically effective amount of an antineoplastic agent and a therapeutically effective amount of a PDE5 inhibitor.
  • the cancer is a multidrug resistant cancer.
  • the subject is a mammal, e.g., a human.
  • the present invention relates to (1) methods of increasing sensitivity of a multidrug resistant cancer cell to an antineoplastic agent by inhibiting ABCC10 transporter activity, comprising contacting the cancer cell with a PDE5 inhibitor; (2) methods of inhibiting growth of a multidrug resistant cancer cell by inhibiting ABCC10 transporter activity, comprising contacting the cell with a combination of an antineoplastic agent and a PDE5 inhibitor; (3) methods of increasing accumulation of an antineoplastic agent in a cancer cell by inhibiting ABCC10 transporter activity, comprising contacting the cell with a PDE5 inhibitor; and (4) methods of decreasing efflux of an antineoplastic agent from a cancer cell by inhibiting ABCC10 transporter activity, comprising contacting the cell with a PDE5 inhibitor.
  • these methods are performed in vivo.
  • the methods are performed in a mammalian subject, e.g., a human subject.
  • both the antineoplastic agent and the PDE5 inhibitor are administered in therapeutically effective amounts.
  • the PDE5 inhibitor is selected from the group consisting of sildenafil, vardenafil, tadalafil, lodenafil, udenafil, benzamidenafil, mirodenafil, avanafil, zaprinast, SLX-2101, UK-371,800, UK-122764, icariin, DA-8159, 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]-pyrazin-2(1H)-one, and their analogs.
  • the antineoplastic agent is selected from the group of antineoplastic agents listed in Table 3; as a nonlimiting example, the antineoplastic agent can be selected from the group consisting of paclitaxel, docetaxel, vinblastine, and vincristine.
  • the invention also provides methods of stimulating the ATPase activity of an ABCB1 transporter in a cell comprising contacting the cell with a PDE5 inhibitor.
  • the PDE5 inhibitor is selected from the group consisting of sildenafil, vardenafil, tadalafil, lodenafil, udenafil, benzamidenafil, mirodenafil, avanafil, zaprinast, SLX-2101, UK-371,800, UK-122764, icariin, DA-8159, 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl) pyrido[3,4-b]-pyrazin-2(1H)-one, and their analogs.
  • the invention also provides methods of stimulating the ATPase activity of an ABCG2 transporter in a cell comprising contacting the cell with a PDE5 inhibitor.
  • the PDE5 inhibitor is selected from the group consisting of sildenafil, vardenafil, tadalafil, lodenafil, udenafil, benzamidenafil, mirodenafil, avanafil, zaprinast, SLX-2101, UK-371,800, UK-122764, icariin, DA-8159, 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl) pyrido[3,4-b]-pyrazin-2(1H)-one, and their analogs.
  • the invention also contemplates methods of stimulating the ATPase activity of an ABCC10 transporter in a cell comprising contacting the cell with a PDE5 inhibitor.
  • the PDE5 inhibitor is selected from the group consisting of sildenafil, vardenafil, tadalafil, lodenafil, udenafil, benzamidenafil, mirodenafil, avanafil, zaprinast, SLX-2101, UK-371,800, UK-122764, icariin, DA-8159, 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl) pyrido[3,4-b]-pyrazin-2(1H)-one, and their analogs.
  • the present invention provides pharmaceutical compositions for treating a multidrug resistant cancer by inhibiting ABCB1 transporter activity in a subject comprising a PDE5 inhibitor and a pharmaceutical excipient.
  • the PDE5 inhibitor is selected from the group consisting of sildenafil, vardenafil, tadalafil, lodenafil, udenafil, benzamidenafil, mirodenafil, avanafil, zaprinast, SLX-2101, UK-371,800, UK-122764, icariin, DA-8159, 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]-pyrazin-2(1H)-one, and their analogs.
  • the pharmaceutical composition of the invention further comprises an antineoplastic agent.
  • the antineoplastic agent is selected from the antineoplastic agent.
  • the present invention provides pharmaceutical compositions for treating a multidrug resistant cancer by inhibiting ABCG2 transporter activity in a subject comprising a PDE5 inhibitor and a pharmaceutical excipient.
  • the PDE5 inhibitor is selected from the group consisting of sildenafil, vardenafil, tadalafil, lodenafil, udenafil, benzamidenafil, mirodenafil, avanafil, zaprinast, SLX-2101, UK-371,800, UK-122764, icariin, DA-8159, 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl) pyrido[3,4-b]-pyrazin-2(1H)-one, and their analogs.
  • the pharmaceutical composition further comprises an antineoplastic agent.
  • the antineoplastic agent is selected from
  • the present invention provides pharmaceutical compositions for treating a multidrug resistant cancer by inhibiting ABCC10 transporter activity in a subject comprising a PDE5 inhibitor and a pharmaceutical excipient.
  • the PDE5 inhibitor is selected from the group consisting of sildenafil, vardenafil, tadalafil, lodenafil, udenafil, benzamidenafil, mirodenafil, avanafil, zaprinast, SLX-2101, UK-371,800, UK-122764, icariin, DA-8159, 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]-pyrazin-2(1H)-one, and their analogs.
  • the pharmaceutical composition of the invention further comprises an antineoplastic agent.
  • the antineoplastic agent is selected from the group consisting of antineoplastic agents listed in Table 3.
  • the subject is a mammal, e.g., a human subject.
  • FIG. 1 depicts the effects of vardenafil and tadalafil on the accumulation ( FIG. 1A ) and efflux ( FIG. 1B ) of [ 3 H]-paclitaxel, as well as ABCB1 expression ( FIG. 1C ) in ABCB1-overexpressing cells.
  • FIG. 1A The accumulation of [ 3 H]-paclitaxel was measured after cells were preincubated with or without vardenafil, tadalafil, or verapamil for 1 h at 37° C., and then incubated with 0.1 ⁇ M [ 3 H]-paclitaxel for another 2 h at 37° C.
  • FIG. 1B The percentage of paclitaxel released was plotted as a function of time.
  • KB-3-1 or KB-C2 cells received either no reversal agent treatment or were treated with vardenafil and verapamil throughout the experiment. Cells were pretreated for 1 h, coincubated with [ 3 H]-paclitaxel, washed, and reincubated in paclitaxel-free medium. Cells were collected and levels of [ 3 H]-paclitaxel determined either at 0, 60, or 120 min.
  • FIG. 1C Effect of vardenafil (left panel) or tadalafil (right panel) on the expression of ABCB1 for 36 and 72 h, respectively. Independent experiments were performed at least three times, and a representative experiment is shown.
  • FIG. 2 depicts the effects of vardenafil and tadalafil on the Vi-sensitive ATPase activity of ABCB1, represented as a function of ATP hydrolysis ( FIG. 2A ), and the photoaffinity labeling of ABCB1 with [ 125 I]-IAAP (iodoarylazidoprazosin), represented as IAAP incorporation ( FIG. 2B ).
  • Mean values are given, and the error bars represent standard error from at least three independent experiments.
  • FIG. 3A is ribbon diagram of open-to-the-cytoplasm 3D-structural conformation of a homology model of human ABCB1 based on the crystal structure coordinates of mouse ABCB1.
  • the docked poses of tadalafil (shown on bottom), and vardenafil and IAAP (shown on top; overlapping) as ball and stick models are shown within the large hydrophobic cavity of ABCB1 at different inhibitor binding sites (see also Ding et al. (2011) PLoS ONE 6(4):e19329).
  • FIG. 3B is the XP-Glide-predicted binding model of vardenafil (left panel) and tadalafil (right panel).
  • Important amino acids are depicted as stick models, whereas the two inhibitors are shown as ball and stick models.
  • the short dotted lines indicate hydrogen bonding interactions, whereas the long dotted lines indicate interacting distances.
  • FIG. 4 demonstrates that sildenafil increased the accumulation of [ 3 H]-paclitaxel or [ 3 H]-mitoxantrone in ABCB1- or ABCG2-overexpressing cells.
  • the accumulation of [ 3 H]-paclitaxel ( FIG. 4A ) or [ 3 H]-mitoxantrone ( FIGS. 4B , 4 C, and 4 D) was measured after cells were preincubated with or without sildenafil, verapamil, or FTC for 1 h at 37° C. and then incubated with 0.1 ⁇ M [ 3 H]-paclitaxel or 0.02 ⁇ M [ 3 H]-mitoxantrone for another 2 h at 37° C.
  • Data points represent the means ⁇ SD of triplicate determinations. * and ** represent p ⁇ 0.05 and p ⁇ 0.01, respectively, for values versus those in the control group. Independent experiments were carried out at least three times, and a representative experiment is shown.
  • FIG. 5 demonstrates that sildenafil inhibited the efflux of BODIPY-prazosin ( FIGS. 5A-5D ) and transport of [ 3 H]-E 2 17 ⁇ G as well as [ 3 H]-methotrexate ( FIGS. 5E and 5F ) by ABCG2.
  • FIGS. 5A-5D sildenafil inhibited the efflux of BODIPY-prazosin
  • FIGS. 5E and 5F transport of [ 3 H]-E 2 17 ⁇ G as well as [ 3 H]-methotrexate
  • flow cytometric data (measured as cell numbers) from HEK293/pcDNA3.1, ABCG2-482-G2, ABCG2-482-R5, and ABCG2-482-T7 cells are shown after incubation with 250 nM BODIPY-prazosin alone (solid line) or with 50 ⁇ M sildenafil (dashed line), and 2.5 ⁇ M FTC (shaded histogram).
  • FIGS. 1-10 flow cytometric data (measured as cell numbers) from HEK293/pcDNA3.1, ABCG2-482-G2, ABCG2-482-R5, and ABCG2-482-T7 cells are shown after incubation with 250 nM BODIPY-prazosin alone (solid line) or with 50 ⁇ M sildenafil (dashed line), and 2.5 ⁇ M FTC (shaded histogram).
  • membrane vesicles were prepared from HEK293/pcDNA3.1 and ABCG2-482-R5 cells, and the rates of the uptake of [ 3 H]-E 2 17 ⁇ G and [ 3 H]-methotrexate into membrane vesicles (10 ⁇ g protein/reaction) were measured. Data points represent the means ⁇ SD of triplicate determinations. * and ** represent p ⁇ 0.05 and p ⁇ 0.01, respectively, for values versus those in the control group. At least three independent experiments were carried out, and a representative experiment is shown.
  • FIG. 6 depicts the effect of sildenafil on the ATPase activity and [ 125 I]-IAAP photoaffinity labeling of ABCB1 and ABCG2.
  • the Vi-sensitive ATPase activities (measured as a function of ATP hydrolysis) of ABCB1 ( FIG. 6A ) and ABCG2 ( FIG. 6B ) in membrane vesicles were determined with different concentrations of sildenafil. Mean values are given, and the error bars represent standard error from at least three independent experiments.
  • the photoaffinity labeling (measured as a function of IAAP incorporation) of ABCB1 ( FIG. 6C ) and ABCG2 ( FIG.
  • FIG. 7 is a model for binding of sildenafil to ABCB1.
  • FIG. 7A is a ribbon diagram of open-to-the-cytoplasm 3D-structural conformation of a homology model of human ABCB1 based on the crystal structure coordinates of mouse Mdr3. Sildenafil is shown as a ball and stick model within the large hydrophobic cavity of ABCB1 characterized by the QZ59-RRR inhibitor binding site (see also Shi et al. (2011) Cancer Research 71(8):1-13).
  • FIG. 7B is an XP-Glide-predicted binding model of sildenafil within the QZ59-RRR binding site. Important amino acids are depicted as stick models, whereas the inhibitor is shown as a ball and stick model. The short dotted line indicates a hydrogen bonding interaction, whereas the long dotted line indicates an interacting distance.
  • FIG. 8 represents survival curves at different concentrations of colchicine ( FIG. 8A ), vinblastine ( FIG. 8B ), paclitaxel ( FIG. 8C ), and cisplatin ( FIG. 8D ) for either KB-3-1 cells with (open squares) or without (filled squares) 10 ⁇ M sildenafil; KB-C2 cells with (open diamonds) or without (filled diamonds) 10 ⁇ M sildenafil; or KB-V1 cells with (open triangles) or without (filled triangles) 10 ⁇ M sildenafil.
  • Cell survival was determined by MTT assay; data points are the means ⁇ SD of triplicate determinations, with a representative experiment shown.
  • FIG. 9 represents survival curves at different concentrations of flavopiridol ( FIG. 9A ), mitoxantrone ( FIG. 9B ), SN-38 ( FIG. 9C ), and cisplatin ( FIG. 9D ) for either S1 cells with (open squares) or without (filled squares) 50 ⁇ M sildenafil; S1/FLV5000 cells with (open diamonds) or without (filled diamonds) 50 ⁇ M sildenafil; or S1-M1-80 cells with (open triangles) or without (filled triangles) 50 ⁇ M sildenafil.
  • Cell survival was determined by MTT assay; data points are the means ⁇ SD of triplicate determinations, with a representative experiment shown.
  • FIG. 10 represents survival curves at different concentrations of flavopiridol ( FIG. 10A ), mitoxantrone ( FIG. 10B ), SN-38 ( FIG. 10C ), and cisplatin ( FIG. 10D ) for either MCF-7 cells with (open squares) or without (filled squares) 50 ⁇ M sildenafil; MCF-7/FLV10000 cells with (open diamonds) or without (filled diamonds) 50 ⁇ M sildenafil; or MCF-7/ADVP3000 cells with (open triangles) or without (filled triangles) 50 ⁇ M sildenafil.
  • Cell survival was determined by MTT assay; data points are the means ⁇ SD of triplicate determinations, with a representative experiment shown.
  • FIG. 11 represents survival curves at different concentrations of flavopiridol ( FIG. 11A ), mitoxantrone ( FIG. 11B ), SN-38 ( FIG. 11C ), and cisplatin ( FIG. 11D ) for either HEK293/pcDNA3 cells with (open squares) or without (filled squares) 50 ⁇ M sildenafil; HEK293/ABCG2-G2 cells with (open diamonds) or without (filled diamonds) 50 ⁇ M sildenafil; HEK293/ABCG2-R5 cells with (open triangles) or without (filled triangles) 50 ⁇ M sildenafil, or HEK293/ABCG2-T7 cells with (open circles) or without (filled circles) 50 ⁇ M sildenafil.
  • Cell survival was determined by MTT assay; data points are the means ⁇ SD of triplicate determinations, with a representative experiment shown.
  • FIG. 12 represents the effects of sildenafil treatment on the growth of tumor xenografts. Mice were treated with either vehicle, sildenafil alone, paclitaxel alone, or a combination of sildenafil and paclitaxel.
  • FIG. 12A demonstrates the sizes of the excised tumors
  • FIG. 12B demonstrates the tumor weight of the excised tumors. * represents p ⁇ 0.05 for values versus those indicated.
  • FIG. 13 demonstrates the effects of sildenafil treatment on tumor volume in mouse xenografts, measured on days 0, 1, 3, 6, 9, 12, 15, and 18 after transplantation. Mice were treated with either vehicle, sildenafil alone, paclitaxel alone, or a combination of sildenafil and paclitaxel. * represents p ⁇ 0.05 for values versus those in the vehicle group; *, # represents p ⁇ 0.05 for values versus those in the paclitaxel alone group.
  • FIG. 14 demonstrates the effects of PDE5 inhibitors on the reversal of ABCC10-mediated drug resistance in HEK/MRP7 cells, showing the survival curves of HEK/MRP7 cells in the presence or absence of sildenafil, vardenafil, or tadalafil at 5 ⁇ M and the parental HEK293-pcDNA3.1 cells at different concentrations of paclitaxel ( FIG. 14A ), docetaxel ( FIG. 14B ), vinblastine ( FIG. 14C ), and cisplatin ( FIG. 14D ).
  • Cell survival was determined by MTT assay as described herein. Data points represent the means ⁇ SD of triplicate determinations. Experiments were performed at least three independent times.
  • FIG. 15 shows the effects of sildenafil, vardenafil, and tadalafil on the accumulation of [ 3 H]-paclitaxel in HEK293-pcDNA3.1 and HEK/MRP7 cells.
  • the intracellular accumulation of [ 3 H]-paclitaxel was measured by scintillation counting after cells were preincubated with or without sildenafil, vardenafil, tadalafil, or cepharanthine for 2 h at 37° C. and then incubated with 0.1 ⁇ M [ 3 H]-paclitaxel for another 2 h at 37° C.
  • Data points represent the means ⁇ SD of triplicate determinations. Experiments were performed at least three independent times. ** represents p ⁇ 0.01 for values versus those in the control group.
  • FIG. 16 demonstrates the effects of sildenafil ( FIG. 16A ), vardenafil ( FIG. 16B ), and tadalafil ( FIG. 16C ) on the efflux of [ 3 H]-paclitaxel in HEK293-pcDNA3.1 and HEK/MRP7 cells.
  • Cells were preincubated with or without sildenafil, vardenafil, or tadalafil for 2 h at 37° C., and further incubated with 0.1 ⁇ M [ 3 H]-paclitaxel for another 2 h at 37° C. Cells were then incubated in fresh medium with or without PDE5 inhibitors for different time periods at 37° C.
  • FIG. 17 shows immunoblot detection ( FIG. 17A ) and immunofluorescence detection ( FIG. 17B ) of MRP7 in HEK/MRP7 cells following incubation with PDE5 inhibitors.
  • Cell lysates were prepared from HEK/MRP7 cells incubated with 5 ⁇ M sildenafil, vardenafil, and tadalafil for different time periods (0, 24, 48, and 72 h).
  • Immunoblot detection of MRP7 FIG. 17A
  • Equal amounts 40 ⁇ g of protein
  • the present invention is based on the finding that phosphodiesterase 5 (PDE5) inhibitors can be useful for treating multidrug resistance in cells, e.g., cancer cells.
  • PDE5 phosphodiesterase 5
  • the present invention provides a method of treating multidrug resistance in cancer cells in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a PDE5 inhibitor.
  • multidrug resistance refers to a phenomenon wherein disease-causing organisms or cells are able to evade treatment with agents designed to target them, often agents that are structurally or mechanistically distinct.
  • multidrug resistance can be viewed as simultaneous resistance to different drugs that have either the same or different targets, and either similar or distinct chemical structures.
  • Organisms or cells may acquire a multidrug resistant phenotype upon exposure to a single drug, e.g., a chemotherapeutic/antineoplastic drug (also referred to herein as an “anticancer drug”).
  • Multidrug resistance in cancer, or multidrug resistant cancer refers to the situation wherein at least one cancer cell becomes resistant, or nonresponsive, to treatment with drugs, e.g., chemotherapeutic/antineoplastic drugs (agents, compounds, etc.), designed to target it, e.g., chemotherapeutic/antineoplastic drugs that may be structurally and/or mechanistically distinct. Therefore, cancer cells sensitive to the treatment with chemotherapeutic or antineoplastic drugs are killed, and the resistant cancer cell divides and propagates into a drug-resistant cancer growth.
  • drugs e.g., chemotherapeutic/antineoplastic drugs (agents, compounds, etc.)
  • chemotherapeutic/antineoplastic drugs e.g., chemotherapeutic/antineoplastic drugs that may be structurally and/or mechanistically distinct. Therefore, cancer cells sensitive to the treatment with chemotherapeutic or antineoplastic drugs are killed, and the resistant cancer cell divides and propagates into a drug-
  • ABC transporters are conserved proteins that translocate substrates across cellular membranes.
  • A-G ATP-binding cassette transporters
  • MDR transporters refers to ABC transporters involved in multidrug resistance in cancer cells.
  • the three MDR transporters with previously known predominant function in multidrug resistance to cancer chemotherapeutics are ABCB1 (P-glycoprotein or Pgp), ABCC1 (multidrug resistance protein 1 or MRP1), and ABCG2 (breast cancer resistant protein or BCRP), with ABCB1 transporter overexpression occurring in nearly 50% of all cancers.
  • ABCC10 MRP7 has now been shown to function in multidrug resistance to cancer chemotherapeutics.
  • cancers or metastases that may be affected by multidrug resistance, and thus may be treated using the methods of the present invention, include but are not limited to the following: cancers of the central nervous system, skin, respiratory tract, oral cavity, eyes, bone, skin, connective tissue, gastrointestinal tract, cardiovascular system (heart, vasculature, etc.), ear, sinuses, salivary glands, urethra, lips, bile duct, gall bladder, etc.; hematological-associated cancers (e.g., lymphomas, leukemias, myeloproliferative cancers, etc.); breast, ovarian, cervical, vaginal, uterine, testicular, renal, penile, bladder, prostate, nasopharyngeal, endocrine (thyroid, parathyroid, pituitary, adrenal, pancreatic, pineal), splenic, etc. cancers; as well as viral-associated cancers such as Kaposi's sarcoma, etc.
  • a subject in need thereof refers to any subject that is determined to be in need of therapy, e.g., therapy for the treatment of multidrug resistance with PDE5 inhibitors, by a treating physician (e.g., an oncologist).
  • a subject in need thereof may be a subject for whom a treatment with one or more chemotherapeutic agents was determined to be unsuccessful.
  • the success of a treatment with any particular therapy is evaluated based on the ability of that therapy to eliminate or reduce tumor burden, i.e., to eradicate or diminish the number of cancer cells.
  • the most suitable way to measure the elimination or reduction in tumor burden may vary by the type of cancer involved.
  • a treating physician will know the most suitable way to measure elimination or reduction in tumor burden, e.g., through obtaining and analyzing a biopsy sample.
  • a subject in need thereof may be a subject suffering from cancer, e.g., multidrug resistant cancer. In one embodiment, the subject in need thereof may be suffering from lung cancer.
  • a subject in need thereof may be a human or nonhuman animal.
  • nonhuman animal includes all vertebrates, such as nonhuman primates, sheep, dogs, cows, chickens, amphibians, reptiles, etc.
  • a subject may be a mammalian subject.
  • the present invention relates to the treatment of multidrug resistance by administering a phosphodiesterase inhibitor, e.g., a PDE5 inhibitor.
  • Phosphodiesterases are protein enzymes that function to break down a phosphodiester bond.
  • Cyclic nucleotide phosphodiesterases function to break down the phosphodiester bond in cyclic nucleotides, e.g., converting cyclic AMP (3′5′-cAMP; also commonly abbreviated as cAMP) and cyclic GMP (3′5′-cGMP; also commonly abbreviated as cGMP) to 5′-AMP and 5′-GMP, respectively; thereby inactivating these important second messengers and halting signal transduction cascades.
  • Increased levels of cyclic GMP are associated with smooth muscle vasodilation, and thus increased blood flow.
  • PDE5 is a phosphodiesterase responsible for converting second messenger cyclic GMP into 5′-GMP, thereby terminating its biological actions and potentially leading to vasoconstriction and reduced blood flow. Inhibitors of PDE5 increase cyclic GMP levels, and have been used in the clinic to treat erectile dysfunction and pulmonary arterial hypertension.
  • PDE5 inhibitors include, but are not limited to, vardenafil (LEVITRA®); sildenafil (VIAGRA®); tadalafil (CIALIS®); lodenafil (HELLEVA®); udenafil (ZYDENA®); benzamidenafil (Zou et al. (2008) J. Pharm. Biomed. Anal. 47:255-59); SLX-2101; mirodenafil; avanafil; UK-371,800 (Bunnage et al. (2008) Bioorg. Med. Chem. Lett.
  • PDE5 inhibitors e.g., inhibitors such as sildenafil, tadalafil, and vardenafil
  • PDE5 inhibitors are useful in blocking multidrug resistance in cancer, and thereby increasing the efficacy of antineoplastic agents.
  • PDE5 inhibitors are capable of reversing multidrug resistance
  • PDE5 inhibitors as well as other agents known to reverse multidrug resistance, are sometimes referred to as “reversal agents.”
  • PDE5 inhibitors can increase the efficacy of antineoplastic drugs in several ways.
  • PDE5 inhibitors can block the efflux of antineoplastic agents through MDR transporters.
  • the ATPase activity of ABC transporters e.g., MDR transporters
  • their transport substrates e.g., either their natural transport substrates or antineoplastic drugs.
  • the ATPase activity of the MDR transporters is stimulated in the presence of PDE5 inhibitors, e.g., sildenafil, tadalafil, and vardenafil.
  • PDE5 inhibitors may act as transport substrates of MDR transporters (e.g., competitive inhibitors of MDR transporters), thereby reducing the ability of MDR transporters to efflux antineoplastic agents.
  • PDE5 inhibitors can decrease cancer cell survival, thus having a positive effect on the outcome of cancer treatment.
  • PDE5 inhibitors can also positively affect cancer treatment because PDE5 inhibitors block cGMP hydrolysis, and thus increase cGMP levels, which may lead to activation of protein kinase G (PKG). Activation of PKG in turn leads to growth suppression or apoptosis.
  • PKG protein kinase G
  • a nonspecific PDE5 inhibitor e.g., a PDE inhibitor that blocks the function of other cGMP phosphodiesterases, in addition to its ability to block PDE5
  • MDR transporters such as ABCB1, ABCC10, and ABCG2.
  • a PDE inhibitor that is able to block the function of other cyclic nucleotide phosphodiesterases e.g., cyclic GMP phosphodiesterases
  • exclusive of an ability to block PDE5 may be effective in blocking MDR transporters such as ABCB1, ABCC10, and ABCG2.
  • Examples of phosphodiesterases that are able to catalyze cyclic GMP to 5′-GMP conversion include, but are not limited to, PDE1, PDE1B, PDE1C, PDE2A, PDE6A, PDE6B, PDE6C, PDE9A, PDE10A, and PDE11A.
  • MDR transporters are expressed in the membranes of various cell types (both normal and cancer cells), including cell types present at sites of drug absorption, e.g., intestinal epithelial cells; therefore, in one embodiment of the invention, a PDE5 inhibitor may block multidrug resistance, at least in part, by blocking the ability of MDR transporters expressed on epithelial cells to efflux antineoplastic drugs, thereby aiding drug absorption. In another embodiment of the invention, a PDE5 inhibitor blocks multidrug resistance by blocking the ability of MDR transporters expressed on cancer cells to efflux antineoplastic drugs.
  • a certain PDE5 inhibitor to block antineoplastic drug efflux.
  • a certain PDE5 inhibitor to block antineoplastic drug efflux.
  • Antineoplastic agents used for treating cancers, or under investigation for use in treating cancers may include, but are not limited to: (1) nitrogen mustards, such as mechlorethamine, cyclophosphamide, ifosfamide, melphalan (L-sarcolysin, L-PAM), uramustine, and chlorambucil; (2) ethylenimines and methylmelamines (aziridines), such as thioTEPA, hexamethylmelamine (HMM, altretamine), and triethylenemelamine (TEM); (3) nitrosoureas, such as carmustine (BCNU), lomustine (CCNU), semustine (Methyl-CCNU), fotemustine, and streptozotocin; (4) alkyl sulfonates, such as busulfan; (5) azines
  • antineoplastic agents of choice for various cancer diagnoses are summarized in, e.g., Basic and Clinical Pharmacology, pp. 945-46 (Bertram Katzung ed., Lange Medical Books/McGraw-Hill 8 th ed. 2001); and Goodman and Gilman's The Pharmacological Basis of Therapeutics, pp. 1383-85 (Joel G. Hardman, Lee E. Limbird, and Alfred Goodman Gilman eds., McGraw-Hill Medical Publishing Division 10 th Ed. 2001). Some sources use different groupings/classes to categorize the various antineoplastic or chemotherapeutic agents (drugs, compounds, etc.), but all appropriate antineoplastic agents are contemplated in the present invention.
  • antineoplastic agents are known to be substrates for different MDR transporters, e.g., ABCB1, ABCC1, ABCC10, and ABCG2 transporters.
  • MDR transporters e.g., ABCB1, ABCC1, ABCC10, and ABCG2 transporters.
  • Antineoplastic agents are substrates for a particular MDR transporter.
  • Exemplary antineoplastic agents that are substrates for ABCB1 are presented in Table 1;
  • exemplary antineoplastic agents that are substrates for ABCG2 are presented in Table 2;
  • exemplary antineoplastic agents that are substrates for ABCC10 are presented in Table 3.
  • paclitaxel, vinblastine, and vincristine are substrates for the ABCB1 transporter, whereas cisplatin is not a substrate for the ABCB1 transporter.
  • flavopiridol, mitoxantrone, SN-38, and methotrexate are substrates for the ABCG2 transporter, whereas cisplatin is not a substrate for the ABCG2 transporter.
  • the present invention encompasses a method of treating multidrug resistance in cancer cells in a subject in need thereof by inhibiting ABCB1 transporter activity, comprising administering to the subject a therapeutically effective, nontoxic amount of a PDE5 inhibitor.
  • the PDE5 inhibitor may be selected from a variety of known PDE5 inhibitors.
  • the PDE5 inhibitor is sildenafil, tadalafil, or vardenafil.
  • the PDE5 inhibitor is administered in combination with a chemotherapeutic agent(s).
  • the chemotherapeutic (antineoplastic) agent(s) is selected from the group consisting of paclitaxel, vincristine, and vinblastine.
  • the present invention encompasses a method of treating multidrug resistance in cancer cells in a subject in need thereof by inhibiting ABCG2 transporter activity, comprising administering to the subject a therapeutically effective, nontoxic amount of a PDE5 inhibitor.
  • the PDE5 inhibitor is sildenafil.
  • the PDE5 inhibitor is administered in combination with a chemotherapeutic agent(s).
  • the chemotherapeutic (antineoplastic) agent(s) is selected from the group consisting of SN-38, flavopiridol, mitoxantrone, and/or methotrexate.
  • the present invention encompasses a method of treating multidrug resistance in cancer cells in a subject in need thereof by inhibiting ABCC10 transporter activity, comprising administering to the subject a therapeutically effective, nontoxic amount of a PDE5 inhibitor.
  • the PDE5 inhibitor is sildenafil or vardenafil.
  • the PDE5 inhibitor is administered in combination with a chemotherapeutic agent(s).
  • the chemotherapeutic (antineoplastic) agent(s) is selected from the group consisting of paclitaxel, docetaxel, vinblastine, and vincristine.
  • An additional embodiment of the invention comprises a method of treating cancer, e.g., multidrug resistant cancer, in a subject by inhibiting MDR transporter activity, e.g., ABCB1, ABCC10, or ABCG2 transporter activity, comprising administering to the subject a therapeutically effective amount of a chemotherapeutic agent and a PDE5 inhibitor.
  • MDR transporter activity e.g., ABCB1, ABCC10, or ABCG2 transporter activity
  • chemotherapeutic agents include vincristine, vinblastine, paclitaxel, docetaxel, SN-38, flavopiridol, mitoxantrone, and methotrexate.
  • PDE5 inhibitors include vardenafil, tadalafil, and sildenafil.
  • the method comprises treating multidrug resistant lung cancer using a combination of a PDE5 inhibitor and paclitaxel.
  • a “therapeutically effective amount” means an amount of a compound, alone or in a combination, required to treat, ameliorate, reduce or prevent multidrug resistance in cancer cells of a subject.
  • the therapeutically effective amount of an active compound(s), e.g., a PDE5 inhibitor(s) varies depending upon the route of administration, age, body weight, and general health of the subject. Ultimately, the attending physician or veterinarian will decide the appropriate amount and dosage regimen.
  • PDE5 inhibitors may be effective in treating other disorders with ABC transporter involvement (may be referred to as ABC-associated disorders), e.g., infectious disorders. It is known in the art that ABC transporters are localized in the membranes of pathogenic organisms, and therefore, are able to efflux drugs designed to target them. Therefore, one embodiment of the present invention encompasses a method of treating a multidrug-resistant infectious disease comprising administering to the subject a therapeutically effective amount of a PDE5 inhibitor. Similarly, the inventors contemplate that PDE5 inhibitors may be effective in treating inflammatory conditions, e.g., gout.
  • compositions comprising PDE5 inhibitors.
  • the compositions are suitable for pharmaceutical use and administration to patients.
  • the compositions can comprise PDE5 inhibitors and pharmaceutical excipients.
  • the compositions can also comprise PDE5 inhibitors and chemotherapeutic agents, i.e., antineoplastic agents, as well as pharmaceutical excipients.
  • pharmaceutical excipients include solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, etc., that are compatible with pharmaceutical administration. Use of these agents for pharmaceutically active substances is well known in the art.
  • the compositions may also contain other active compounds providing supplemental, additional, or enhanced therapeutic functions.
  • the pharmaceutical compositions may also be included in a container, pack, or dispenser, together with instructions for administration.
  • a pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Methods to accomplish such administration are known to those of ordinary skill in the art.
  • Pharmaceutical compositions may be topically or orally administered, or capable of transmission across mucous membranes. Examples of administration of a pharmaceutical composition include oral ingestion or inhalation. Administration may also be intravenous, intraperitoneal, intramuscular, intracavity, subcutaneous, cutaneous, or transdermal.
  • Solutions or suspensions used for intradermal or subcutaneous application typically include at least one of the following components: a sterile diluent, such as water, saline solution, fixed oils, polyethylene glycol, glycerine, propylene glycol, or other synthetic solvent; antibacterial agents, such as benzyl alcohol or methyl parabens; antioxidants, such as ascorbic acid or sodium bisulfite; chelating agents, such as ethylenediaminetetraacetic acid (EDTA); buffers, such as acetate, citrate, or phosphate; and tonicity agents, such as sodium chloride or dextrose.
  • a sterile diluent such as water, saline solution, fixed oils, polyethylene glycol, glycerine, propylene glycol, or other synthetic solvent
  • antibacterial agents such as benzyl alcohol or methyl parabens
  • antioxidants such as ascorbic acid or sodium bisulfite
  • chelating agents such as ethylenediamine
  • Solutions or suspensions used for intravenous administration include a carrier such as physiological saline, bacteriostatic water, CREMOPHOR EL® (BASF Corp., Ludwigshafen, Germany), ethanol, or polyol.
  • a carrier such as physiological saline, bacteriostatic water, CREMOPHOR EL® (BASF Corp., Ludwigshafen, Germany), ethanol, or polyol.
  • the composition must be sterile and fluid for easy syringability. Proper fluidity can often be obtained using lecithin or surfactants.
  • the composition must also be stable under the conditions of manufacture and storage.
  • Prevention from introduction or growth of microorganisms can be achieved with antibacterial and antifungal agents, e.g., parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, etc.
  • isotonic agents sucrose
  • polyalcohols e.g., mannitol and sorbitol
  • sodium chloride may be included in the composition.
  • Prolonged absorption of the composition can be accomplished by adding an agent that delays absorption, e.g., aluminum monostearate or gelatin.
  • Oral compositions include an inert diluent or edible carrier.
  • PDE5 inhibitors or combinations of PDE5 inhibitors and antineoplastic agents, can be incorporated with excipients and placed, e.g., in tablets, troches, capsules, or gelatin.
  • Pharmaceutically compatible binding agents or adjuvant materials can be included in the composition.
  • compositions may contain, for example, (1) a binder, such as microcrystalline cellulose, gum tragacanth or gelatin; (2) an excipient, such as starch or lactose; (3) a disintegrating agent, such as alginic acid, PRIMOJEL®, or corn starch; (4) a lubricant, such as magnesium stearate; (5) a glidant, such as colloidal silicon dioxide; and/or (6) a sweetening or flavoring agent.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose
  • a disintegrating agent such as alginic acid, PRIMOJEL®, or corn starch
  • a lubricant such as magnesium stearate
  • a glidant such as colloidal silicon dioxide
  • the composition may also be administered by a transmucosal or transdermal route.
  • Transmucosal administration can be accomplished by lozenges, nasal sprays, inhalers, or suppositories.
  • Transdermal administration can be accomplished with composition-containing ointments, salves, gels, or creams known in the art.
  • penetrants appropriate to the barrier to be permeated are used.
  • a PDE5 inhibitor either alone or in combination with a chemotherapeutic agent, may be delivered in an aerosol spray from a pressured container or dispenser, which contains a propellant (e.g., liquid or gas), or a nebulizer.
  • a propellant e.g., liquid or gas
  • a PDE5 inhibitor either alone or in combination with a chemotherapeutic agent, is prepared with carriers to protect the PDE5 inhibitor and/or the chemotherapeutic agent against rapid elimination from the body.
  • Biodegradable polymers e.g., ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid
  • Methods for the preparation of such formulations are known by those skilled in the art.
  • Liposomal suspensions can also be used as pharmaceutically acceptable carriers.
  • the liposomes can be prepared according to established methods known in the art.
  • PDE5 inhibitors and antineoplastic agents are administered in a formulation and via a route already utilized in the clinic for those particular agents.
  • sildenafil, vardenafil, and tadalafil are administered orally.
  • sildenafil, vardenafil, and tadalafil are administered intravenously.
  • compositions of the invention are administered in therapeutically effective amounts. Appropriate dosages can be determined by a physician based upon clinical indications. For example, the compositions may be given as a bolus dose or a continuous infusion.
  • Examples of dosage ranges for PDE5 inhibitors that can be administered to a subject include: 1 ⁇ g/kg to 20 mg/kg, 1 ⁇ g/kg to 10 mg/kg, 1 ⁇ g/kg to 1 mg/kg, 10 ⁇ g/kg to 1 mg/kg, 10 ⁇ g/kg to 100 ⁇ g/kg, 100 ⁇ g/kg to 1 mg/kg, 250 ⁇ g/kg to 2 mg/kg, 250 ⁇ g/kg to 1 mg/kg, 500 ⁇ g/kg to 2 mg/kg, 500 ⁇ g/kg to 1 mg/kg, 1 mg/kg to 2 mg/kg, 1 mg/kg to 5 mg/kg, 5 mg/kg to 10 mg/kg, 10 mg/kg to 20 mg/kg, 15 mg/kg to 20 mg/kg, 10 mg/kg to 25 mg/kg, 15 mg/kg to 25 mg/kg, 20 mg/kg to 25 mg/kg, and 20 mg/kg to 30 mg/kg (or higher).
  • the dosage range of PDE5 is 10 ⁇ g/kg to 3 mg/kg. These dosages may be administered daily, weekly, biweekly, monthly, or less frequently, for example, biannually, depending on dosage, method of administration, cancer(s) to be treated, and individual subject characteristics.
  • Dosage unit form refers to physically discrete units suited for the patient.
  • Each dosage unit contains a predetermined quantity of PDE5 inhibitor, either alone or in combination with a chemotherapeutic agent, calculated to produce a therapeutic effect in association with the carrier.
  • the dosage unit depends on the characteristics of the PDE5 inhibitor and/or the chemotherapeutic agent, and the particular therapeutic effect to be achieved.
  • One embodiment of the present invention comprises a method of increasing sensitivity of a multidrug resistant cancer cell to a chemotherapeutic agent by inhibiting MDR transporter activity, e.g., ABCB1, ABCC10, or ABCG2 transporter activity, comprising contacting the cancer cell with a PDE5 inhibitor.
  • the method of increasing sensitivity of a multidrug resistant cancer cell may be performed in vivo, e.g., in a human or an animal cancer patient, or in an animal cancer model (e.g., a cancer xenograft in a mouse).
  • the method of increasing sensitivity of a multidrug resistant cancer cell may also be performed in vitro, e.g., in a cultured cancer cell, e.g., a primary or immortalized cancer cell.
  • a cultured cancer cell may be a cell that overexpresses a particular MDR transporter, e.g., ABCB1, ABCC10, or ABCG2 transporter.
  • exemplary embodiments of PDE5 inhibitors include but are not limited to sildenafil, vardenafil, and tadalafil.
  • Another embodiment of the present invention comprises a method of inhibiting growth (also referred to as proliferation) of a multidrug resistant cancer cell by inhibiting MDR transporter activity, e.g., ABCB1, ABCC10, or ABCG2 transporter activity, comprising contacting the cell with a combination of a chemotherapeutic agent and a PDE5 inhibitor.
  • MDR transporter activity e.g., ABCB1, ABCC10, or ABCG2 transporter activity
  • inhibition of cell growth or proliferation is presented as the concentration of the drug, e.g., an antineoplastic drug, required to inhibit the growth of cells, e.g., drug resistant cancer cells, by 50%, otherwise known as the IC 50 of the drug.
  • Assays for measuring inhibition of cell growth e.g., inhibition due to treatment with an antineoplastic agent or a combination of an antineoplastic agent and a PDE5 inhibitor, are known in the art.
  • a skilled artisan can use an MTT cytotoxicity assay described in the Examples.
  • a skilled artisan can use a colony formation assay.
  • the invention comprises a method of increasing accumulation of a chemotherapeutic agent in a cancer cell by inhibiting MDR transporter activity, e.g., ABCB1, ABCC10, or ABCG2 transporter activity, comprising contacting the cell with a PDE5 inhibitor.
  • MDR transporter activity e.g., ABCB1, ABCC10, or ABCG2 transporter activity
  • the invention comprises a method of decreasing efflux of a chemotherapeutic agent from a cancer cell by inhibiting MDR transporter activity, e.g., ABCB1, ABCC10, or ABCG2 transporter activity, comprising contacting the cell with a PDE5 inhibitor.
  • MDR transporter activity e.g., ABCB1, ABCC10, or ABCG2 transporter activity
  • the invention comprises a method of stimulating ATPase activity of an MDR transporter, e.g., ABCB1, ABCC10, or ABCG2 transporter, in a cell comprising contacting the cell with a PDE5 inhibitor.
  • MDR transporter e.g., ABCB1, ABCC10, or ABCG2 transporter
  • PDE5 inhibitors can stimulate the ATPase activity of MDR transporters.
  • the ability of PDE5 inhibitors to stimulate ATPase activity of MDR transporters demonstrates that PDE5 inhibitors serve as substrates for the MDR transporters, thereby inhibiting the efflux of other agents, e.g., chemotherapeutic agents.
  • ATPase activity of a transporter e.g., stimulation of the ATPase activity of a transporter, based on, for example, Ambudkar et al. (1998) Methods Enzymol. 292:504-14.
  • the invention also comprises a method of identifying a PDE5 inhibitor capable of blocking multidrug resistance in cancer cells comprising administering a PDE5 inhibitor to a cancer cell, wherein the PDE5 inhibitor is administered in a combination with a chemotherapeutic agent, and determining at least one of the following: (1) increased inhibition of growth of the cancer cell in the presence of the PDE5 inhibitor, in comparison with the same determination in the absence of the PDE5 inhibitor; (2) increased accumulation of the chemotherapeutic agent in the cancer cell in the presence of the PDE5 inhibitor, in comparison with the same determination in the absence of the PDE5 inhibitor; and (3) decreased efflux of the chemotherapeutic agent from the cancer cell in the presence of the PDE5 inhibitor, in comparison with the same determination in the absence of the PDE5 inhibitor.
  • the method of identifying a PDE5 inhibitor capable of blocking multidrug resistance in a cancer cell comprises: (a) a step of administering to the cancer cell a chemotherapeutic agent known to induce multidrug resistance in that cancer cell; (b) a step of measuring either (1) growth of the cancer cell, (2) accumulation of the chemotherapeutic agent in the cancer cell, or (3) efflux of the chemotherapeutic agent from the cancer cell; (c) a step of administering a PDE5 inhibitor to the cancer cell; (d) a step of measuring either (1) growth of the cancer cell, (2) accumulation of the chemotherapeutic agent in the cancer cell, or (3) efflux of the chemotherapeutic agent from the cancer cell in the presence of the PDE5 inhibitor; and (e) a step of comparing the measurements in steps (b) and (d), wherein a determination of (1) decreased growth of the cancer call, (2) increased accumulation of the chemotherapeutic agent in the cancer cell, or (3) decreased efflux of the chemotherapeutic agent from
  • the step of contacting the cell is carried out in vivo, in vitro, or ex vivo.
  • the step of contacting a cell is carried out in a human or nonhuman vertebrate subject, e.g., a mammalian subject, e.g., a human subject.
  • Vardenafil and tadalafil were purchased from Toronto Research Chemicals Inc. (Ontario, Canada).
  • [ 3 H]-paclitaxel 37.9 Ci/mmol was purchased from Moravek Biochemicals Inc (Brea, Calif.).
  • Monoclonal antibody C-219 (against ABCB1) was acquired from Signet Laboratories Inc. (Dedham, Mass.).
  • GPDH Anti-glyceraldehyde-3-phosphate dehydrogenase
  • 14C10 Anti-glyceraldehyde-3-phosphate dehydrogenase (GAPDH) monoclonal antibody (14C10) was obtained from Cell Signaling Technology, Inc. (Danvers, Mass.). Fumitremorgin C (FTC) was synthesized by Thomas McCloud Developmental Therapeutics Program, Natural Products Extraction Laboratory, NCI, NIH (Bethesda, Md.). ONO1078 was a gift from Dr. Akiyama (Kagoshima University, Japan).
  • Paclitaxel vincristine (VCR), colchicine, 7-ethyl-10-hydroxy-20 (S)-camptothecin (SN-38), verapamil, dimethyl sulfoxide (DMSO), 1-(4,5-dimethylthiazol-2-yl)-3,5-diphenylformazan (MTT) and all other chemicals were purchased from Sigma Chemical Company.
  • the ABCB1/Pgp-overexpressing drug-resistant cell line KB-C2 was established in a cell culture medium by a step-wise selection of the parental human epidermoid carcinoma cell line KB-3-1 using colchicine at concentrations up to 2 ⁇ g/ml.
  • the KB-C2 and KB-3-1 were kindly provided by Dr. Akiyama (Kagoshima University, Japan).
  • HEK293-pcDNA3.1 and wild-type HEK/ABCG2 transfected cells were established by selection with G418 after transfecting HEK293 with either (1) empty pcDNA3.1 vector or (2) pcDNA3.1 vector containing full length of ABCG2 coding arginine (R) at amino acid position 482, and were then cultured in a medium with 2 mg/ml of G418 (ABCG2-482-R2 cells).
  • the HEK/MRP1 (ABCC1) and HEK/ABCB1 cells were generated by transfecting the HEK293 cells with either the MRP1 expression vector or the ABCB1 expression vector.
  • DMEM Dulbecco's modified Eagle's medium
  • the MTT assay was used to assess cytotoxicity.
  • the cultured cells were harvested with trypsin and resuspended in a final concentration of 4 ⁇ 10 3 cells/well for KB-3-1; 7.5 ⁇ 10 3 cells/well for KB-C2; and 8 ⁇ 10 3 for all of the other cell lines used in this study. Cells were seeded evenly in 96-well multiplates.
  • different concentrations of chemotherapeutic drugs (20 ⁇ l/well) were added into designated wells after 1 h with or without exposure to potential reversal compounds: vardenafil, tadalafil, verapamil, ONO-1078, or FTC (20 ⁇ l/well).
  • the degree of resistance was calculated by dividing the IC 50 (concentrations required to inhibit growth, i.e., proliferation, by 50%) of a chemotherapeutic agent in the resistant cells by the IC 50 of the chemotherapeutic agent in the parental sensitive cells.
  • the degree of the reversal of multidrug resistance (MDR) was calculated by dividing the IC 50 of an anticancer drug, e.g., an antineoplastic or chemotherapeutic drug (agent, compound), in a cell type in the absence of a reversal agent by the IC 50 of the anticancer drug in the same cell type in the presence of the reversal agent.
  • the IC 50 values were calculated from survival curves using the Bliss method.
  • the intracellular accumulation of [ 3 H]-paclitaxel was measured as follows. Confluent cells in 24-well plates were preincubated with or without the reversal agents, e.g., vardenafil, tadalafil, or verapamil, for 1 h at 37° C. Intracellular paclitaxel accumulation was measured by incubating cells with 0.1 ⁇ M [ 3 H]-paclitaxel for 2 h in the presence or absence of the reversal agents at 37° C. The cells were washed three times with ice-cold PBS, then suspended in fresh medium with or without reversal agents at 37° C.
  • the reversal agents e.g., vardenafil, tadalafil, or verapamil
  • KB-C2 cells were incubated with 10 ⁇ M vardenafil or tadalafil for 0, 36 and 72 h. Following incubation, the cells were harvested and rinsed twice with ice-cold PBS, and total cell lysates were collected with cell lysis buffer (1 ⁇ PBS, 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS, 100 ⁇ g/ml phenylmethylsulfonyl fluoride, 10 ⁇ g/ml aprotinin, 10 ⁇ g/ml leupeptin) for 30 min with gentle rocking, and clarified by centrifugation at 12,000 rpm for 10 min at 4° C.
  • cell lysis buffer (1 ⁇ PBS, 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS, 100 ⁇ g/ml phenylmethylsulfonyl fluoride, 10 ⁇ g/ml aprotinin, 10 ⁇ g/ml leupept
  • Equal amounts (100 ⁇ g of protein) of cell lysates were resolved by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and electrophoretically transferred onto polyvinylidene fluoride (PVDF) membranes.
  • PVDF polyvinylidene fluoride
  • the membranes were washed three times for 15 min with TBST buffer and incubated at room temperature for 2 h with HRP-conjugated secondary antibody at 1:1000 dilutions.
  • the membranes underwent three additional washes for 15 min with TBST buffer and the protein-antibody complexes were visualized by the enhanced PHOTOTOPE® HRP Detection Kit (Cell Signaling Technology, Inc.) and exposed to Kodak medical X-ray processor (Eastman Kodak Co., Rochester, N.Y.).
  • HRP-conjugated secondary antibody 1:1000 dilutions.
  • the membranes underwent three additional washes for 15 min with TBST buffer and the protein-antibody complexes were visualized by the enhanced PHOTOTOPE® HRP Detection Kit (Cell Signaling Technology, Inc.) and exposed to Kodak medical X-ray processor (Eastman Kodak Co., Rochester, N.Y.).
  • Kodak medical X-ray processor Eastman Kodak
  • vardenafil or tadalafil at 10 mM were added into the wells. After incubation for 72 h with vardenafil or tadalafil, cells were washed with PBS and fixed with 4% paraformaldehyde for 15 min at room temperature and then rinsed with PBS three times.
  • the monoclonal antibody C219 against ABCB1 (1:500) was added for overnight incubation, followed by incubation for 1 h in Alexa Fluor 488 goat antimouse IgG (1:1000; Molecular Probe, Carlsbad, Calif.). Propidium iodide was used for nuclear staining.
  • the Vi-sensitive ATPase activity of ABCB1 in membrane vesicles of High FiveTM insect cells was measured as follows.
  • the membrane vesicles (10 ⁇ g of protein) were incubated in ATPase assay buffer (50 mM MES [pH 6.8], 50 mM KCl, 5 mM sodium azide, 2 mM EGTA, 2 mM dithiothreitol, 1 mM ouabain, and 10 mM MgCl 2 ) with or without 0.3 mM orthovanadate (freshly prepared) at 37° C. for 5 min, then incubated with different concentrations of drug at 37° C. for 3 min.
  • ATPase assay buffer 50 mM MES [pH 6.8], 50 mM KCl, 5 mM sodium azide, 2 mM EGTA, 2 mM dithiothreitol, 1 mM ouabain, and 10 mM MgCl 2
  • the ATPase reaction was started by the addition of 5 mM ATP, and the total volume was 0.1 ml. After incubation at 37° C. for 20 min, the reactions were stopped by the addition of 0.1 ml of 5% SDS solution and vortexed and kept at room temperature. The liberated Pi was measured as described in Ambudkar et al., supra.
  • Vardenafil, tadalafil (modeled as R,R isomer), and IAAP were constructed using the fragment dictionary of Maestro 9.0 and the energy minimized by Macromodel program v9.7 (Schrödinger, Inc., New York, N.Y.) using the OPLSAA force field (as described Jorgensen et al. (1996) J. Am. Chem. Soc. 118:11225-36), with the steepest descent followed by a truncated Newton conjugate gradient protocol. Partial atomic charges were computed using the OPLS-AA force field.
  • the low-energy 3D structures of vardenafil, tadalafil, and IAAP were generated with the following parameters present in LigPrep v2.3: different protonation states at physiological pH, all possible tautomers and ring conformations.
  • the X-ray crystal structure of mouse ABCB1 in the apoprotein state (PDB ID: 3G5U) and in complex with inhibitors QZ59-RRR (PDB ID: 3G6O) and QZ59-SSS (PDB ID: 3G61) was obtained from the RCSB Protein Data Bank; these were used to build the homology model of human ABCB1.
  • the homology modeling was conducted using the default parameters of Prime v2.1 as implemented in Maestro 9.0.
  • the input file for the amino acid sequence of human ABCB1 in the Prime structure prediction application was obtained as a FASTA file (UniProt Accession Number P08183.3) extracted from uniprot.org.
  • the initial structure thus obtained was refined by means of default parameters mentioned in a protein preparation facility implemented in Maestro v9.0 and Impact program v5.5 (Schrödinger, Inc., New York, N.Y.), in which the protonation states of residues were adjusted to the dominant ionic forms at pH 7.4.
  • the refined human ABCB1 homology model was further used to generate four different receptor grids by selecting QZ59-RRR (site-1) and QZ59-SSS (site-2) bound ligands, all amino acid residues known to contribute to verapamil binding (site-3), and two residues known to be common to the three previous sites (site-4), as shown in Table 7.
  • the docking calculations were performed using the “Extra Precision” (XP) mode of Glide program v5.5 (Schrödinger, Inc.) and the default parameters.
  • the top scoring pose-ABCB1 complex was then subjected to energy minimization using Macromodel program v9.7 using the OPLS-AA force field and used for graphical analysis. All computations were carried out on a Dell Precision 470n dual processor with the Linux OS (Red Hat Enterprise WS 4.0).
  • Vardenafil Enhances Drug Sensitivity of ABCB1- but Not ABCC1- or ABCG2-Overexpressing Cancer Cells
  • vardenafil or tadalafil were tested.
  • vardenafil produced a concentration-dependent increase in cytotoxicity of colchicine and paclitaxel in ABCB1-overexpressing drug-selected cell line KB-C2, but not parental sensitive KB-C2 cells (Table 4).
  • the effect of vincristine and paclitaxel cytotoxicity on ABCB1-transfected HEK293/ABCB1 cells was also measured.
  • Vardenafil increased the sensitivity of ABCB1-transfected HEK/ABCB1 cells to vincristine and paclitaxel (Table 5), but not the control cell line HEK293/pcDNA3.1. Therefore, the effect of vardenafil was specific to ABCB1-overexpressing cells, but had no significant toxic effects on parental cells when combined with ABCB1 transporter substrate anticancer drugs. Vardenafil did not reverse MDR induced by cells expressing ABCC1 and ABCG2 (Table 6), or significantly alter the IC 50 values of cisplatin, which is not a substrate of ABCB1 (Tables 4 and 5).
  • Vardenafil Increases Accumulation of Intracellular Paclitaxel in ABCB1-Overexpressing Cells by Inhibiting Drug Efflux
  • FIG. 1B The effect of vardenafil on paclitaxel efflux was also determined ( FIG. 1B ).
  • the intracellular levels of paclitaxel were measured over 2 h.
  • a significantly higher concentration of paclitaxel was effluxed from the KB-C2 cells compared to KB-3-1 cells, and the amount of effluxed paclitaxel increased with time; at 1 h time point, 70% of accumulated paclitaxel was effluxed from KB-C2 cells in the absence of vardenafil, and 10 ⁇ M of vardenafil significantly blocked ABCB1 efflux function, with 75% of accumulated paclitaxel being retained inside KB-C2 cells. No significant change in concentration of effluxed paclitaxel in parental KB-3-1 cells in the absence or presence of vardenafil was noted.
  • the ABCB1-overexpressing KB-C2 cells were incubated with 10 ⁇ M vardenafil or tadalafil for 36 and 72 h and then assayed to determine the difference in protein levels via Western blot.
  • the protein level of ABCB1 in KB-C2 cells was not significantly altered after the cells were incubated with vardenafil or tadalafil. It was possible that the ABCB1 transporter on the membrane had translocated inside the cell as a result of treatment, and the Western blot of whole lysates could not distinguish this possibility.
  • the drug-efflux function of ABCB1 is coupled to ATP hydrolysis by the ATPase enzyme that is usually stimulated in the presence of ABCB1 substrates. Therefore, to assess the effect of vardenafil and tadalafil on the ATPase activity of ABCB1, the rate of ABCB1-mediated ATP hydrolysis was measured in isolated membrane vesicles in the presence of various concentrations of vardenafil and tadalafil under conditions that suppressed activity of other major ATPases. Vardenafil produced a concentration-dependent increase in the ATPase activity of ABCB1 over a range of concentrations ( FIG.
  • the PDE5 inhibitors vardenafil and tadalafil are described for the first time as ABCB1 inhibitors. Their predicted binding conformation within the large cavity of ABCB1 required determination. Because the crystal structure of the human ABCB1 remains to be elucidated and the binding conformation of vardenafil and tadalafil within the large cavity of ABCB1 transporter is unknown, a homology model of human ABCB1, based on the mouse ABCB1-QZ59-RRR cocrystal structure as a template ( FIG. 3A ), was utilized for the Glide docking study of vardenafil and tadalafil.
  • the XP-Glide-predicted binding mode of vardenafil indicates the importance of hydrophobic and electrostatic interactions within the large drug binding cavity of ABCB1 ( FIG. 3B , left panel).
  • the N-ethylpiperazine (D-ring) of vardenafil forms hydrophobic contacts with Met69, Phe336, Leu339, and Ile340
  • the C-ring along with its ethoxy substituent enters into favorable hydrophobic interactions with Phe72, Leu975, and Phe978.
  • the A-ring, along with its methyl and propyl substituents and the B-ring are engaged in hydrophobic interactions with the side chains of Phe728, Ala729, Phe732, and Val982.
  • vardenafil also appears to form favorable electrostatic interactions with residues Tyr953 and Tyr307.
  • the sulfonyl oxygen atom forms a hydrogen bond with the hydroxyl group of Tyr953 (—SO 2 —HO-Tyr953), whereas the carbonyl function of the B-ring is located at a distance of 4.0 ⁇ from the side chain hydroxyl group of Tyr307.
  • the XP-Glide-predicted binding mode of tadalafil in site-4 of the large drug binding cavity of ABCB1 is shown in FIG. 3B (right panel).
  • the A- and B-rings of the indole moiety bind to the hydrophobic pocket formed by the side chains of Phe303, Leu304, Tyr307, and Phe343.
  • Phe343 also has hydrophobic contacts with the B-, C-, and E-rings. Both E- and F-rings of the benzodioxole moiety are surrounded by the side chains of Phe336, Leu339, Phe978, and Val982.
  • the carbonyl oxygen atom (close to E-ring) of the D-ring is stabilized by a hydrogen bonding interaction with the side chain amide group of Gln725 (—CO—H 2 NOC-Gln725).
  • the lower efficacy of tadalafil compared to vardenafil may be due to the orientation of its hydrophobic N-methyl substituent of the D-ring towards the unfavorable polar backbone of Met986 and Gly989 and the polar side chain amide group of Gln990.
  • [ 3 H]-paclitaxel (37.9 Ci/mmol), [ 3 H]-mitoxantrone (4 Ci/mmol), and [ 3 H]-methotrexate (23 Ci/mmol) were purchased from Moravek Biochemicals, Inc.
  • [ 3 H]-E 2 17 ⁇ G (40.5 Ci/mmol) and [ 125 I]-IAAP (2,200 Ci/mmol) were obtained from PerkinElmer Life Sciences.
  • the fluorescent compound BODIPY-prazosin was purchased from Invitrogen.
  • Monoclonal antibodies C-219 (against ABCB1) and BXP-21 (against ABCG2) were acquired from Signet Laboratories, Inc.
  • Sildenafil was purified from 100 mg VIAGRA® tablets as described by Francis et al. (2003) J. Impot. Res. 15:369-72. Fumitremorgin C (FTC) was synthesized by Thomas McCloud Developmental Therapeutics Program, Natural Products Extraction Laboratory, National Cancer Institute (NCI), NIH. Other chemicals were purchased from Sigma Chemical Co.
  • the ABCB1-overexpressing drug-resistant cell line KB-C2 was established by stepwise selection of the parental human epidermoid carcinoma cell line KB-3-1 in increasing concentrations of colchicine and was cultured in medium containing 2 ⁇ g/mL of colchicine.
  • the ABCB1-overexpressing drug resistant cell line KB-V1 (generously provided by Dr. Gottesman, NCI, NIH) was established by a stepwise concentration increase of vinblastine in the culture medium of KB-3-1 cells and subsequently culturing these cells in medium with 1 ⁇ g/mL of vinblastine.
  • An ABCC1-overexpressing MDR cell line, KB-CV60 was also derived from KB-3-1 cells and was maintained in medium with 1 ⁇ g/mL of cepharanthine and 60 ng/mL of vincristine. Both KB-C2 and KB-CV60 cell lines were kindly provided by Dr. Akiyama (Kagoshima University, Japan).
  • HEK293/pcDNA3.1, ABCG2-482-R5, ABCG2-482-G2, and ABCG2-482-T7 cells were established by selection with 2 mg/mL G418 after transfecting HEK293 with either (1) empty pcDNA3.1 vector or (2) pcDNA3.1 vector containing full length ABCG2 coding either (2a) wild-type arginine (R) or mutant (2b) glycine (G) or (2c) threonine (T) at amino acid position 482, respectively.
  • the wild-type ABCG2-overexpressing drug-resistant cell line MCF-7/Flv1000 was cultured in the medium with 1 ⁇ M of flavopiridol.
  • the mutated ABCG2-overexpressing drug resistant cell line MCF-7/ADVP3000 was maintained in the medium with 5 ⁇ g/mL of verapamil and 3 ⁇ g/mL of doxorubicin.
  • Another G482 mutant ABCG2-overexpressing drug resistant cell line, S1-M1-80 was maintained in the medium with 80 ⁇ M of mitoxantrone.
  • DMEM Dulbecco's modified Eagle's medium
  • bovine serum at 37° C. in a humidified atmosphere of 5% CO 2 .
  • Flow cytometric assays were carried out as follows. The cells were trypsinized and then resuspended in complete media (phenol red-free Iscove's modified Eagle's medium, IMEM, with 10% fetal calf serum) containing 250 nM BODIPY-prazosin alone or with various concentrations of the inhibitors for 30 min at 37° C. Cells were then washed once in cold complete medium and then incubated for another 1 h at 37° C. in substrate-free media with or without the same described concentrations of the inhibitors to generate the efflux histograms.
  • complete media phenol red-free Iscove's modified Eagle's medium, IMEM, with 10% fetal calf serum
  • Transport assays were carried out using the rapid filtration method as described in Chen et al. (2003) Cancer Res. 63:4048-54. Membrane vesicles were incubated with various concentrations of inhibitors for 1 h on ice, and then transport reactions were carried out at 37° C.
  • samples were passed through 0.22 ⁇ m GVWP filters (Millipore) presoaked in the stop solution.
  • the filters were washed three times with 3 mL of ice-cold stop solution. Radioactivity was measured by liquid scintillation counting.
  • the vanadate (Vi)-sensitive ATPase activity of ABCB1 or ABCG2 in the membrane vesicles of HIGH FIVETM insect cells was measured as described in Ambudkar et al., supra.
  • the membrane vesicles (100 ⁇ g of protein/mL) were incubated in ATPase assay buffer (50 mM MES, pH 6.8, 50 mM KCl, 5 mM sodium azide, 2 mM EGTA, 2 mM dithiothreitol, 1 mM ouabain, and 10 mM MgCl 2 ) with or without 0.3 mM vanadate at 37° C. for 5 min, and then incubated with different concentrations of drugs at 37° C.
  • ABCB1 and ABCG2 were immunoprecipitated using BXP-21 antibodies as described in Shukla et al., supra. Both ABCB1 and ABCG2 samples were subjected to SDS-PAGE in a 7% Trisacetate NuPAGE gel, the gel was dried, and exposed to Bio-Max MR film (Eastman Kodak Co.) at ⁇ 70° C. for 8 to 12 h. The radioactivity incorporated into the ABCB1 or ABCG2 band was quantified using the STORM 860 PhosphorImager System and ImageQuaNT (Molecular Dynamics).
  • Sildenafil and IAAP structures were built using the fragment dictionary of Maestro 9.0 and energy minimized by Macromodel program v9.7 (2009; Schrödinger, Inc.) using the OPLS-AA force field (Jorgensen et al., supra) with the steepest descent followed by a truncated Newton conjugate gradient protocol. Partial atomic charges were computed using the OPLS-AA force field.
  • the low-energy 3D structures of sildenafil and IAAP were generated with the following parameters present in LigPrep v2.3: different protonation states at physiologic pH, all possible tautomers and ring conformations.
  • the X-ray crystal structure of ABCB1 in apoprotein state (PDB ID: 3G5U) and in complex with inhibitors QZ59-RRR (PDB ID: 3G6O) and QZ59-SSS (PDB ID: 3G61) obtained from the RCSB Protein Data Bank were used to build the homology model of human ABCB1. Aller et al., supra. Homology modeling was carried out using the default parameters of Prime v2.1 as implemented in Maestro 9.0.
  • the input file for the amino acid sequence of human ABCB1 in the Prime structure prediction application was obtained as a FASTA file (UniProt Accession number P08183.3) extracted from www.uniprot.org.
  • the refined human ABCB1 homology model was used further to generate four different receptor grids by selecting QZ59-RRR (site-1) and QZ59-SSS (site-2) bound ligands, all amino acid residues known to contribute to verapamil binding (site-3), and two residues known to be common to three previous sites (site-4), as shown in Table 11.
  • sildenafil moderately increased the sensitivity (in KB-C2 and KB-V1 cells) to all three drugs, and at 10 ⁇ M, sildenafil increased the sensitivity with efficacy comparable to that of the equivalent concentration of verapamil. Neither sildenafil nor verapamil altered the cytotoxicity of the three drugs in parental KB-3-1 cells.
  • the IC 50 values of cisplatin which is not a substrate for ABCB1 and exhibited equal sensitivity in KB-3-1, KB-C2, and KB-V1 cells, were not affected by either sildenafil or verapamil in these three cell lines.
  • sildenafil On ABCC1- and ABCG2-mediated drug resistance were determined.
  • sildenafil at 10 ⁇ M showed no significant reduction in the IC 50 value of vincristine, a known ABCC1 substrate (not shown).
  • MCF-7/Flv1000 and MCF-7/AdVp3000 cells Compared with parental MCF-7 cells, MCF-7/Flv1000 and MCF-7/AdVp3000 cells exhibited high levels of resistance to ABCG2 substrates flavopiridol, mitoxantrone, and SN-38, but not to non-ABCG2 substrate cisplatin (Table 9). Similarly, the IC 50 values for flavopiridol, mitoxantrone, and SN-38 in S1-M1-80 were significantly higher than in parental S1 cells.
  • sildenafil decreased the IC 50 values for flavopiridol, mitoxantrone, and SN-38 in the ABCG2-expressing cell lines down to levels observed when cytotoxicity assays were carried out in the presence of the known specific ABCG2 inhibitor FTC at 2.5 ⁇ M.
  • the IC 50 values of these ABCG2 substrate drugs were not significantly different in the parental cells or the S1/FLV5000 cells (which do not express ABCG2, but also are resistant to flavopiridol in the presence or absence of sildenafil).
  • the non-ABCG2 substrate cisplatin was used, its IC 50 values were not affected by either sildenafil or FTC in any of the cell lines.
  • FIGS. 8-11 show that the survival curves in the presence of sildenafil were shifted remarkably to the left in the ABCB1- or ABCG2-overexpressing cells. Based on the above results, it appears that sildenafil significantly inhibits ABCB1-mediated drug efflux and partially reverses ABCG2-mediated efflux.
  • b Data are expressed as means ⁇ SD of at least three independent experiments carried out in triplicate.
  • c Fold-resistance values (values in parentheses) were calculated by dividing all IC 50 values obtained for colchicine, vinblastine, paclitaxel, and cisplatin by the respective IC 50 value for colchicine, vinblastine, paclitaxel, and cisplatin in the KB-3-1 cells in the absence of a reversal agent.
  • b Data are expressed as means ⁇ SD of at least three independent experiments performed in triplicate.
  • c Fold-resistance values (values in parentheses) were calculated by dividing all IC 50 values obtained for flavopiridol, mitoxantrone, SN-38, and cisplatin by the respective IC 50 value for flavopiridol, mitoxantrone, SN-38, and cisplatin in the S1 cells in the absence of a reversal agent.
  • b Data are expressed as means ⁇ SD of at least three independent experiments carried out in triplicate.
  • c Fold-resistance values (values in parentheses) were calculated by dividing all IC 50 values obtained for flavopiridol, mitoxantrone, SN-38, and cisplatin by the respective IC 50 value for flavopiridol, mitoxantrone, SN-38, and cisplatin in the HEK293/pcDNA3 cells in the absence of a reversal agent.
  • Sildenafil Increases Accumulation of [ 3 H]-Paclitaxel in ABCB1-Overexpressing Cells, and [ 3 H]-Mitoxantrone and BODIPY-Prazosin in ABCG2-Overexpressing Cells
  • sildenafil at 10 ⁇ M significantly increased the intracellular level of [ 3 H]-paclitaxel, close to the effect of verapamil at 10 ⁇ M in KB-C2 and KB-V1 cells. Neither sildenafil nor verapamil altered the intracellular levels of [ 3 H]-paclitaxel in parental KB-3-1 cells. Similarly, the intracellular levels of [ 3 H]-mitoxantrone were measured in the ABCG2-overexpressing cells in the presence or absence of sildenafil ( FIG. 4B-D ). The intracellular levels of [ 3 H]-mitoxantrone in cells expressing either wild-type or mutant ABCG2 were significantly less than that in parental cells.
  • sildenafil In the presence of sildenafil, either at 10 or 50 ⁇ M, all ABCG2-overexpressing cell lines displayed elevated intracellular [ 3 H]-mitoxantrone levels, and intracellular levels of [ 3 H]-mitoxantrone increased with increasing concentrations of sildenafil. However, the effects of sildenafil at 50 ⁇ M were less than those observed for FTC at 2.5 ⁇ M. Neither sildenafil nor FTC affected the intracellular levels of [ 3 H]-mitoxantrone in parental cells.
  • sildenafil at 50 ⁇ M enhanced the accumulation of BODIPY-prazosin in cells expressing either wild-type or mutant ABCG2, but demonstrated weaker enhancement than that of FTC at 2.5 ⁇ M.
  • Representative histograms for BODIPY-prazosin are shown in FIGS. 5A-D .
  • sildenafil significantly inhibited the rates of both methotrexate and E 2 17 ⁇ G uptake in the membrane vesicles of wild-type ABCG2 in a concentration-dependent manner, but its inhibitory effect was weaker than that of FTC at the same concentration.
  • FTC also moderately decreased the uptake rates of both methotrexate and E 2 17 ⁇ G in the membrane vesicles of HEK293/pcDNA3.1, but sildenafil did not.
  • the substrates of ABC transporters stimulate their ATPase activity, and among the reversal agents, some (e.g., verapamil) stimulate the activity, whereas others (e.g., cyclosporine A) inhibit ATP hydrolysis (Ambudkar et al. (1999) Annu. Rev. Pharmacol Toxicol. 39:361-98).
  • some e.g., verapamil
  • others e.g., cyclosporine A
  • ATP hydrolysis Ambudkar et al. (1999) Annu. Rev. Pharmacol Toxicol. 39:361-98.
  • the membrane vesicles of HIGH FIVETM insect cells overexpressing ABCB1 or ABCG2 were used in the presence of various concentrations of sildenafil under conditions that suppressed the activity of other major membrane ATPases. As shown in FIGS.
  • sildenafil at the indicated concentrations potently stimulated the ATPase activity of ABCB1, but mildly stimulated the ATPase activity of ABCG2.
  • concentrations of sildenafil required for 50% stimulation of ATPase activity of ABCB1 and ABCG2 were 5-10 and 0.25-0.5 ⁇ M, respectively. These results indicate that sildenafil may be a substrate for ABCB1 and ABCG2.
  • sildenafil interacts at the prazosin binding site of ABCB1 or ABCG2
  • the ability of sildenafil to prevent photolabeling of ABCB1 and ABCG2 with [ 125 I]-IAAP was examined by using the membrane vesicles from HIGH FIVETM insect cells transfected with ABCB1 or ABCG2. As shown in FIGS.
  • sildenafil dose dependently inhibited the photoaffinity labeling of ABCB1 or ABCG2 with [ 125 I]-IAAP.
  • the 50% inhibition of the photoaffinity labeling of ABCB1 and ABCG2 with [ 125 I]-IAAP by sildenafil was observed at sildenafil concentrations of 10 and 100 ⁇ M, respectively.
  • sildenafil competes with IAAP to bind both ABCB1 and ABCG2, and that sildenafil interacts with the transmembrane regions of both transporters.
  • PDE5A homology model of human ABCB1 based on the mouse (Mdr3) ABCB1-QZ59-RRR cocrystal structure was used to predict sildenafil-ABCB1 binding conformation.
  • a human ABCB1 homology model ( FIG. 7A ) was then used to dock sildenafil using Glide docking software to investigate its potential binding mode.
  • Three binding sites were reported in the crystal structure of mouse Mdr3 as represented by ABCB1-QZ59-RRR (site-1), ABCB1-QZ59-SSS (site-2), and ABCB1-verapamil (site-3). Aller et al., supra. To identify a favorable binding site on ABCB1 for sildenafil, docking experiments using these sites were carried out.
  • sildenafil displaces IAAP in a dose-dependent manner
  • IAAP was also docked to these sites for comparison.
  • binding energy data for the docked poses of sildenafil and IAAP were compared at each of the binding sites (Table 11).
  • both ligands sildenafil and IAAP were found to bind most favorably within the QZ59-RRR binding site of ABCB1. Bound conformation of sildenafil in context of site-1 is addressed below.
  • the XP-Glide-predicted binding mode of sildenafil shows the importance of hydrophobic interactions within the large drug binding cavity of ABCB1 ( FIG. 7B ).
  • the N-methylpiperazine (D-ring) of sildenafil was found to be stabilized by hydrophobic contacts with the side chains of Met69 of transmembrane (TM) 1 and Phe336, Leu339, and Ile340 of TM6.
  • the C-ring along with its ethoxy substituent enters into favorable hydrophobic interactions with residues Phe72 of TM1, Leu975 and Phe978 of TM12.
  • the A-ring (along with its methyl and propyl substituents) and the B-ring forms hydrophobic interactions with the side chains of Phe728, Ala729, and Phe732 of TM7 and Val982 of TM12.
  • sildenafil also seems to be stabilized by electrostatic interactions with key residues Tyr953 of TM11 and Tyr307 of TM5.
  • the sulfonyl oxygen atom enters into a hydrogen bonding interaction with the hydroxyl group of Tyr953 (—SO 2 —HO-Tyr953), whereas the carbonyl oxygen atom present in the B-ring is located at a distance of 3.9 ⁇ from the side chain hydroxyl group of Tyr307.
  • Transmembrane domain numbering is as reported previously (Aller et al., supra).
  • Paclitaxel as antineoplastic drug
  • saline as vehicle
  • Sildenafil was purified from 100 mg VIAGRA® tablets as described in Example 2.1.a.
  • KB-3-1 cells and the ABCB1/Pgp-overexpressing drug-resistant cell line KB-C2 cells were obtained as described in Example 2.1.b.
  • mice from Taconic (Hudson, N.Y.) were used for all xenografts. The mice were provided with sterilized food and water. The animal protocol was reviewed and approved by the Institutional Animal Care and Use Committee at St. John's University.
  • KB-3-1 or KB-C2 cells were grown in flasks, harvested and implanted (at 2.5 ⁇ 10 6 cells in 0.2 mL for KB-3-1 cells and 4.0 ⁇ 10 6 cells in 0.2 mL for KB-C2 cells) subcutaneously under the shoulder in the nude mice.
  • the mice were randomized into four groups, six mice per group: (1) vehicle, (2) sildenafil (given at 75 mg/kg p.o.), (3) paclitaxel (given at 18 mg/kg i.p.), and (4) paclitaxel and sildenafil, all treated every three days for a total of six administrations.
  • mice were euthanized when the tumor weight exceeded 1 gram in the control group, and the tumors were excised and weighed.
  • sildenafil in blocking MDR in vivo was evaluated using mouse tumor xenografts. As demonstrated in FIGS. 12 and 13 , there was a significant reduction in tumor weight and volume in mice treated with both sildenafil and paclitaxel in comparison with mice treated with paclitaxel alone.
  • Sildenafil, vardenafil, and tadalafil were purchased from Toronto Research Chemicals Inc. (Ontario, Canada). Cepharanthine was provided by Daiichi Sankyo Pharmaceutical Co. Ltd (Tokyo, Japan). Dulbecco's modified Eagle's medium (DMEM), fetal bovine serum (FBS), penicillin/streptomycin and trypsin 0.25% were products of Hyclone (Logan, Utah). Monoclonal antibody 14C10 (against GAPDH) was acquired from Cell Signaling Technology, Inc. (Danvers, Mass.). Polyclonal antibody D-19 (against MRP7) was obtained from Santa Cruz Biotechnology Inc (Santa Cruz, Calif.).
  • DMEM Dulbecco's modified Eagle's medium
  • FBS fetal bovine serum
  • trypsin 0.25% were products of Hyclone (Logan, Utah).
  • Monoclonal antibody 14C10 (against GAPDH) was acquired from
  • Alexa Flour 488 donkey anti-goat secondary antibody for immunocytochemistry was purchased from Molecular Probes (Eugene, Oreg.).
  • [ 3 H]-paclitaxel (46.5 Ci/mmol) was purchased from Moravek Biochemicals Inc (Brea, Calif.). Paclitaxel, docetaxel, vinblastine, DMSO, MTT, and other chemicals were purchased from Sigma Chemical (St. Louis, Mo.).
  • the MRP7 cDNA was generously provided by Dr. Gary Kruh (University of Illinois, Chicago, Ill.) and inserted into the pcDNA3.1 expression vector.
  • the MRP7 expression vector and parental plasmid were introduced into HEK293 cells by electroporation as previously described (Chen et al. (2003) Mol. Pharmacol. 63:351-58). Individual colonies were selected in medium containing G418 (1 mg/ml) and cultured for further analysis.
  • DMEM Dulbecco's modified Eagle's medium
  • MTT colorimetric assay was performed to analyze the drug sensitivity as previously described (Shi et al. (2007) Cancer Res. 67:11012-20).
  • HEK293-pcDNA3.1 and HEK/MRP7 cells were seeded into 96-well plates in triplicate at 6000 cells/well. After incubation in DMEM supplemented with 10% bovine serum at 37° C. for 24 h, three different nontoxic concentrations of sildenafil, vardenafil, and tadalafil (1.25, 2.5, and 5 ⁇ M) were added into plates 1 h prior to the addition of the substrates of MRP7 (paclitaxel, docetaxel, and vinblastine).
  • HEK293-pcDNA3.1 and HEK/MRP7 cells were trypsinized and four aliquots from each cell line were suspended in the medium, aliquots were preincubated with medium-only (control), sildenafil, vardenafil, or tadalafil (5 ⁇ M each) at 37° C. for 2 h, then incubated with 0.1 ⁇ M [ 3 H]-paclitaxel for another 2 h.
  • the cells were treated the same as in the drug accumulation study, and then washed three times with ice-cold PBS, and suspended in fresh medium with or without PDE5 inhibitors. Aliquots were evenly collected at various time points (0, 30, 60, and 120 min). Samples from both accumulation and efflux experiments were washed with ice-cold PBS three times and placed in scintillation fluid, and the radioactivity was measured in a Packard TRI-CARB® 1900CA liquid scintillation analyzer from Packard Instrument Company, Inc. (Downers Grove, Ill.).
  • HEK/MRP7 cells were incubated with 5 ⁇ M sildenafil, vardenafil, or tadalafil for different time periods (0, 24, 48 and 72 h), then harvested and rinsed twice with cold PBS.
  • the total cell lysates were collected with radioimmunoprecipitation assay (RIPA) buffer (1 ⁇ PBS, 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS, 100 ⁇ g/ml phenylmethylsulfonyl fluoride, 10 ⁇ g/ml aprotinin, 10 ⁇ g/ml leupeptin) for 30 min with occasional rocking followed by centrifugation at 12,000 rpm at 4° C. for 15 min.
  • RIPA radioimmunoprecipitation assay
  • the protein concentration was determined by a bicinchoninic acid (BCATM)-based protein assay (Thermo Scientific, Rockford, Ill.). Equal amounts of total cell lysates (40 ⁇ g of protein) were resolved by 4-12% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and electrophoretically transferred onto PVDF membranes.
  • BCATM bicinchoninic acid
  • SDS-PAGE 4-12% sodium dodecyl sulfate polyacrylamide gel electrophoresis
  • the membranes were washed three times for 15 min with TBST buffer and incubated at room temperature for 2 h with horseradish peroxide (HRP)-conjugated secondary antibody (1:2000 dilution).
  • HRP horseradish peroxide
  • the protein-antibody complex was detected by the enhanced Phototope TM-HRP Detection Kit (Cell Signaling Technology, Inc.) and exposed to Kodak medical X-ray processor (Eastman Kodak Co.).
  • the protein expression was quantified by Scion Image software (Scion Co., Frederick, Md.).
  • HEK/MRP7 cells (1 ⁇ 10 4 ) were seeded in 24 well plates and cultured overnight. Sildenafil, vardenafil, or tadalafil at 5 ⁇ M were added into the wells and then cultured at 37° C. for 72 h in a humidified incubator containing 5% CO 2 . Cells were washed with PBS and fixed with 4% paraformaldehyde for 15 min at room temperature and then rinsed with PBS three times. Nonspecific reaction was blocked with 1% BSA for 1 h at room temperature. A polyclonal antibody D19 against MRP7 (1:200) was added and incubated overnight.
  • HEK/MRP7 cells compared to parental HEK293-pcDNA3.1 cells exhibited a significant resistance to various MRP7 substrates such as paclitaxel, docetaxel, and vinblastine, which is consistent with previous reports (e.g., Shi et al. (2011) Cancer Res. 71:3029-41).
  • Sildenafil, vardenafil, and tadalafil dose-dependently decreased the IC 50 values of the above-mentioned MRP7 substrates for HEK/MRP7 cells.
  • Tadalafil showed the least reversal effect of the three PDE5 inhibitors.
  • Cepharanthine a known MRP7 inhibitor used as a positive control at 2.5 ⁇ M, completely reversed the resistance of HEK/MRP7 cells to paclitaxel, docetaxel, and vinblastine.
  • sildenafil, vardenafil, or tadalafil did not significantly reverse the resistance of HEK/MRP7 cells to cisplatin, a nonsubstrate of MRP7 (Table 12, FIG. 14 ).
  • Data are means ⁇ SD of at least three independent experiments performed in triplicate.
  • bFold-resistance was determined by dividing the IC 50 values of HEK/MRP7 cells by the IC 50 values of HEK293-pcDNA3.1 cells in the absence or presence of sildenafil, vardenafil, tadalafil or verapamil. *represents p ⁇ 0.05; **represents p ⁇ 0.01
  • PDE5 Inhibitors Increase the Intracellular Accumulation of [ 3 H]-Paclitaxel in HEK/MRP7 Cells
  • PDE5 Inhibitors Inhibit the Efflux of [ 3 H]-Paclitaxel Mediated by MRP7 in HEK/MRP7 Cells
  • Reversal of MRP7-mediated MDR can be achieved by either altering MRP7 expression or inhibiting MRP7 function.
  • MRP7 expression To evaluate the effects of sildenafil, vardenafil, and tadalafil on MRP7 expression, HEK/MRP7 cells were treated with sildenafil, vardenafil, or tadalafil at 5 ⁇ M for 0, 24, 48, and 72 h and the MRP7 expression levels were examined by Western blot analysis.
  • the results shown in FIG. 17A indicated that PDE5 inhibitors do not significantly alter the protein expression levels of MRP7 in HEK/MRP7 cells.
  • PDE5 Inhibitors do not Alter the Localization of MRP7
  • the ABC transporters could be downregulated if they were translocated or dislodged from the plasma membrane to a cytosolic region.
  • an immunofluorescence assay was performed to examine whether the location of MRP7 was altered after treatment with a PDE5 inhibitor. As shown in FIG. 17 , there was no alteration of MRP7 protein localization in plasma membranes after the treatment with sildenafil, vardenafil, or tadalafil at 5 ⁇ M for 72 h.

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to methods of treating multidrug resistance in cancerous cells with phosphodiesterase (PDE) inhibitors, e.g., PDE5 inhibitors. More specifically, the invention relates to methods of treating multidrug resistance that arises, e.g., during administration of chemotherapeutic/antineoplastic (anticancer) agents for treatment of cancer, with a PDE5 inhibitor (e.g., sildenafil, vardenafil, and tadalafil). The invention also relates to methods of treating cancer, e.g., multidrug resistant cancer, using a PDE5 inhibitor in combination with an antineoplastic therapeutic agent. Further, the invention relates to pharmaceutical compositions for treating multidrug resistant cancers comprising a PDE5 inhibitor, or a combination of a PDE5 inhibitor and an antineoplastic agent.

Description

    RELATED APPLICATIONS
  • This application is a continuation of U.S. application Ser. No. 13/432,315, filed Mar. 28, 2012, now U.S. Pat. No. 8,673,914, which claims the benefit of U.S. Provisional Patent Application No. 61/465,948, filed Mar. 28, 2011, and U.S. Provisional Patent Application No. 61/501,984, filed Jun. 28, 2011. The entire contents of all the above-mentioned applications are herein incorporated by reference.
  • STATEMENT AS TO FEDERALLY SPONSORED RESEARCH
  • This invention was made partially with Government support under NIH grant number 1R15CA143701. The Government has certain rights in the invention.
  • BACKGROUND OF THE INVENTION
  • 1. Field of the Invention
  • The present invention relates to a method of treating multidrug resistance with phosphodiesterase (PDE) inhibitors, e.g., PDE5 inhibitors. More specifically, the present invention relates to a method of treating multidrug resistance, e.g., multidrug resistance that arises, e.g., during administration of chemotherapeutic or antineoplastic agents for treatment of cancer, with a PDE5 inhibitor (e.g., vardenafil, sildenafil, and tadalafil). In addition, the invention relates to a method of treating cancer, e.g., multidrug resistant cancer, using a PDE5 inhibitor in combination with an anticancer agent, i.e., a chemotherapeutic/antineoplastic agent. Further, the invention relates to a pharmaceutical composition for treating a multidrug resistant cancer comprising a PDE5 inhibitor, or a combination of a PDE5 inhibitor and an antineoplastic agent.
  • 2. Description of Related Art
  • Multidrug resistance (MDR) is a phenomenon in which disease-causing organisms or cells are able to evade treatment with drugs designed to target them. Multidrug resistance to cancer chemotherapy, a major hurdle to successfully treating cancer, commonly arises due to the ability of one or more cancer cells to resist treatment with a variety of agents, e.g., drugs that may be distinct in one or both of the following: structure and mechanism of action. Therefore, whereas a portion of cancer cells, i.e., drug-sensitive cancer cells, are killed with a chemotherapeutic agent, drug-resistant cancer cells survive, such that the resultant or remaining cancers consist primarily of drug-resistant (or medicine-tolerant) cells.
  • One known cause of multidrug resistance to cancer chemotherapy is the overexpression of ATP-binding cassette (ABC) transporters in the membranes of cancers cells (Szakacs et al. (2006) Nat. Rev. Drug Discov. 5:219-34). These pumps efflux agents, including structurally and functionally unrelated chemotherapeutic agents, out of cancer cells, significantly decreasing the chance of successful treatment. In mammals, the ABC transporters have been divided into seven subfamilies (i.e., subfamilies A-G), based on genome sequence similarities (Gottesman et al. (2002) Nat. Rev. Cancer 2:48-58). Studies to date have consistently shown that the three major ABC transporters involved in multidrug resistance in most cancer cells are P-glycoprotein (Pgp/ABCB1/MDR1), breast cancer resistant protein (ABCG2/BCRP/ABCP), and members of the multidrug resistance protein family, e.g., multidrug resistant protein 1 (ABCC1/MRP1).
  • Overexpression of ABCB1 transporter occurs in 40-50% of all cancers; therefore, there is a major ongoing effort to design a strategy for inhibiting this transporter. Three generations of compounds that inhibit ABCB1 have been studied. The first generation of ABCB1 inhibitors included such drugs as verapamil, quinine, and cyclosporine A; however, these first generation agents produced undesirable adverse effects at concentrations necessary to inhibit ABCB1 (Krishna et al. (2000) Eur. J. Pharm. Sci. 11:265-83). The second generation of ABCB1 inhibitors, e.g., valspodar and biricodar, produced unpredictable interactions with other transport proteins and inhibited cytochrome P450 3A4 (CYP3A4), an enzyme responsible for metabolizing many xenobiotics, including chemotherapeutic drugs. Therefore, the use of the second generation ABCB1 inhibitors resulted in decreased clearance and increased toxicity of many chemotherapeutic agents (Gottesman et al., supra). The third generation of inhibitors, e.g., LY335979 (zosuquidar), GF120918 (elacridar), and MS-209 (dofequidar), were derived from second generation agents, and had nanomolar affinity for the ABCB1 transporter. However, to date third generation inhibitors have not been approved for use in patients due to several factors, including adverse side effects, unfavorable pharmacokinetic profiles, and/or lack of significant efficacy in late-phase clinical trials (Modok et al. (2006) Curr. Opin. Pharmacol. 6:350-54; Wu et al. (2009) Curr. Mol. Pharmacol. 1:93-105). Thus, it would be beneficial to determine whether other drugs, including drugs already used in the clinic, are capable of inhibiting ABC transporters and, therefore, can be used for the treatment of multidrug resistance.
  • Multidrug resistant protein 7 (MRP7; ABCC10), a member of MRP subfamily, is similar in topology to other MRPs (including MRP1) with two nucleotide-binding domains and three transmembrane domains (Kruh et al. (2007) Pflugers Arch. 453:675-84; Chen et al. (2003) Mol. Pharmacol. 63:351-58). ABCC10 is able to confer resistance to several natural product chemotherapeutic drugs, including taxanes and vinca alkaloids, which are also substrates of Pgp (Hopper-Borge et al. (2004) Cancer Res. 64:4927-30). ABCC10 has been reported to confer resistance to vinorelbine and paclitaxel in non-small cell lung cancer cells (Bessho et al. (2009) Oncol. Rep. 21:263-68; Oguri et al. (2008) Mol. Cancer Ther. 7:1150-55) and to vincristine in human salivary gland adenocarcinoma cells (Naramoto et al. (2007) Int. J. Oncol. 30:393-401). The in vivo functions of ABCC10 have recently been confirmed using an Mrp7 knockout mouse model (Hopper-Borge et al. (2011) Cancer Res. 71(10):3649-57).
  • Phosphodiesterase type 5 (PDE5) inhibitors are widely used in the treatment of male erectile dysfunction and in improving breathing efficiency in pulmonary hypertension. As agents already used in the clinic for other purposes, these drugs were investigated for their effects on MDR and ABC transporters.
  • SUMMARY OF THE INVENTION
  • The present invention relates to methods of treating multidrug resistance in a cancer cell in a subject in need thereof by inhibiting ABCB1 transporter activity, comprising administering to the subject a therapeutically effective amount of a phosphodiesterase 5 (PDE5) inhibitor. In one embodiment, the PDE5 inhibitor is administered in combination with an antineoplastic agent.
  • The present invention further relates to methods of treating a cancer in a subject by inhibiting ABCB1 transporter activity, comprising administering to the subject a therapeutically effective amount of an antineoplastic agent and a therapeutically effective amount of a PDE5 inhibitor. In one embodiment, the cancer is a multidrug resistant cancer.
  • In some embodiments of the above methods of treating, the subject is a mammal, e.g., a human.
  • In addition, the present invention relates to (1) methods of increasing sensitivity of a multidrug resistant cancer cell to an antineoplastic agent by inhibiting ABCB1 transporter activity, comprising contacting the cancer cell with a PDE5 inhibitor; (2) methods of inhibiting growth of a multidrug resistant cancer cell by inhibiting ABCB1 transporter activity, comprising contacting the cell with a combination of an antineoplastic agent and a PDE5 inhibitor; (3) methods of increasing accumulation of an antineoplastic agent in a cancer cell by inhibiting ABCB1 transporter activity, comprising contacting the cell with a PDE5 inhibitor; and (4) methods of decreasing efflux of an antineoplastic agent from a cancer cell by inhibiting ABCB1 transporter activity, comprising contacting the cell with a PDE5 inhibitor. In some embodiments, these methods are performed in vivo. For example, the methods are performed in a mammalian subject, e.g., a human subject. In further embodiments of the above methods, both the antineoplastic agent and the PDE5 inhibitor are administered in therapeutically effective amounts.
  • In some embodiments of the above methods related to ABCB1 transporter activity, the PDE5 inhibitor is selected from the group consisting of sildenafil, vardenafil, tadalafil, lodenafil, udenafil, benzamidenafil, mirodenafil, avanafil, zaprinast, SLX-2101, UK-371,800, UK-122764, icariin, DA-8159, 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]-pyrazin-2(1H)-one, and their analogs. In some embodiments of the above methods, the antineoplastic agent is selected from the group consisting of antineoplastic agents listed in Table 1; as a nonlimiting example, the antineoplastic agent can be selected from the group consisting of paclitaxel, vinblastine, and vincristine.
  • The present invention also relates to methods of treating multidrug resistance in a cancer cell in a subject in need thereof by inhibiting ABCG2 transporter activity, comprising administering to the subject a therapeutically effective amount of a PDE5 inhibitor. In one embodiment, the PDE5 inhibitor is administered in combination with an antineoplastic agent.
  • The present invention further relates to methods of treating a cancer in a subject by inhibiting ABCG2 transporter activity, comprising administering to the subject a therapeutically effective amount of an antineoplastic agent and a therapeutically effective amount of a PDE5 inhibitor. In one embodiment, the cancer is a multidrug resistant cancer.
  • In some embodiments of the above methods of treating, the subject is a mammal, e.g., a human.
  • Further, the present invention relates to (1) methods of increasing sensitivity of a multidrug resistant cancer cell to an antineoplastic agent by inhibiting ABCG2 transporter activity, comprising contacting the cancer cell with a PDE5 inhibitor; (2) methods of inhibiting growth of a multidrug resistant cancer cell by inhibiting ABCG2 transporter activity, comprising contacting the cell with a combination of an antineoplastic agent and a PDE5 inhibitor; (3) methods of increasing accumulation of an antineoplastic agent in a cancer cell by inhibiting ABCG2 transporter activity, comprising contacting the cell with a PDE5 inhibitor; and (4) methods of decreasing efflux of an antineoplastic agent from a cancer cell by inhibiting ABCG2 transporter activity, comprising contacting the cell with a PDE5 inhibitor. In some embodiments, these methods are performed in vivo. For example, the methods are performed in a mammalian subject, e.g., a human subject. In further embodiments of the above methods, both the antineoplastic agent and the PDE5 inhibitor are administered in therapeutically effective amounts.
  • In some embodiments of the above methods related to ABCG2 transporter activity, the PDE5 inhibitor is selected from the group consisting of sildenafil, vardenafil, tadalafil, lodenafil, udenafil, benzamidenafil, mirodenafil, avanafil, zaprinast, SLX-2101, UK-371,800, UK-122764, icariin, DA-8159, 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]-pyrazin-2(1H)-one, and their analogs. In some embodiments of the above methods, the antineoplastic agent is selected from the group of antineoplastic agents listed in Table 2; as a nonlimiting example, the antineoplastic agent can be selected from the group consisting of SN-38, flavopiridol, mitoxantrone, and methotrexate.
  • The present invention also relates to methods of treating multidrug resistance in a cancer cell in a subject in need thereof by inhibiting ABCC10 transporter activity, comprising administering to the subject a therapeutically effective amount of a PDE5 inhibitor. In one embodiment, the PDE5 inhibitor is administered in combination with an antineoplastic agent.
  • The present invention further relates to methods of treating a cancer in a subject by inhibiting ABCC10 transporter activity, comprising administering to the subject a therapeutically effective amount of an antineoplastic agent and a therapeutically effective amount of a PDE5 inhibitor. In one embodiment, the cancer is a multidrug resistant cancer.
  • In some embodiments of the above methods of treating, the subject is a mammal, e.g., a human.
  • Further, the present invention relates to (1) methods of increasing sensitivity of a multidrug resistant cancer cell to an antineoplastic agent by inhibiting ABCC10 transporter activity, comprising contacting the cancer cell with a PDE5 inhibitor; (2) methods of inhibiting growth of a multidrug resistant cancer cell by inhibiting ABCC10 transporter activity, comprising contacting the cell with a combination of an antineoplastic agent and a PDE5 inhibitor; (3) methods of increasing accumulation of an antineoplastic agent in a cancer cell by inhibiting ABCC10 transporter activity, comprising contacting the cell with a PDE5 inhibitor; and (4) methods of decreasing efflux of an antineoplastic agent from a cancer cell by inhibiting ABCC10 transporter activity, comprising contacting the cell with a PDE5 inhibitor. In some embodiments, these methods are performed in vivo. For example, the methods are performed in a mammalian subject, e.g., a human subject. In further embodiments of the above methods, both the antineoplastic agent and the PDE5 inhibitor are administered in therapeutically effective amounts.
  • In some embodiments of the above methods related to ABCC10 transporter activity, the PDE5 inhibitor is selected from the group consisting of sildenafil, vardenafil, tadalafil, lodenafil, udenafil, benzamidenafil, mirodenafil, avanafil, zaprinast, SLX-2101, UK-371,800, UK-122764, icariin, DA-8159, 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]-pyrazin-2(1H)-one, and their analogs. In some embodiments of the above methods, the antineoplastic agent is selected from the group of antineoplastic agents listed in Table 3; as a nonlimiting example, the antineoplastic agent can be selected from the group consisting of paclitaxel, docetaxel, vinblastine, and vincristine.
  • The invention also provides methods of stimulating the ATPase activity of an ABCB1 transporter in a cell comprising contacting the cell with a PDE5 inhibitor. In one embodiment, the PDE5 inhibitor is selected from the group consisting of sildenafil, vardenafil, tadalafil, lodenafil, udenafil, benzamidenafil, mirodenafil, avanafil, zaprinast, SLX-2101, UK-371,800, UK-122764, icariin, DA-8159, 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl) pyrido[3,4-b]-pyrazin-2(1H)-one, and their analogs.
  • The invention also provides methods of stimulating the ATPase activity of an ABCG2 transporter in a cell comprising contacting the cell with a PDE5 inhibitor. In one embodiment, the PDE5 inhibitor is selected from the group consisting of sildenafil, vardenafil, tadalafil, lodenafil, udenafil, benzamidenafil, mirodenafil, avanafil, zaprinast, SLX-2101, UK-371,800, UK-122764, icariin, DA-8159, 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl) pyrido[3,4-b]-pyrazin-2(1H)-one, and their analogs.
  • The invention also contemplates methods of stimulating the ATPase activity of an ABCC10 transporter in a cell comprising contacting the cell with a PDE5 inhibitor. In one embodiment, the PDE5 inhibitor is selected from the group consisting of sildenafil, vardenafil, tadalafil, lodenafil, udenafil, benzamidenafil, mirodenafil, avanafil, zaprinast, SLX-2101, UK-371,800, UK-122764, icariin, DA-8159, 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl) pyrido[3,4-b]-pyrazin-2(1H)-one, and their analogs.
  • Further, the present invention provides pharmaceutical compositions for treating a multidrug resistant cancer by inhibiting ABCB1 transporter activity in a subject comprising a PDE5 inhibitor and a pharmaceutical excipient. In one embodiment, the PDE5 inhibitor is selected from the group consisting of sildenafil, vardenafil, tadalafil, lodenafil, udenafil, benzamidenafil, mirodenafil, avanafil, zaprinast, SLX-2101, UK-371,800, UK-122764, icariin, DA-8159, 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]-pyrazin-2(1H)-one, and their analogs. In some embodiments, the pharmaceutical composition of the invention further comprises an antineoplastic agent. In some embodiments, the antineoplastic agent is selected from the group consisting of antineoplastic agents listed in Table 1.
  • Yet further, the present invention provides pharmaceutical compositions for treating a multidrug resistant cancer by inhibiting ABCG2 transporter activity in a subject comprising a PDE5 inhibitor and a pharmaceutical excipient. In one embodiment, the PDE5 inhibitor is selected from the group consisting of sildenafil, vardenafil, tadalafil, lodenafil, udenafil, benzamidenafil, mirodenafil, avanafil, zaprinast, SLX-2101, UK-371,800, UK-122764, icariin, DA-8159, 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl) pyrido[3,4-b]-pyrazin-2(1H)-one, and their analogs. In some embodiments, the pharmaceutical composition further comprises an antineoplastic agent. In some embodiments, the antineoplastic agent is selected from the group consisting of antineoplastic agents listed in Table 2.
  • Further, the present invention provides pharmaceutical compositions for treating a multidrug resistant cancer by inhibiting ABCC10 transporter activity in a subject comprising a PDE5 inhibitor and a pharmaceutical excipient. In one embodiment, the PDE5 inhibitor is selected from the group consisting of sildenafil, vardenafil, tadalafil, lodenafil, udenafil, benzamidenafil, mirodenafil, avanafil, zaprinast, SLX-2101, UK-371,800, UK-122764, icariin, DA-8159, 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]-pyrazin-2(1H)-one, and their analogs. In some embodiments, the pharmaceutical composition of the invention further comprises an antineoplastic agent. In some embodiments, the antineoplastic agent is selected from the group consisting of antineoplastic agents listed in Table 3. In some embodiments of the pharmaceutical composition of the present invention, the subject is a mammal, e.g., a human subject.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 depicts the effects of vardenafil and tadalafil on the accumulation (FIG. 1A) and efflux (FIG. 1B) of [3H]-paclitaxel, as well as ABCB1 expression (FIG. 1C) in ABCB1-overexpressing cells. FIG. 1A: The accumulation of [3H]-paclitaxel was measured after cells were preincubated with or without vardenafil, tadalafil, or verapamil for 1 h at 37° C., and then incubated with 0.1 μM [3H]-paclitaxel for another 2 h at 37° C. * and ** represent p<0.05 and p<0.01, respectively, for values for [3H]-paclitaxel accumulation in samples treated with vardenafil, tadalafil, and verapamil versus those in the control group. FIG. 1B The percentage of paclitaxel released was plotted as a function of time. KB-3-1 or KB-C2 cells received either no reversal agent treatment or were treated with vardenafil and verapamil throughout the experiment. Cells were pretreated for 1 h, coincubated with [3H]-paclitaxel, washed, and reincubated in paclitaxel-free medium. Cells were collected and levels of [3H]-paclitaxel determined either at 0, 60, or 120 min. Data points represent the means±standard deviations (SD) of triplicate determinations. FIG. 1C: Effect of vardenafil (left panel) or tadalafil (right panel) on the expression of ABCB1 for 36 and 72 h, respectively. Independent experiments were performed at least three times, and a representative experiment is shown.
  • FIG. 2 depicts the effects of vardenafil and tadalafil on the Vi-sensitive ATPase activity of ABCB1, represented as a function of ATP hydrolysis (FIG. 2A), and the photoaffinity labeling of ABCB1 with [125I]-IAAP (iodoarylazidoprazosin), represented as IAAP incorporation (FIG. 2B). Mean values are given, and the error bars represent standard error from at least three independent experiments.
  • FIG. 3A is ribbon diagram of open-to-the-cytoplasm 3D-structural conformation of a homology model of human ABCB1 based on the crystal structure coordinates of mouse ABCB1. The docked poses of tadalafil (shown on bottom), and vardenafil and IAAP (shown on top; overlapping) as ball and stick models are shown within the large hydrophobic cavity of ABCB1 at different inhibitor binding sites (see also Ding et al. (2011) PLoS ONE 6(4):e19329). FIG. 3B is the XP-Glide-predicted binding model of vardenafil (left panel) and tadalafil (right panel). Important amino acids are depicted as stick models, whereas the two inhibitors are shown as ball and stick models. The short dotted lines indicate hydrogen bonding interactions, whereas the long dotted lines indicate interacting distances.
  • FIG. 4 demonstrates that sildenafil increased the accumulation of [3H]-paclitaxel or [3H]-mitoxantrone in ABCB1- or ABCG2-overexpressing cells. The accumulation of [3H]-paclitaxel (FIG. 4A) or [3H]-mitoxantrone (FIGS. 4B, 4C, and 4D) was measured after cells were preincubated with or without sildenafil, verapamil, or FTC for 1 h at 37° C. and then incubated with 0.1 μM [3H]-paclitaxel or 0.02 μM [3H]-mitoxantrone for another 2 h at 37° C. Data points represent the means±SD of triplicate determinations. * and ** represent p<0.05 and p<0.01, respectively, for values versus those in the control group. Independent experiments were carried out at least three times, and a representative experiment is shown.
  • FIG. 5 demonstrates that sildenafil inhibited the efflux of BODIPY-prazosin (FIGS. 5A-5D) and transport of [3H]-E217βG as well as [3H]-methotrexate (FIGS. 5E and 5F) by ABCG2. In FIGS. 5A-5D, flow cytometric data (measured as cell numbers) from HEK293/pcDNA3.1, ABCG2-482-G2, ABCG2-482-R5, and ABCG2-482-T7 cells are shown after incubation with 250 nM BODIPY-prazosin alone (solid line) or with 50 μM sildenafil (dashed line), and 2.5 μM FTC (shaded histogram). In FIGS. 5E and 5F, membrane vesicles were prepared from HEK293/pcDNA3.1 and ABCG2-482-R5 cells, and the rates of the uptake of [3H]-E217βG and [3H]-methotrexate into membrane vesicles (10 μg protein/reaction) were measured. Data points represent the means±SD of triplicate determinations. * and ** represent p<0.05 and p<0.01, respectively, for values versus those in the control group. At least three independent experiments were carried out, and a representative experiment is shown.
  • FIG. 6 depicts the effect of sildenafil on the ATPase activity and [125I]-IAAP photoaffinity labeling of ABCB1 and ABCG2. The Vi-sensitive ATPase activities (measured as a function of ATP hydrolysis) of ABCB1 (FIG. 6A) and ABCG2 (FIG. 6B) in membrane vesicles were determined with different concentrations of sildenafil. Mean values are given, and the error bars represent standard error from at least three independent experiments. The photoaffinity labeling (measured as a function of IAAP incorporation) of ABCB1 (FIG. 6C) and ABCG2 (FIG. 6D) with [125I]-IAAP was conducted in the presence of different concentrations of sildenafil. The autoradiograms and quantification of incorporation of IAAP into ABCB1 and ABCG2 bands were obtained from at least two independent experiments. Cyclosporine A (CSA) and verapamil (Vera) were used as positive controls for inhibition of [125I]-IAAP photolabeling of ABCB1, and FTC was used as a positive control for ABCG2. * and ** represent p<0.05 and p<0.01, respectively, for values versus those in the control group.
  • FIG. 7 is a model for binding of sildenafil to ABCB1. FIG. 7A is a ribbon diagram of open-to-the-cytoplasm 3D-structural conformation of a homology model of human ABCB1 based on the crystal structure coordinates of mouse Mdr3. Sildenafil is shown as a ball and stick model within the large hydrophobic cavity of ABCB1 characterized by the QZ59-RRR inhibitor binding site (see also Shi et al. (2011) Cancer Research 71(8):1-13). FIG. 7B is an XP-Glide-predicted binding model of sildenafil within the QZ59-RRR binding site. Important amino acids are depicted as stick models, whereas the inhibitor is shown as a ball and stick model. The short dotted line indicates a hydrogen bonding interaction, whereas the long dotted line indicates an interacting distance.
  • FIG. 8 represents survival curves at different concentrations of colchicine (FIG. 8A), vinblastine (FIG. 8B), paclitaxel (FIG. 8C), and cisplatin (FIG. 8D) for either KB-3-1 cells with (open squares) or without (filled squares) 10 μM sildenafil; KB-C2 cells with (open diamonds) or without (filled diamonds) 10 μM sildenafil; or KB-V1 cells with (open triangles) or without (filled triangles) 10 μM sildenafil. Cell survival was determined by MTT assay; data points are the means±SD of triplicate determinations, with a representative experiment shown.
  • FIG. 9 represents survival curves at different concentrations of flavopiridol (FIG. 9A), mitoxantrone (FIG. 9B), SN-38 (FIG. 9C), and cisplatin (FIG. 9D) for either S1 cells with (open squares) or without (filled squares) 50 μM sildenafil; S1/FLV5000 cells with (open diamonds) or without (filled diamonds) 50 μM sildenafil; or S1-M1-80 cells with (open triangles) or without (filled triangles) 50 μM sildenafil. Cell survival was determined by MTT assay; data points are the means±SD of triplicate determinations, with a representative experiment shown.
  • FIG. 10 represents survival curves at different concentrations of flavopiridol (FIG. 10A), mitoxantrone (FIG. 10B), SN-38 (FIG. 10C), and cisplatin (FIG. 10D) for either MCF-7 cells with (open squares) or without (filled squares) 50 μM sildenafil; MCF-7/FLV10000 cells with (open diamonds) or without (filled diamonds) 50 μM sildenafil; or MCF-7/ADVP3000 cells with (open triangles) or without (filled triangles) 50 μM sildenafil. Cell survival was determined by MTT assay; data points are the means±SD of triplicate determinations, with a representative experiment shown.
  • FIG. 11 represents survival curves at different concentrations of flavopiridol (FIG. 11A), mitoxantrone (FIG. 11B), SN-38 (FIG. 11C), and cisplatin (FIG. 11D) for either HEK293/pcDNA3 cells with (open squares) or without (filled squares) 50 μM sildenafil; HEK293/ABCG2-G2 cells with (open diamonds) or without (filled diamonds) 50 μM sildenafil; HEK293/ABCG2-R5 cells with (open triangles) or without (filled triangles) 50 μM sildenafil, or HEK293/ABCG2-T7 cells with (open circles) or without (filled circles) 50 μM sildenafil. Cell survival was determined by MTT assay; data points are the means±SD of triplicate determinations, with a representative experiment shown.
  • FIG. 12 represents the effects of sildenafil treatment on the growth of tumor xenografts. Mice were treated with either vehicle, sildenafil alone, paclitaxel alone, or a combination of sildenafil and paclitaxel. FIG. 12A demonstrates the sizes of the excised tumors, and FIG. 12B demonstrates the tumor weight of the excised tumors. * represents p<0.05 for values versus those indicated.
  • FIG. 13 demonstrates the effects of sildenafil treatment on tumor volume in mouse xenografts, measured on days 0, 1, 3, 6, 9, 12, 15, and 18 after transplantation. Mice were treated with either vehicle, sildenafil alone, paclitaxel alone, or a combination of sildenafil and paclitaxel. * represents p<0.05 for values versus those in the vehicle group; *, # represents p<0.05 for values versus those in the paclitaxel alone group.
  • FIG. 14 demonstrates the effects of PDE5 inhibitors on the reversal of ABCC10-mediated drug resistance in HEK/MRP7 cells, showing the survival curves of HEK/MRP7 cells in the presence or absence of sildenafil, vardenafil, or tadalafil at 5 μM and the parental HEK293-pcDNA3.1 cells at different concentrations of paclitaxel (FIG. 14A), docetaxel (FIG. 14B), vinblastine (FIG. 14C), and cisplatin (FIG. 14D). Cell survival was determined by MTT assay as described herein. Data points represent the means±SD of triplicate determinations. Experiments were performed at least three independent times.
  • FIG. 15 shows the effects of sildenafil, vardenafil, and tadalafil on the accumulation of [3H]-paclitaxel in HEK293-pcDNA3.1 and HEK/MRP7 cells. The intracellular accumulation of [3H]-paclitaxel was measured by scintillation counting after cells were preincubated with or without sildenafil, vardenafil, tadalafil, or cepharanthine for 2 h at 37° C. and then incubated with 0.1 μM [3H]-paclitaxel for another 2 h at 37° C. Data points represent the means±SD of triplicate determinations. Experiments were performed at least three independent times. ** represents p<0.01 for values versus those in the control group.
  • FIG. 16 demonstrates the effects of sildenafil (FIG. 16A), vardenafil (FIG. 16B), and tadalafil (FIG. 16C) on the efflux of [3H]-paclitaxel in HEK293-pcDNA3.1 and HEK/MRP7 cells. Cells were preincubated with or without sildenafil, vardenafil, or tadalafil for 2 h at 37° C., and further incubated with 0.1 μM [3H]-paclitaxel for another 2 h at 37° C. Cells were then incubated in fresh medium with or without PDE5 inhibitors for different time periods at 37° C. Cells were then collected, and the intracellular levels of [3H]-paclitaxel were measured by scintillation counting. A time course versus percentage of intracellular [3H]-paclitaxel was plotted (0, 30, 60, and 120 min). Data points represent the means±SD of triplicate determinations. Experiments were performed at least three independent times.
  • FIG. 17 shows immunoblot detection (FIG. 17A) and immunofluorescence detection (FIG. 17B) of MRP7 in HEK/MRP7 cells following incubation with PDE5 inhibitors. Cell lysates were prepared from HEK/MRP7 cells incubated with 5 μM sildenafil, vardenafil, and tadalafil for different time periods (0, 24, 48, and 72 h). Immunoblot detection of MRP7 (FIG. 17A) was performed using polyclonal anti-MRP7 antibody; GAPDH was used as an internal control for equal loading. Equal amounts (40 μg of protein) of total cell lysates were used for each sample. The localization of MRP7 by immunofluorescence (FIG. 17B) was performed on paraformaldehyde-fixed cells using polyclonal antibody D19 against MRP7 (1:200) and Alexa Flour® 488 donkey anti-goat IgG (1:2000). Propidium iodide was used for nuclear counterstaining Results from a representative experiment are shown; similar results were obtained in two other trials.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is based on the finding that phosphodiesterase 5 (PDE5) inhibitors can be useful for treating multidrug resistance in cells, e.g., cancer cells.
  • Methods of Treatment
  • The present invention provides a method of treating multidrug resistance in cancer cells in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a PDE5 inhibitor.
  • As used herein, “multidrug resistance” (commonly abbreviated as “MDR”) refers to a phenomenon wherein disease-causing organisms or cells are able to evade treatment with agents designed to target them, often agents that are structurally or mechanistically distinct. As such, multidrug resistance can be viewed as simultaneous resistance to different drugs that have either the same or different targets, and either similar or distinct chemical structures. Organisms or cells may acquire a multidrug resistant phenotype upon exposure to a single drug, e.g., a chemotherapeutic/antineoplastic drug (also referred to herein as an “anticancer drug”). Multidrug resistance in cancer, or multidrug resistant cancer, refers to the situation wherein at least one cancer cell becomes resistant, or nonresponsive, to treatment with drugs, e.g., chemotherapeutic/antineoplastic drugs (agents, compounds, etc.), designed to target it, e.g., chemotherapeutic/antineoplastic drugs that may be structurally and/or mechanistically distinct. Therefore, cancer cells sensitive to the treatment with chemotherapeutic or antineoplastic drugs are killed, and the resistant cancer cell divides and propagates into a drug-resistant cancer growth.
  • The multidrug resistant phenotype in cancer cells is usually a result of either intrinsic and/or acquired overexpression of the ATP-binding cassette transporters (ABC transporters). ABC transporters are conserved proteins that translocate substrates across cellular membranes. There are nearly fifty known ABC transporters in the human genome, divided into seven families (i.e., A-G), with a variety of transport functions, including transport of biological compounds, toxins, xenobiotics and drugs.
  • As used herein, “MDR transporters” refers to ABC transporters involved in multidrug resistance in cancer cells. The three MDR transporters with previously known predominant function in multidrug resistance to cancer chemotherapeutics are ABCB1 (P-glycoprotein or Pgp), ABCC1 (multidrug resistance protein 1 or MRP1), and ABCG2 (breast cancer resistant protein or BCRP), with ABCB1 transporter overexpression occurring in nearly 50% of all cancers. In addition, as disclosed herein, ABCC10 (MRP7) has now been shown to function in multidrug resistance to cancer chemotherapeutics.
  • The types of cancers or metastases that may be affected by multidrug resistance, and thus may be treated using the methods of the present invention, include but are not limited to the following: cancers of the central nervous system, skin, respiratory tract, oral cavity, eyes, bone, skin, connective tissue, gastrointestinal tract, cardiovascular system (heart, vasculature, etc.), ear, sinuses, salivary glands, urethra, lips, bile duct, gall bladder, etc.; hematological-associated cancers (e.g., lymphomas, leukemias, myeloproliferative cancers, etc.); breast, ovarian, cervical, vaginal, uterine, testicular, renal, penile, bladder, prostate, nasopharyngeal, endocrine (thyroid, parathyroid, pituitary, adrenal, pancreatic, pineal), splenic, etc. cancers; as well as viral-associated cancers such as Kaposi's sarcoma, etc. In one embodiment of the invention, the cancer type that is affected by multidrug resistance and is thus to be treated with the methods of the present invention is lung cancer.
  • A subject in need thereof, as used herein, refers to any subject that is determined to be in need of therapy, e.g., therapy for the treatment of multidrug resistance with PDE5 inhibitors, by a treating physician (e.g., an oncologist). For example, a subject in need thereof may be a subject for whom a treatment with one or more chemotherapeutic agents was determined to be unsuccessful. The success of a treatment with any particular therapy is evaluated based on the ability of that therapy to eliminate or reduce tumor burden, i.e., to eradicate or diminish the number of cancer cells. The most suitable way to measure the elimination or reduction in tumor burden may vary by the type of cancer involved. A treating physician will know the most suitable way to measure elimination or reduction in tumor burden, e.g., through obtaining and analyzing a biopsy sample. A subject in need thereof may be a subject suffering from cancer, e.g., multidrug resistant cancer. In one embodiment, the subject in need thereof may be suffering from lung cancer.
  • A subject in need thereof may be a human or nonhuman animal. The term “nonhuman animal” includes all vertebrates, such as nonhuman primates, sheep, dogs, cows, chickens, amphibians, reptiles, etc. A subject may be a mammalian subject.
  • As described herein, the present invention relates to the treatment of multidrug resistance by administering a phosphodiesterase inhibitor, e.g., a PDE5 inhibitor. Phosphodiesterases are protein enzymes that function to break down a phosphodiester bond. Cyclic nucleotide phosphodiesterases, a subclass of phosphodiesterases, function to break down the phosphodiester bond in cyclic nucleotides, e.g., converting cyclic AMP (3′5′-cAMP; also commonly abbreviated as cAMP) and cyclic GMP (3′5′-cGMP; also commonly abbreviated as cGMP) to 5′-AMP and 5′-GMP, respectively; thereby inactivating these important second messengers and halting signal transduction cascades. Increased levels of cyclic GMP are associated with smooth muscle vasodilation, and thus increased blood flow. PDE5 is a phosphodiesterase responsible for converting second messenger cyclic GMP into 5′-GMP, thereby terminating its biological actions and potentially leading to vasoconstriction and reduced blood flow. Inhibitors of PDE5 increase cyclic GMP levels, and have been used in the clinic to treat erectile dysfunction and pulmonary arterial hypertension.
  • PDE5 inhibitors include, but are not limited to, vardenafil (LEVITRA®); sildenafil (VIAGRA®); tadalafil (CIALIS®); lodenafil (HELLEVA®); udenafil (ZYDENA®); benzamidenafil (Zou et al. (2008) J. Pharm. Biomed. Anal. 47:255-59); SLX-2101; mirodenafil; avanafil; UK-371,800 (Bunnage et al. (2008) Bioorg. Med. Chem. Lett. 18:6033-36); UK-122,764; zaprinast; 3-[4-(2-hydroxyethyl)piperazin-1-yl]-7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)pyrido[3,4-b]-pyrazin-2(1H)-one (Hughes et al. (2010) J. Med. Chem. 53:2656-60); icariin and its derivatives (e.g., 3,7-bis(2-hydroxyethyl)icaritin) (Dell'Agli et al. (2008) J. Nat. Prod. 71:1513-17); DA-8159 (Oh et al. (2000) Arch. Pharm. Res. (NY) 23:471-76); and their analogs. These and other known PDE5 inhibitors are further described in Hughes et al. (2009) Biorg. Med. Chem. Lett. 19:5209-13; Rotella et al. (2000) J. Med. Chem. 43:1257-63; Duan et al. (2009) Bioorg. Med. Chem. Lett. 19:2777-79; Giovannoni et al. (2006) J. Med. Chem. 49:5363-71; Yoo et al. (2007) Bioorg. Med. Chem. Lett. 17:4271-74; Daugan et al. (2003) J. Med. Chem. 46:4525-32; Arnold et al. (2007) Bioorg. Med. Chem. Lett. 17:2376-79; and Eros et al. (2008) Curr. Med. Chem. 15:1570-85, all incorporated by reference herein in their entireties.
  • As taught herein, PDE5 inhibitors, e.g., inhibitors such as sildenafil, tadalafil, and vardenafil, are useful in blocking multidrug resistance in cancer, and thereby increasing the efficacy of antineoplastic agents. Thus, because the present invention shows that PDE5 inhibitors are capable of reversing multidrug resistance, for the purposes of this invention, PDE5 inhibitors, as well as other agents known to reverse multidrug resistance, are sometimes referred to as “reversal agents.” PDE5 inhibitors can increase the efficacy of antineoplastic drugs in several ways. PDE5 inhibitors can block the efflux of antineoplastic agents through MDR transporters. For example, it is known that the ATPase activity of ABC transporters, e.g., MDR transporters, is stimulated in the presence of their transport substrates, e.g., either their natural transport substrates or antineoplastic drugs. As shown herein, the ATPase activity of the MDR transporters is stimulated in the presence of PDE5 inhibitors, e.g., sildenafil, tadalafil, and vardenafil. Therefore, in one embodiment, PDE5 inhibitors may act as transport substrates of MDR transporters (e.g., competitive inhibitors of MDR transporters), thereby reducing the ability of MDR transporters to efflux antineoplastic agents. By increasing the efficacy of antineoplastic agents, PDE5 inhibitors can decrease cancer cell survival, thus having a positive effect on the outcome of cancer treatment.
  • PDE5 inhibitors can also positively affect cancer treatment because PDE5 inhibitors block cGMP hydrolysis, and thus increase cGMP levels, which may lead to activation of protein kinase G (PKG). Activation of PKG in turn leads to growth suppression or apoptosis.
  • The methods and compositions of the present invention encompass the use of specific or selective PDE5 inhibitors. However, one skilled in the art will understand that a nonspecific PDE5 inhibitor (e.g., a PDE inhibitor that blocks the function of other cGMP phosphodiesterases, in addition to its ability to block PDE5) may be effective in blocking MDR transporters, such as ABCB1, ABCC10, and ABCG2. In addition, one skilled in the art will understand that a PDE inhibitor that is able to block the function of other cyclic nucleotide phosphodiesterases (e.g., cyclic GMP phosphodiesterases), exclusive of an ability to block PDE5, may be effective in blocking MDR transporters such as ABCB1, ABCC10, and ABCG2. Examples of phosphodiesterases that are able to catalyze cyclic GMP to 5′-GMP conversion include, but are not limited to, PDE1, PDE1B, PDE1C, PDE2A, PDE6A, PDE6B, PDE6C, PDE9A, PDE10A, and PDE11A.
  • MDR transporters are expressed in the membranes of various cell types (both normal and cancer cells), including cell types present at sites of drug absorption, e.g., intestinal epithelial cells; therefore, in one embodiment of the invention, a PDE5 inhibitor may block multidrug resistance, at least in part, by blocking the ability of MDR transporters expressed on epithelial cells to efflux antineoplastic drugs, thereby aiding drug absorption. In another embodiment of the invention, a PDE5 inhibitor blocks multidrug resistance by blocking the ability of MDR transporters expressed on cancer cells to efflux antineoplastic drugs.
  • Depending on the type of MDR transporter involved, it may be preferable to use a certain PDE5 inhibitor to block antineoplastic drug efflux. For example, when ABCG2 transporter-mediated efflux is involved, it may be preferable to use, e.g., sildenafil, because the ABCG2 transporter is relatively insensitive to inhibition by other PDE5 inhibitors, e.g., vardenafil and tadalafil.
  • Numerous chemotherapeutic agents (antineoplastic agents) have been developed for treating cancer. In the clinic, depending on cancer diagnosis, different antineoplastic agents are preferred. Antineoplastic agents used for treating cancers, or under investigation for use in treating cancers, may include, but are not limited to: (1) nitrogen mustards, such as mechlorethamine, cyclophosphamide, ifosfamide, melphalan (L-sarcolysin, L-PAM), uramustine, and chlorambucil; (2) ethylenimines and methylmelamines (aziridines), such as thioTEPA, hexamethylmelamine (HMM, altretamine), and triethylenemelamine (TEM); (3) nitrosoureas, such as carmustine (BCNU), lomustine (CCNU), semustine (Methyl-CCNU), fotemustine, and streptozotocin; (4) alkyl sulfonates, such as busulfan; (5) azines and hydrazines (triazenes), such as dacarbazine (DTIC) and procarbazine (MATULANE®); (6) platins, such as cisplatin, carboplatin and oxaliplatin; (7) pteridines, such as methotrexate; (8) pyrimidine analogs, such as 5-fluorouracil, fluorodeoxyuridine (floxuridine, FUDR®), cytarabine (AraC), and gemcitabine (GEMZAR®); (9) purine analogs and related inhibitors, such as 6-mercaptopurine (6-MP), thioguanine (6-TG), and pentostatin; (10) antimitotic drugs, for example, (a) vinca alkaloids, such as vinblastine, vincristine, vindesine, and vinorelbine; and (b) taxanes, such as paclitaxel and docetaxel; (11) camptothecins, such as topotecan, irinotecan, diflomotecan, and 9-aminocamptothecin; (12) epipodophyllotoxins, such as etoposide and teniposide; (13) antibiotics, for example, (a) actinomycins, such as actinomycin D (DACTINOMYCIN®), mitomycin, and plicamycin; (b) anthracyclines, such as daunorubicin (daunomycin), doxorubicin, idarubicin, epirubicin, valrubicin, bisantrene, and mitoxantrone; and (c) other antibiotics, e.g., azacitidine (5-azacytidine) and pentostatin (deoxycoformycin); (14) hormones and antagonists, for example, (a) adrenocorticosteroids, such as prednisone, dexamethasone, methylprednisolone, and mitotane (o,p′-DDD); (b) progestins, such as hydroxyprogesterone (17-hydroxyprogesterone), medroxyprogesterone, medroxyprogesterone acetate, megestrol, and megestrol acetate; (c) estrogens, such as DES and ethinyl estradiol; (d) antiestrogens, such as tamoxifen; (e) antiandrogens, such as flutamide; (f) androgens, such as testosterone and fluoxymesterone; (g) gonadotropin-releasing hormone analogs, such as leuprorelin (leuprolide acetate); and (h) aromatases, such as aminoglutethimide, anastrozole, letrozole, and exemestane; (15) heavy metal compounds, such as arsenic trioxide; (16) substituted ureas, such as hydroxyurea; (17) retinoids, such as tretinoin (retinoic acid), alitretinoin, and isotretinoin; (18) tyrosine kinase inhibitors, such as imatinib, gefitinib, lapatinib, erlotinib, sunitinib, nilotinib, vandetanib, dasatinib, afatinib, neratinib, axitinib, pazopanib, sorafenib, toceranib, lestaurtinib, cediranib, regorafenib, semaxanib, bosutinib, tandutinib, aparinib, AG1478, and crizotinib; (19) angiogenesis inhibitors, such as marimastat, angiostatin, endostatin, and thalidomide; (20) enzymes, such as L-asparaginase; (21) CDK inhibitors, such as flavopiridol, and (22) biologic response modifiers, such as natural and synthetic interferons. Exemplary antineoplastic agents of choice for various cancer diagnoses are summarized in, e.g., Basic and Clinical Pharmacology, pp. 945-46 (Bertram Katzung ed., Lange Medical Books/McGraw-Hill 8th ed. 2001); and Goodman and Gilman's The Pharmacological Basis of Therapeutics, pp. 1383-85 (Joel G. Hardman, Lee E. Limbird, and Alfred Goodman Gilman eds., McGraw-Hill Medical Publishing Division 10th Ed. 2001). Some sources use different groupings/classes to categorize the various antineoplastic or chemotherapeutic agents (drugs, compounds, etc.), but all appropriate antineoplastic agents are contemplated in the present invention.
  • Different agents, e.g., antineoplastic agents, are known to be substrates for different MDR transporters, e.g., ABCB1, ABCC1, ABCC10, and ABCG2 transporters. A skilled artisan will know which antineoplastic agents are substrates for a particular MDR transporter. Exemplary antineoplastic agents that are substrates for ABCB1 are presented in Table 1; exemplary antineoplastic agents that are substrates for ABCG2 are presented in Table 2; and exemplary antineoplastic agents that are substrates for ABCC10 are presented in Table 3. For example, a skilled artisan will know that paclitaxel, vinblastine, and vincristine are substrates for the ABCB1 transporter, whereas cisplatin is not a substrate for the ABCB1 transporter. Similarly, a skilled artisan will know that flavopiridol, mitoxantrone, SN-38, and methotrexate are substrates for the ABCG2 transporter, whereas cisplatin is not a substrate for the ABCG2 transporter.
  • TABLE 1
    Representative Antineoplastic Agents That
    Are Substrates for the ABCB1 Transporter
    Classical Antineoplastic Agents Tyrosine Kinase Inhibitors
    Vinblastine Tandutinib
    Vincristine Erlotinib
    Vinorelbine Dasatinib
    Vindesine Imatinib
    Paclitaxel Nilotinib
    Docetaxel Lapatinib
    Doxorubicin Sunitinib
    Daunorubicin Apatinib
    Idarubicin AG1478
    Bisantrene
    Mitoxantrone
    Etoposide
    Teniposide
    Dactinomycin
    Mitomycin
    Plicamycin
    Methotrexate
    Topotecan
    Irinotecan
    SN-38
  • TABLE 2
    Representative Antineoplastic Agents That
    Are Substrates for the ABCG2 Transporter
    Classical Antineoplastic Agents Tyrosine Kinase Inhibitors
    Aza-anthrapyrazole Tandutinib
    (BBR 3390)
    9-Aminocamptothecin Erlotinib
    Bisantrene Dasatinib
    Diflomotecan Gefitinib
    Doxorubicin Imatinib
    Daunorubicin Lapatinib
    Epirubicin Sorafenib
    Etoposide
    Flavopiridol
    GV-196771
    Irinotecan (CPT-11)
    J-107088
    Methotrexate and its polyglutamates
    Mitoxantrone
    NB-506
    Quinazoline
    SN-38
    Teniposide
    Topotecan
  • TABLE 3
    Representative Antineoplastic Agents That
    Are Substrates for the ABCC10 Transporter
    Classical Antineoplastic Agents Tyrosine Kinase Inhibitors
    Vinblastine Nilotinib
    Vincristine Erlotinib
    Vinorelbine Lapatinib
    Paclitaxel Imatinib
    Docetaxel Tandutinib
    Etoposide Masitinib
    Teniposide
    Dactinomycin
    SN-38
    Gemcitabine
    Cytarabine
    Epothilone-B
  • In one embodiment, the present invention encompasses a method of treating multidrug resistance in cancer cells in a subject in need thereof by inhibiting ABCB1 transporter activity, comprising administering to the subject a therapeutically effective, nontoxic amount of a PDE5 inhibitor. The PDE5 inhibitor may be selected from a variety of known PDE5 inhibitors. In one embodiment, the PDE5 inhibitor is sildenafil, tadalafil, or vardenafil. In another embodiment, the PDE5 inhibitor is administered in combination with a chemotherapeutic agent(s). In a further embodiment, the chemotherapeutic (antineoplastic) agent(s) is selected from the group consisting of paclitaxel, vincristine, and vinblastine.
  • In a different embodiment, the present invention encompasses a method of treating multidrug resistance in cancer cells in a subject in need thereof by inhibiting ABCG2 transporter activity, comprising administering to the subject a therapeutically effective, nontoxic amount of a PDE5 inhibitor. In one embodiment, the PDE5 inhibitor is sildenafil. In another embodiment, the PDE5 inhibitor is administered in combination with a chemotherapeutic agent(s). In a further embodiment, the chemotherapeutic (antineoplastic) agent(s) is selected from the group consisting of SN-38, flavopiridol, mitoxantrone, and/or methotrexate.
  • In a different embodiment, the present invention encompasses a method of treating multidrug resistance in cancer cells in a subject in need thereof by inhibiting ABCC10 transporter activity, comprising administering to the subject a therapeutically effective, nontoxic amount of a PDE5 inhibitor. In one embodiment, the PDE5 inhibitor is sildenafil or vardenafil. In another embodiment, the PDE5 inhibitor is administered in combination with a chemotherapeutic agent(s). In a further embodiment, the chemotherapeutic (antineoplastic) agent(s) is selected from the group consisting of paclitaxel, docetaxel, vinblastine, and vincristine.
  • An additional embodiment of the invention comprises a method of treating cancer, e.g., multidrug resistant cancer, in a subject by inhibiting MDR transporter activity, e.g., ABCB1, ABCC10, or ABCG2 transporter activity, comprising administering to the subject a therapeutically effective amount of a chemotherapeutic agent and a PDE5 inhibitor. Exemplary embodiments of chemotherapeutic agents include vincristine, vinblastine, paclitaxel, docetaxel, SN-38, flavopiridol, mitoxantrone, and methotrexate. Exemplary embodiments of PDE5 inhibitors include vardenafil, tadalafil, and sildenafil. In one embodiment, the method comprises treating multidrug resistant lung cancer using a combination of a PDE5 inhibitor and paclitaxel. A “therapeutically effective amount” means an amount of a compound, alone or in a combination, required to treat, ameliorate, reduce or prevent multidrug resistance in cancer cells of a subject. The therapeutically effective amount of an active compound(s), e.g., a PDE5 inhibitor(s), varies depending upon the route of administration, age, body weight, and general health of the subject. Ultimately, the attending physician or veterinarian will decide the appropriate amount and dosage regimen.
  • In addition to methods of treating cancer, the inventors contemplate that PDE5 inhibitors may be effective in treating other disorders with ABC transporter involvement (may be referred to as ABC-associated disorders), e.g., infectious disorders. It is known in the art that ABC transporters are localized in the membranes of pathogenic organisms, and therefore, are able to efflux drugs designed to target them. Therefore, one embodiment of the present invention encompasses a method of treating a multidrug-resistant infectious disease comprising administering to the subject a therapeutically effective amount of a PDE5 inhibitor. Similarly, the inventors contemplate that PDE5 inhibitors may be effective in treating inflammatory conditions, e.g., gout.
  • Pharmaceutical Compositions and Methods of Administration
  • Certain embodiments of the invention include compositions comprising PDE5 inhibitors. The compositions are suitable for pharmaceutical use and administration to patients. The compositions can comprise PDE5 inhibitors and pharmaceutical excipients. The compositions can also comprise PDE5 inhibitors and chemotherapeutic agents, i.e., antineoplastic agents, as well as pharmaceutical excipients. As used herein, “pharmaceutical excipients” include solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, etc., that are compatible with pharmaceutical administration. Use of these agents for pharmaceutically active substances is well known in the art. The compositions may also contain other active compounds providing supplemental, additional, or enhanced therapeutic functions. The pharmaceutical compositions may also be included in a container, pack, or dispenser, together with instructions for administration.
  • A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Methods to accomplish such administration are known to those of ordinary skill in the art. Pharmaceutical compositions may be topically or orally administered, or capable of transmission across mucous membranes. Examples of administration of a pharmaceutical composition include oral ingestion or inhalation. Administration may also be intravenous, intraperitoneal, intramuscular, intracavity, subcutaneous, cutaneous, or transdermal.
  • Solutions or suspensions used for intradermal or subcutaneous application typically include at least one of the following components: a sterile diluent, such as water, saline solution, fixed oils, polyethylene glycol, glycerine, propylene glycol, or other synthetic solvent; antibacterial agents, such as benzyl alcohol or methyl parabens; antioxidants, such as ascorbic acid or sodium bisulfite; chelating agents, such as ethylenediaminetetraacetic acid (EDTA); buffers, such as acetate, citrate, or phosphate; and tonicity agents, such as sodium chloride or dextrose. The pH can be adjusted with acids or bases by methods known in the art. Such preparations may be enclosed in, e.g., ampoules, disposable syringes, or multiple dose vials.
  • Solutions or suspensions used for intravenous administration include a carrier such as physiological saline, bacteriostatic water, CREMOPHOR EL® (BASF Corp., Ludwigshafen, Germany), ethanol, or polyol. In all cases, the composition must be sterile and fluid for easy syringability. Proper fluidity can often be obtained using lecithin or surfactants. The composition must also be stable under the conditions of manufacture and storage. Prevention from introduction or growth of microorganisms can be achieved with antibacterial and antifungal agents, e.g., parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, etc. In many cases, isotonic agents (sugar), polyalcohols (e.g., mannitol and sorbitol), or sodium chloride may be included in the composition. Prolonged absorption of the composition can be accomplished by adding an agent that delays absorption, e.g., aluminum monostearate or gelatin.
  • Oral compositions include an inert diluent or edible carrier. For the purpose of oral administration, PDE5 inhibitors, or combinations of PDE5 inhibitors and antineoplastic agents, can be incorporated with excipients and placed, e.g., in tablets, troches, capsules, or gelatin. Pharmaceutically compatible binding agents or adjuvant materials can be included in the composition. The compositions may contain, for example, (1) a binder, such as microcrystalline cellulose, gum tragacanth or gelatin; (2) an excipient, such as starch or lactose; (3) a disintegrating agent, such as alginic acid, PRIMOJEL®, or corn starch; (4) a lubricant, such as magnesium stearate; (5) a glidant, such as colloidal silicon dioxide; and/or (6) a sweetening or flavoring agent.
  • The composition may also be administered by a transmucosal or transdermal route. Transmucosal administration can be accomplished by lozenges, nasal sprays, inhalers, or suppositories. Transdermal administration can be accomplished with composition-containing ointments, salves, gels, or creams known in the art. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used. For administration by inhalation, a PDE5 inhibitor, either alone or in combination with a chemotherapeutic agent, may be delivered in an aerosol spray from a pressured container or dispenser, which contains a propellant (e.g., liquid or gas), or a nebulizer.
  • In certain embodiments, a PDE5 inhibitor, either alone or in combination with a chemotherapeutic agent, is prepared with carriers to protect the PDE5 inhibitor and/or the chemotherapeutic agent against rapid elimination from the body. Biodegradable polymers (e.g., ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid) are often used. Methods for the preparation of such formulations are known by those skilled in the art. Liposomal suspensions can also be used as pharmaceutically acceptable carriers. The liposomes can be prepared according to established methods known in the art.
  • In some embodiments of the invention, PDE5 inhibitors and antineoplastic agents are administered in a formulation and via a route already utilized in the clinic for those particular agents. For example, sildenafil, vardenafil, and tadalafil are administered orally. In another example, sildenafil, vardenafil, and tadalafil are administered intravenously.
  • The compositions of the invention are administered in therapeutically effective amounts. Appropriate dosages can be determined by a physician based upon clinical indications. For example, the compositions may be given as a bolus dose or a continuous infusion.
  • Examples of dosage ranges for PDE5 inhibitors that can be administered to a subject include: 1 μg/kg to 20 mg/kg, 1 μg/kg to 10 mg/kg, 1 μg/kg to 1 mg/kg, 10 μg/kg to 1 mg/kg, 10 μg/kg to 100 μg/kg, 100 μg/kg to 1 mg/kg, 250 μg/kg to 2 mg/kg, 250 μg/kg to 1 mg/kg, 500 μg/kg to 2 mg/kg, 500 μg/kg to 1 mg/kg, 1 mg/kg to 2 mg/kg, 1 mg/kg to 5 mg/kg, 5 mg/kg to 10 mg/kg, 10 mg/kg to 20 mg/kg, 15 mg/kg to 20 mg/kg, 10 mg/kg to 25 mg/kg, 15 mg/kg to 25 mg/kg, 20 mg/kg to 25 mg/kg, and 20 mg/kg to 30 mg/kg (or higher). In one embodiment, the dosage range of PDE5 is 10 μg/kg to 3 mg/kg. These dosages may be administered daily, weekly, biweekly, monthly, or less frequently, for example, biannually, depending on dosage, method of administration, cancer(s) to be treated, and individual subject characteristics.
  • In certain circumstances, it may be advantageous to formulate 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 for the patient. Each dosage unit contains a predetermined quantity of PDE5 inhibitor, either alone or in combination with a chemotherapeutic agent, calculated to produce a therapeutic effect in association with the carrier. The dosage unit depends on the characteristics of the PDE5 inhibitor and/or the chemotherapeutic agent, and the particular therapeutic effect to be achieved.
  • Additional Applications of the Invention
  • One embodiment of the present invention comprises a method of increasing sensitivity of a multidrug resistant cancer cell to a chemotherapeutic agent by inhibiting MDR transporter activity, e.g., ABCB1, ABCC10, or ABCG2 transporter activity, comprising contacting the cancer cell with a PDE5 inhibitor. The method of increasing sensitivity of a multidrug resistant cancer cell may be performed in vivo, e.g., in a human or an animal cancer patient, or in an animal cancer model (e.g., a cancer xenograft in a mouse). The method of increasing sensitivity of a multidrug resistant cancer cell may also be performed in vitro, e.g., in a cultured cancer cell, e.g., a primary or immortalized cancer cell. A cultured cancer cell may be a cell that overexpresses a particular MDR transporter, e.g., ABCB1, ABCC10, or ABCG2 transporter. Exemplary embodiments of PDE5 inhibitors include but are not limited to sildenafil, vardenafil, and tadalafil.
  • Another embodiment of the present invention comprises a method of inhibiting growth (also referred to as proliferation) of a multidrug resistant cancer cell by inhibiting MDR transporter activity, e.g., ABCB1, ABCC10, or ABCG2 transporter activity, comprising contacting the cell with a combination of a chemotherapeutic agent and a PDE5 inhibitor. Commonly, inhibition of cell growth or proliferation is presented as the concentration of the drug, e.g., an antineoplastic drug, required to inhibit the growth of cells, e.g., drug resistant cancer cells, by 50%, otherwise known as the IC50 of the drug. Assays for measuring inhibition of cell growth, e.g., inhibition due to treatment with an antineoplastic agent or a combination of an antineoplastic agent and a PDE5 inhibitor, are known in the art. In one example, a skilled artisan can use an MTT cytotoxicity assay described in the Examples. In another example, a skilled artisan can use a colony formation assay.
  • In yet another embodiment, the invention comprises a method of increasing accumulation of a chemotherapeutic agent in a cancer cell by inhibiting MDR transporter activity, e.g., ABCB1, ABCC10, or ABCG2 transporter activity, comprising contacting the cell with a PDE5 inhibitor.
  • In another embodiment, the invention comprises a method of decreasing efflux of a chemotherapeutic agent from a cancer cell by inhibiting MDR transporter activity, e.g., ABCB1, ABCC10, or ABCG2 transporter activity, comprising contacting the cell with a PDE5 inhibitor.
  • In another embodiment, the invention comprises a method of stimulating ATPase activity of an MDR transporter, e.g., ABCB1, ABCC10, or ABCG2 transporter, in a cell comprising contacting the cell with a PDE5 inhibitor. The present invention teaches that PDE5 inhibitors can stimulate the ATPase activity of MDR transporters. As described herein, the ability of PDE5 inhibitors to stimulate ATPase activity of MDR transporters demonstrates that PDE5 inhibitors serve as substrates for the MDR transporters, thereby inhibiting the efflux of other agents, e.g., chemotherapeutic agents. One skilled in the art will know how to measure the ATPase activity of a transporter, e.g., stimulation of the ATPase activity of a transporter, based on, for example, Ambudkar et al. (1998) Methods Enzymol. 292:504-14.
  • The invention also comprises a method of identifying a PDE5 inhibitor capable of blocking multidrug resistance in cancer cells comprising administering a PDE5 inhibitor to a cancer cell, wherein the PDE5 inhibitor is administered in a combination with a chemotherapeutic agent, and determining at least one of the following: (1) increased inhibition of growth of the cancer cell in the presence of the PDE5 inhibitor, in comparison with the same determination in the absence of the PDE5 inhibitor; (2) increased accumulation of the chemotherapeutic agent in the cancer cell in the presence of the PDE5 inhibitor, in comparison with the same determination in the absence of the PDE5 inhibitor; and (3) decreased efflux of the chemotherapeutic agent from the cancer cell in the presence of the PDE5 inhibitor, in comparison with the same determination in the absence of the PDE5 inhibitor. Specifically, in one embodiment, the method of identifying a PDE5 inhibitor capable of blocking multidrug resistance in a cancer cell comprises: (a) a step of administering to the cancer cell a chemotherapeutic agent known to induce multidrug resistance in that cancer cell; (b) a step of measuring either (1) growth of the cancer cell, (2) accumulation of the chemotherapeutic agent in the cancer cell, or (3) efflux of the chemotherapeutic agent from the cancer cell; (c) a step of administering a PDE5 inhibitor to the cancer cell; (d) a step of measuring either (1) growth of the cancer cell, (2) accumulation of the chemotherapeutic agent in the cancer cell, or (3) efflux of the chemotherapeutic agent from the cancer cell in the presence of the PDE5 inhibitor; and (e) a step of comparing the measurements in steps (b) and (d), wherein a determination of (1) decreased growth of the cancer call, (2) increased accumulation of the chemotherapeutic agent in the cancer cell, or (3) decreased efflux of the chemotherapeutic agent from the cancer cell in the presence of the PDE5 inhibitor indicates that the PDE5 inhibitor is capable of blocking multidrug resistance. In addition to measuring (1) growth of the cancer cell, (2) accumulation of the chemotherapeutic agent in the cancer cell, or (3) efflux of the chemotherapeutic agent from the cancer cell, a skilled artisan will know of other assays that may be used to determine whether a PDE5 inhibitor has a beneficial effect on multidrug resistance.
  • As used herein, the step of contacting the cell, e.g., the cancer cell, is carried out in vivo, in vitro, or ex vivo. In one embodiment, the step of contacting a cell is carried out in a human or nonhuman vertebrate subject, e.g., a mammalian subject, e.g., a human subject.
  • The entire contents of all references, patent applications, and patents cited throughout this application are hereby incorporated by reference herein.
  • EXAMPLES
  • The invention will be further illustrated in the following nonlimiting examples. These Examples are set forth to aid in the understanding of the invention but are not intended to, and should not be construed to, limit its scope in any way. The Examples do not include detailed descriptions of conventional methods that are well known to those of ordinary skill in the art. Additionally, the present invention is further illustrated in Ding et al. (2011) PLoS ONE 6(4):e19329, and Shi et al. (2011) Cancer Research 71(8):1-13, incorporated herein by reference in their entireties.
  • Example 1 Effects of Vardenafil and Tadalafil on ABCB1/P-Glycoprotein-Mediated Multidrug Resistance Example 1.1 Materials and Methods Example 1.1.a Reagents
  • Vardenafil and tadalafil were purchased from Toronto Research Chemicals Inc. (Ontario, Canada). [3H]-paclitaxel (37.9 Ci/mmol) was purchased from Moravek Biochemicals Inc (Brea, Calif.). [125I]-Iodoarylazidoprazosin (IAAP) (2,200 Ci/mmol) was obtained from Perkin Elmer Life Sciences (Boston, Mass.). Monoclonal antibody C-219 (against ABCB1) was acquired from Signet Laboratories Inc. (Dedham, Mass.). Anti-glyceraldehyde-3-phosphate dehydrogenase (GAPDH) monoclonal antibody (14C10) was obtained from Cell Signaling Technology, Inc. (Danvers, Mass.). Fumitremorgin C (FTC) was synthesized by Thomas McCloud Developmental Therapeutics Program, Natural Products Extraction Laboratory, NCI, NIH (Bethesda, Md.). ONO1078 was a gift from Dr. Akiyama (Kagoshima University, Japan). Paclitaxel, vincristine (VCR), colchicine, 7-ethyl-10-hydroxy-20 (S)-camptothecin (SN-38), verapamil, dimethyl sulfoxide (DMSO), 1-(4,5-dimethylthiazol-2-yl)-3,5-diphenylformazan (MTT) and all other chemicals were purchased from Sigma Chemical Company.
  • Example 1.1.b Cell Lines and Cell Culture
  • The ABCB1/Pgp-overexpressing drug-resistant cell line KB-C2 was established in a cell culture medium by a step-wise selection of the parental human epidermoid carcinoma cell line KB-3-1 using colchicine at concentrations up to 2 μg/ml. The KB-C2 and KB-3-1 were kindly provided by Dr. Akiyama (Kagoshima University, Japan). HEK293-pcDNA3.1 and wild-type HEK/ABCG2 transfected cells were established by selection with G418 after transfecting HEK293 with either (1) empty pcDNA3.1 vector or (2) pcDNA3.1 vector containing full length of ABCG2 coding arginine (R) at amino acid position 482, and were then cultured in a medium with 2 mg/ml of G418 (ABCG2-482-R2 cells). Similarly, the HEK/MRP1 (ABCC1) and HEK/ABCB1 cells were generated by transfecting the HEK293 cells with either the MRP1 expression vector or the ABCB1 expression vector. All of the cell lines were grown in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% bovine serum, 100 units/ml penicillin, and 100 mg/ml streptomycin in a humidified incubator containing 5% CO2 at 37° C.
  • Example 1.1.c Cell Cytotoxicity by MTT Assay
  • The MTT assay was used to assess cytotoxicity. The cultured cells were harvested with trypsin and resuspended in a final concentration of 4×103 cells/well for KB-3-1; 7.5×103 cells/well for KB-C2; and 8×103 for all of the other cell lines used in this study. Cells were seeded evenly in 96-well multiplates. In the reversal experiments, different concentrations of chemotherapeutic drugs (20 μl/well) were added into designated wells after 1 h with or without exposure to potential reversal compounds: vardenafil, tadalafil, verapamil, ONO-1078, or FTC (20 μl/well). After 68 h of incubation, 20 μl of the MTT solution (4 mg/ml) was added to each well, and the plate was further incubated for 4 h at 37° C., allowing viable cells to convert the yellow-colored MTT into dark-blue formazan crystals. Subsequently, the medium was discarded, and 100 μl of DMSO was added into each well to dissolve the formazan crystals, generating purple color. The absorbance was determined at 570 nm by an OPSYS Microplate Reader from DYNEX Technologies, Inc. (Chantilly, Va.). The degree of resistance was calculated by dividing the IC50 (concentrations required to inhibit growth, i.e., proliferation, by 50%) of a chemotherapeutic agent in the resistant cells by the IC50 of the chemotherapeutic agent in the parental sensitive cells. The degree of the reversal of multidrug resistance (MDR) was calculated by dividing the IC50 of an anticancer drug, e.g., an antineoplastic or chemotherapeutic drug (agent, compound), in a cell type in the absence of a reversal agent by the IC50 of the anticancer drug in the same cell type in the presence of the reversal agent. The IC50 values were calculated from survival curves using the Bliss method.
  • Example 1.1.d [3H]-Paclitaxel Accumulation and Efflux
  • The intracellular accumulation of [3H]-paclitaxel was measured as follows. Confluent cells in 24-well plates were preincubated with or without the reversal agents, e.g., vardenafil, tadalafil, or verapamil, for 1 h at 37° C. Intracellular paclitaxel accumulation was measured by incubating cells with 0.1 μM [3H]-paclitaxel for 2 h in the presence or absence of the reversal agents at 37° C. The cells were washed three times with ice-cold PBS, then suspended in fresh medium with or without reversal agents at 37° C. Aliquots of the extracellular medium (40 μl) were collected at various time points (0, 60, 120 min), and finally the cells were collected and lysed in 10 mM lysis buffer (pH 7.4, containing 1% Triton X-100 and 0.2% SDS). Each sample was placed in scintillation fluid and the radioactivity was measured in a Packard TRI-CARB1 1900CA liquid scintillation analyzer from Packard Instrument Company, Inc. (Downers Grove, Ill.).
  • Example 1.1.e Western Blot and Immunofluorescence Analysis
  • To determine the effect of vardenafil or tadalafil on the expression of ABCB1, KB-C2 cells were incubated with 10 μM vardenafil or tadalafil for 0, 36 and 72 h. Following incubation, the cells were harvested and rinsed twice with ice-cold PBS, and total cell lysates were collected with cell lysis buffer (1×PBS, 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS, 100 μg/ml phenylmethylsulfonyl fluoride, 10 μg/ml aprotinin, 10 μg/ml leupeptin) for 30 min with gentle rocking, and clarified by centrifugation at 12,000 rpm for 10 min at 4° C. Equal amounts (100 μg of protein) of cell lysates were resolved by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and electrophoretically transferred onto polyvinylidene fluoride (PVDF) membranes. After incubation in a blocking solution containing 5% nonfat milk in TBST buffer (10 mM Tris-HCL (pH 8.0), 150 mM NaCl, and 0.1% Tween 20) for 1 h at room temperature, membranes were immunoblotted overnight with primary monoclonal antibodies C219 (1:200) against ABCB1, or 14C10 (1:200) against GAPDH, at 4° C. Subsequently, the membranes were washed three times for 15 min with TBST buffer and incubated at room temperature for 2 h with HRP-conjugated secondary antibody at 1:1000 dilutions. The membranes underwent three additional washes for 15 min with TBST buffer and the protein-antibody complexes were visualized by the enhanced PHOTOTOPE® HRP Detection Kit (Cell Signaling Technology, Inc.) and exposed to Kodak medical X-ray processor (Eastman Kodak Co., Rochester, N.Y.). For the immunofluorescence analysis, cells (2×103) were seeded in 24-well plates and, after 12 h of incubation at 37° C. in a humidified atmosphere of 5% CO2, vardenafil or tadalafil at 10 mM were added into the wells. After incubation for 72 h with vardenafil or tadalafil, cells were washed with PBS and fixed with 4% paraformaldehyde for 15 min at room temperature and then rinsed with PBS three times. The monoclonal antibody C219 against ABCB1 (1:500) (Signet Laboratories Inc., Dedham, Mass.) was added for overnight incubation, followed by incubation for 1 h in Alexa Fluor 488 goat antimouse IgG (1:1000; Molecular Probe, Carlsbad, Calif.). Propidium iodide was used for nuclear staining.
  • Example 1.1.f ATPase Assay of ABCB1
  • The Vi-sensitive ATPase activity of ABCB1 in membrane vesicles of High Five™ insect cells was measured as follows. The membrane vesicles (10 μg of protein) were incubated in ATPase assay buffer (50 mM MES [pH 6.8], 50 mM KCl, 5 mM sodium azide, 2 mM EGTA, 2 mM dithiothreitol, 1 mM ouabain, and 10 mM MgCl2) with or without 0.3 mM orthovanadate (freshly prepared) at 37° C. for 5 min, then incubated with different concentrations of drug at 37° C. for 3 min. The ATPase reaction was started by the addition of 5 mM ATP, and the total volume was 0.1 ml. After incubation at 37° C. for 20 min, the reactions were stopped by the addition of 0.1 ml of 5% SDS solution and vortexed and kept at room temperature. The liberated Pi was measured as described in Ambudkar et al., supra.
  • Example 1.1.g Photoaffinity Labeling of ABCB1 with [125I]-IAAP
  • The photoaffinity labeling of ABCB1 with [125I]-IAAP was performed as described in Sauna et al. (2000) Proc. Natl. Acad. Sci. USA 97:2515-20. The membrane vesicles from High Five™ insect cells expressing ABCB1 (50 μg of protein) were incubated at room temperature with different concentrations of drugs in the ATPase assay buffer with [125I]-IAAP (7 nM) for 5 min under subdued light. The samples were photo cross-linked by using a 365 nm UV light source for 10 min at room temperature. Subsequently, the samples were run in a 7% Tris-acetate NuPAGE gel on SDS-PAGE; and the gels were dried and exposed to Bio-Max MR film (Eastman Kodak Co., Rochester, N.Y.) at −70° C. for 8-12 h. The radioactivity incorporated into the ABCB1 band was quantified using the STORM 860 PhosphorImager system and ImageQuaNT (Molecular Dynamics, CA).
  • Example 1.1.h Ligand-ABCB1 Structure Preparation
  • Vardenafil, tadalafil (modeled as R,R isomer), and IAAP were constructed using the fragment dictionary of Maestro 9.0 and the energy minimized by Macromodel program v9.7 (Schrödinger, Inc., New York, N.Y.) using the OPLSAA force field (as described Jorgensen et al. (1996) J. Am. Chem. Soc. 118:11225-36), with the steepest descent followed by a truncated Newton conjugate gradient protocol. Partial atomic charges were computed using the OPLS-AA force field. The low-energy 3D structures of vardenafil, tadalafil, and IAAP were generated with the following parameters present in LigPrep v2.3: different protonation states at physiological pH, all possible tautomers and ring conformations.
  • Example 1.1.i Protein Structure Preparation
  • The X-ray crystal structure of mouse ABCB1 in the apoprotein state (PDB ID: 3G5U) and in complex with inhibitors QZ59-RRR (PDB ID: 3G6O) and QZ59-SSS (PDB ID: 3G61) was obtained from the RCSB Protein Data Bank; these were used to build the homology model of human ABCB1. The homology modeling was conducted using the default parameters of Prime v2.1 as implemented in Maestro 9.0. The input file for the amino acid sequence of human ABCB1 in the Prime structure prediction application was obtained as a FASTA file (UniProt Accession Number P08183.3) extracted from uniprot.org. The cocrystal structures of ABCB1 from the mouse model in complex with QZ59-RRR and QZ59-SSS inhibitors were used as templates for modeling site-1 and site-2, respectively; while apoprotein-ABCB1 was used as a template for modeling site-3 and site-4. The resultant alignment of human ABCB1 and mouse ABCB1 sequences produced 87% sequence identity and 93% similarity. Based on the resultant alignment that was constructed using default parameters, the side chains were optimized and residues were minimized. The initial structure thus obtained was refined by means of default parameters mentioned in a protein preparation facility implemented in Maestro v9.0 and Impact program v5.5 (Schrödinger, Inc., New York, N.Y.), in which the protonation states of residues were adjusted to the dominant ionic forms at pH 7.4. The refined human ABCB1 homology model was further used to generate four different receptor grids by selecting QZ59-RRR (site-1) and QZ59-SSS (site-2) bound ligands, all amino acid residues known to contribute to verapamil binding (site-3), and two residues known to be common to the three previous sites (site-4), as shown in Table 7.
  • Example 1.1.j Docking Protocol
  • The docking calculations were performed using the “Extra Precision” (XP) mode of Glide program v5.5 (Schrödinger, Inc.) and the default parameters. The top scoring pose-ABCB1 complex was then subjected to energy minimization using Macromodel program v9.7 using the OPLS-AA force field and used for graphical analysis. All computations were carried out on a Dell Precision 470n dual processor with the Linux OS (Red Hat Enterprise WS 4.0).
  • Example 1.1.k Statistical Analysis
  • All experiments were repeated at least three times and the differences were determined by using the Student's t-test. The statistical significance was determined at p<0.05.
  • Example 1.2 Vardenafil Enhances Drug Sensitivity of ABCB1- but Not ABCC1- or ABCG2-Overexpressing Cancer Cells
  • Cytotoxic effects of vardenafil and tadalafil on different cell lines using the MTT assay were tested, and neither vardenafil nor tadalafil inhibited the growth of any cell lines tested at concentrations of up to 20 μM.
  • Subsequently, the effect of vardenafil or tadalafil on the sensitivity of anticancer drugs in ABCB1-, ABCC1-, and ABCG2-overexpressing MDR cells was tested. At 5 and 10 μM, vardenafil produced a concentration-dependent increase in cytotoxicity of colchicine and paclitaxel in ABCB1-overexpressing drug-selected cell line KB-C2, but not parental sensitive KB-C2 cells (Table 4). In order to eliminate the possibility of multiple factors playing a role in drug-selected cell lines, the effect of vincristine and paclitaxel cytotoxicity on ABCB1-transfected HEK293/ABCB1 cells was also measured. Vardenafil increased the sensitivity of ABCB1-transfected HEK/ABCB1 cells to vincristine and paclitaxel (Table 5), but not the control cell line HEK293/pcDNA3.1. Therefore, the effect of vardenafil was specific to ABCB1-overexpressing cells, but had no significant toxic effects on parental cells when combined with ABCB1 transporter substrate anticancer drugs. Vardenafil did not reverse MDR induced by cells expressing ABCC1 and ABCG2 (Table 6), or significantly alter the IC50 values of cisplatin, which is not a substrate of ABCB1 (Tables 4 and 5).
  • TABLE 4
    The Effects of Vardenafil and Tadalafil on the Reversal
    of ABCB1-Mediated Resistance to Colchicine, Paclitaxel,
    and Cisplatin in Drug-Selected Cell Line
    IC50 ± SD (μM) (fold reversal)
    Compounds KB-3-1 KB-C2 (ABCB1)
    Colchicine 0.0063 ± 0.0013 (1.00)  2.9017 ± 0.6127 (1.00)
    + Vardenafil 0.0070 ± 0.0014 (0.90) 0.1053 ± 0.0215** (27.6)
    5 μM
    + Vardenafil 0.0068 ± 0.0012 (0.93)  0.0157 ± 0.0063** (184.8)
    10 μM
    + Tadalafil 0.0073 ± 0.0009 (0.86) 0.6923 ± 0.1518** (4.19)
    5 μM
    + Tadalafil 0.0071 ± 0.0015 (0.89) 0.4547 ± 0.1033** (6.38)
    10 μM
    + Verapamil 0.0064 ± 0.0011 (0.98) 0.0347 ± 0.0071** (83.6)
    10 μM
    Paclitaxel 0.0066 ± 0.0016 (1.00)  0.7354 ± 0.0141 (1.00)
    + Vardenafil 0.0074 ± 0.0015 (0.89) 0.0281 ± 0.0067** (26.2)
    5 μM
    + Vardenafil 0.0072 ± 0.0017 (0.92) 0.0136 ± 0.0025** (54.1)
    10 μM
    + Tadalafil 0.0071 ± 0.0020 (0.93) 0.2911 ± 0.0669** (2.53)
    5 μM
    + Tadalafil 0.0065 ± 0.0028 (1.02) 0.1641 ± 0.0311** (4.48)
    10 μM
    + Verapamil 0.0060 ± 0.0009 (1.10) 0.0132 ± 0.0035** (55.7)
    10 μM
    Cisplatin 1.9032 ± 0.0709 (1.00)  1.7877 ± 0.2171 (1.00)
    + Vardenafil 2.1316 ± 0.3653 (0.89)  2.0555 ± 0.7811 (0.87)
    5 μM
    + Vardenafil 2.0364 ± 0.6313 (0.93)  1.8651 ± 0.5409 (0.96)
    10 μM
    + Tadalafil 1.9899 ± 0.4975 (0.96)  1.7700 ± 0.7257 (1.01)
    5 μM
    + Tadalafil 1.7890 ± 0.6083 (1.06)  1.9221 ± 0.9034 (0.93)
    10 μM
    + Verapamil 1.8703 ± 0.2330 (1.02)  1.7890 ± 0.6472 (1.00)
    10 μM
    Cell survival was determined by MTT assay as described in Example 1.1.c. Data are expressed as the mean ± SD of at least three independent experiments performed in triplicate. The fold-reversal values of MDR (values given in parentheses) were calculated by dividing the IC50 values of anticancer drugs in cells in the absence of an inhibitor by those obtained in the same cell type in the presence of the inhibitor.
    **represents p < 0.01, for values versus those obtained in the absence of inhibitor.
  • TABLE 5
    The Effects of Vardenafil and Tadalafil on the Reversal
    of ABCB1-Mediated Resistance to Vincristine, Paclitaxel,
    and Cisplatin in ABCBl-Transfected Cell Line
    IC50 ± SD (μM) (fold reversal)
    Compounds HEK293/pcDNA3.1 HEK/ABCB1 (ABCB1)
    Vincristine 11.55 ± 1.59 (1.00) 169.84 ± 12.93 (1.00)
    + Vardenafil 5 μM 13.13 ± 0.93 (0.88) 41.41 ± 6.12** (4.10)
    + Vardenafil 10 μM  9.72 ± 2.13 (1.19)  14.64 ± 2.36** (11.60)
    + Tadalafil 5 μM 10.02 ± 1.24 (1.15)  112.94 ± 5.04 (1.50)
    + Tadalafil 10 μM  8.57 ± 0.62 (1.35)  69.27 ± 3.48* (2.45)
    + Verapamil 10 μM  8.23 ± 1.11 (1.40) 18.21 ± 4.82** (9.33)
    Paclitaxel 23.73 ± 5.21 (1.00) 219.14 ± 13.16 (1.00)
    + Vardenafil 5 μM 21.85 ± 2.04 (1.09) 56.83 ± 7.74** (3.86)
    + Vardenafil 10 μM 20.09 ± 3.06 (1.18) 25.23 ± 4.17** (8.69)
    + Tadalafil 5 μM 24.04 ± 2.95 (0.99) 159.78 ± 11.52 (1.37)
    + Tadalafil 10 μM 19.64 ± 3.17 (1.21)  96.92 ± 4.83* (2.26)
    + Verapamil 10 μM 20.17 ± 2.52 (1.18) 31.03 ± 2.19** (7.06)
    Cisplatin 890.32 ± 33.92 (1.00)  850.84 ± 82.53 (1.00)
    + Vardenafil 5 μM 839.03 ± 48.37 (1.06)  893.19 ± 32.60 (0.95)
    + Vardenafil 10 μM 892.44 ± 19.26 (1.00)  782.82 ± 59.31 (1.09)
    + Tadalafil 5 μM 901.29 ± 67.23 (0.99)  909.06 ± 98.44 (0.94)
    + Tadalafil 10 μM 792.85 ± 92.60 (1.12)  783.60 ± 84.20 (1.09)
    + Verapamil 10 μM 853.62 ± 61.04 (1.04)  725.71 ± 48.56 (1.17)
    Cell survival was determined by MTT assay as described in Example 1.1.c. Data are expressed as the mean ± SD of at least three independent experiments performed in triplicate. The fold-reversal values of MDR (values given in parentheses) were calculated by dividing the IC50 values of anticancer drugs in cells in the absence of an inhibitor by those obtained in the same cell type in the presence of the inhibitor.
    **represents p < 0.01,
    *represents p < 0.05, for values versus those obtained in the absence of inhibitor.
  • TABLE 6
    The Effect of Vardenafil and Tadalafil on the
    Reversal of ABCG2-Mediated Resistance to SN-38
    and ABCC1-Mediated Resistance to Vincristine
    Compounds IC50 ± SD (μM) (fold reversal)
    HEK293/pcDNA3.1     ABCG2-482-R2 (ABCG2)
    SN-38 0.1157 ± 0.0151 (1.00) 3.6471 ± 0.5472 (1.00)
    + Vardenafil 0.1049 ± 0.0186 (1.10) 3.7652 ± 0.6264 (0.97)
    10 μM
    + Tadalafil 0.1202 ± 0.0250 (0.96) 3.5173 ± 0.8198 (1.04)
    10 μM
    + FTC 0.1021 ± 0.0238 (1.13) 0.9484 ± 0.1634** (3.84) 
    10 μM
    HEK293/pcDNA3.1    HEK/MRP1 (ABCC1)
    Vincristine 0.0012 ± 0.0002 (1.00) 0.0106 ± 0.0022 (1.00)
    + Vardenafil 0.0013 ± 0.0001 (0.92) 0.0099 ± 0.0016 (1.07)
    10 μM
    + Tadalafil 0.0011 ± 0.0002 (1.09) 0.0103 ± 0.0021 (1.03)
    10 μM
    + ONO-1078 0.0009 ± 0.0001 (1.33) 0.0018 ± 0.0003** (5.9)  
    10 μM
    Cell survival was determined by MTT assay as described in Example 1.1.c. Data are expressed as the mean ± SD of at least three independent experiments performed in triplicate. The fold-reversal values of MDR (values given in parentheses) were calculated by dividing the IC50 values of anticancer drugs in cells in the absence of an inhibitor by those obtained in the same cell type in the presence of the inhibitor.
    **represents p < 0.01, for values versus those obtained in the absence of inhibitor.
  • Example 1.3 Vardenafil Increases Accumulation of Intracellular Paclitaxel in ABCB1-Overexpressing Cells by Inhibiting Drug Efflux
  • To determine the reversal mechanism of vardenafil and tadalafil for ABCB1 transporter, the accumulation of [3H]-paclitaxel was measured after cells were preincubated with or without vardenafil, tadalafil, or verapamil (control) for 1 h at 37° C. and then incubated with [3H]-paclitaxel for another 2 h at 37° C. (FIG. 1A). The intracellular concentration of paclitaxel in KB-C2 cells was approximately 55% of that in the parental KB-3-1 cells. However, 10 μM of vardenafil significantly increased the intracellular accumulation of paclitaxel in KB-C2 by 1.6-fold without altering the levels accumulated in KB-3-1 cells. Although tadalafil at 10 μM had significant effects on paclitaxel accumulation, this accumulation was reduced in comparison with vardenafil and verapamil (control).
  • The effect of vardenafil on paclitaxel efflux was also determined (FIG. 1B). The intracellular levels of paclitaxel were measured over 2 h. A significantly higher concentration of paclitaxel was effluxed from the KB-C2 cells compared to KB-3-1 cells, and the amount of effluxed paclitaxel increased with time; at 1 h time point, 70% of accumulated paclitaxel was effluxed from KB-C2 cells in the absence of vardenafil, and 10 μM of vardenafil significantly blocked ABCB1 efflux function, with 75% of accumulated paclitaxel being retained inside KB-C2 cells. No significant change in concentration of effluxed paclitaxel in parental KB-3-1 cells in the absence or presence of vardenafil was noted.
  • Example 1.4 Vardenafil does not Alter Membrane Expression of ABCB1
  • To determine whether reversal of ABCB1-mediated MDR occurred by either decreasing ABCB1 expression or inhibiting ABCB1 activity, the ABCB1-overexpressing KB-C2 cells were incubated with 10 μM vardenafil or tadalafil for 36 and 72 h and then assayed to determine the difference in protein levels via Western blot. According to FIG. 1C, the protein level of ABCB1 in KB-C2 cells was not significantly altered after the cells were incubated with vardenafil or tadalafil. It was possible that the ABCB1 transporter on the membrane had translocated inside the cell as a result of treatment, and the Western blot of whole lysates could not distinguish this possibility. Therefore, an immunofluorescence assay was performed to detect the location of the ABCB1 transporter. The results suggest that neither vardenafil nor tadalafil altered the expression of ABCB1 in the membrane of the KB-C2 cells after 72 h of incubation (not shown). Thus, vardenafil and tadalafil altered neither the expression nor the localization of ABCB1 transporter. These data are in agreement with the conclusion that PDE5 inhibitors inhibit ABCB1 function rather than its expression.
  • Example 1.5 The Effect of Vardenafil and Tadalafil on the ATPase Activity of ABCB1
  • The drug-efflux function of ABCB1 is coupled to ATP hydrolysis by the ATPase enzyme that is usually stimulated in the presence of ABCB1 substrates. Therefore, to assess the effect of vardenafil and tadalafil on the ATPase activity of ABCB1, the rate of ABCB1-mediated ATP hydrolysis was measured in isolated membrane vesicles in the presence of various concentrations of vardenafil and tadalafil under conditions that suppressed activity of other major ATPases. Vardenafil produced a concentration-dependent increase in the ATPase activity of ABCB1 over a range of concentrations (FIG. 2A), with the concentration of vardenafil required for a 50% stimulation of ATPase activity being 2.69±0.72 μM. In contrast, tadalafil mildly stimulated ATPase activity, such that at the highest concentration tested (25 μM), the concentration required for 50% stimulation was not reached.
  • Example 1.6 Effect of Vardenafil and Tadalafil on the Photoaffinity Labeling of ABCB1 with [125I]-IAAP
  • In order to determine whether vardenafil and tadalafil interacted with the substrate binding site of ABCB1, the effects of vardenafil and tadalafil on photoaffinity labeling of ABCB1 transporter with [125I]-IAAP using membrane vesicles was measured. Vardenafil inhibited the photoaffinity labeling of ABCB1 with [125I]-IAAP in a concentration-dependent manner (FIG. 2B). At concentrations of 0.69 μM and 10 μM, vardenafil inhibited the [125I]-IAAP photolabeling of ABCB1 by 50% and 90%, respectively, whereas concentrations of up to 50 μM tadalafil did not produce a 50% inhibition of the [125I]-IAAP photolabeling of ABCB1 transporter. These data demonstrate that the tested PDE5 inhibitors interacted with the drug binding site of ABCB1 and, as these agents stimulated the ATPase activity of ABCB1 (Example 1.5), they may be transport substrates for ABCB1.
  • Example 1.7 Model for Binding of Vardenafil and Tadalafil to ABCB1
  • In the Examples above, the PDE5 inhibitors vardenafil and tadalafil are described for the first time as ABCB1 inhibitors. Their predicted binding conformation within the large cavity of ABCB1 required determination. Because the crystal structure of the human ABCB1 remains to be elucidated and the binding conformation of vardenafil and tadalafil within the large cavity of ABCB1 transporter is unknown, a homology model of human ABCB1, based on the mouse ABCB1-QZ59-RRR cocrystal structure as a template (FIG. 3A), was utilized for the Glide docking study of vardenafil and tadalafil. Four binding sites were reported in the crystal structure of mouse ABCB1 as represented by the following sites: ABCB1-QZ59-RRR (site-1), ABCB1-QZ59-SSS (site-2), ABCB1-verapamil (site-3), and the site common to the above three sites (site-4). Aller et al. (2009) Science 323:1718-22. As the photoaffinity labeling data suggested that vardenafil displaces IAAP in a concentration-dependent manner, IAAP was also docked to these sites for comparison. These data also indicated that vardenafil and IAAP share the same binding site, i.e., site-1; however, the tadalafil binding site is somewhat different from the vardenafil docking site—the Glide docking score predicts that it is site-4 (Table 7). A comparison of the binding energy data for the docked poses of vardenafil, tadalafil, and IAAP at each of the binding sites (Table 7) suggested that the more potent of the two ABCB1 inhibitors, vardenafil, exhibited the most favorable binding energy within the QZ59-RRR binding site of ABCB1, whereas tadalafil interacted most favorably with site-4. Thus, the following section addresses the bound conformation of vardenafil and tadalafil in site-1 and in site-4, respectively.
  • TABLE 7
    Binding Energies of Vardenafil, Tadalafil, and IAAP within Each
    of the Predicted Binding Sites of ABCB1.
    Glide score kcal/mol
    Ligands Site-1a Site-2b Site-3c Site-4d
    Figure US20140288078A1-20140925-C00001
    −8.56 −6.26 −5.13 −4.87
    Figure US20140288078A1-20140925-C00002
    −6.26 −5.05 −7.35 −7.85
    Figure US20140288078A1-20140925-C00003
    −8.89 −5.79 −4.54 −5.16
    a-Site represented by bound QZ59-RRR.
    b-Site represented by bound ligand QZ59-SSS.
    c-Verapamil binding site.
    d-Site grid generated using residues Phe728 and Val982, which are known to be common to above three sites.
  • The XP-Glide-predicted binding mode of vardenafil indicates the importance of hydrophobic and electrostatic interactions within the large drug binding cavity of ABCB1 (FIG. 3B, left panel). Whereas the N-ethylpiperazine (D-ring) of vardenafil forms hydrophobic contacts with Met69, Phe336, Leu339, and Ile340, the C-ring along with its ethoxy substituent enters into favorable hydrophobic interactions with Phe72, Leu975, and Phe978. The A-ring, along with its methyl and propyl substituents and the B-ring, are engaged in hydrophobic interactions with the side chains of Phe728, Ala729, Phe732, and Val982. In addition, vardenafil also appears to form favorable electrostatic interactions with residues Tyr953 and Tyr307. For example, the sulfonyl oxygen atom forms a hydrogen bond with the hydroxyl group of Tyr953 (—SO2—HO-Tyr953), whereas the carbonyl function of the B-ring is located at a distance of 4.0 Å from the side chain hydroxyl group of Tyr307.
  • The XP-Glide-predicted binding mode of tadalafil in site-4 of the large drug binding cavity of ABCB1 is shown in FIG. 3B (right panel). The A- and B-rings of the indole moiety bind to the hydrophobic pocket formed by the side chains of Phe303, Leu304, Tyr307, and Phe343. Moreover, Phe343 also has hydrophobic contacts with the B-, C-, and E-rings. Both E- and F-rings of the benzodioxole moiety are surrounded by the side chains of Phe336, Leu339, Phe978, and Val982. The carbonyl oxygen atom (close to E-ring) of the D-ring is stabilized by a hydrogen bonding interaction with the side chain amide group of Gln725 (—CO—H2NOC-Gln725). The lower efficacy of tadalafil compared to vardenafil may be due to the orientation of its hydrophobic N-methyl substituent of the D-ring towards the unfavorable polar backbone of Met986 and Gly989 and the polar side chain amide group of Gln990.
  • Example 2 Effect of Sildenafil on ABCB1- and ABCG2-Mediated Multidrug Resistance Example 2.1 Materials and Methods Example 2.1.a Materials
  • [3H]-paclitaxel (37.9 Ci/mmol), [3H]-mitoxantrone (4 Ci/mmol), and [3H]-methotrexate (23 Ci/mmol) were purchased from Moravek Biochemicals, Inc. [3H]-E217βG (40.5 Ci/mmol) and [125I]-IAAP (2,200 Ci/mmol) were obtained from PerkinElmer Life Sciences. The fluorescent compound BODIPY-prazosin was purchased from Invitrogen. Monoclonal antibodies C-219 (against ABCB1) and BXP-21 (against ABCG2) were acquired from Signet Laboratories, Inc. Sildenafil was purified from 100 mg VIAGRA® tablets as described by Francis et al. (2003) J. Impot. Res. 15:369-72. Fumitremorgin C (FTC) was synthesized by Thomas McCloud Developmental Therapeutics Program, Natural Products Extraction Laboratory, National Cancer Institute (NCI), NIH. Other chemicals were purchased from Sigma Chemical Co.
  • Example 2.1.b Cell Lines and Cell Culture
  • The ABCB1-overexpressing drug-resistant cell line KB-C2 was established by stepwise selection of the parental human epidermoid carcinoma cell line KB-3-1 in increasing concentrations of colchicine and was cultured in medium containing 2 μg/mL of colchicine. The ABCB1-overexpressing drug resistant cell line KB-V1 (generously provided by Dr. Gottesman, NCI, NIH) was established by a stepwise concentration increase of vinblastine in the culture medium of KB-3-1 cells and subsequently culturing these cells in medium with 1 μg/mL of vinblastine. An ABCC1-overexpressing MDR cell line, KB-CV60, was also derived from KB-3-1 cells and was maintained in medium with 1 μg/mL of cepharanthine and 60 ng/mL of vincristine. Both KB-C2 and KB-CV60 cell lines were kindly provided by Dr. Akiyama (Kagoshima University, Japan). HEK293/pcDNA3.1, ABCG2-482-R5, ABCG2-482-G2, and ABCG2-482-T7 cells were established by selection with 2 mg/mL G418 after transfecting HEK293 with either (1) empty pcDNA3.1 vector or (2) pcDNA3.1 vector containing full length ABCG2 coding either (2a) wild-type arginine (R) or mutant (2b) glycine (G) or (2c) threonine (T) at amino acid position 482, respectively. The wild-type ABCG2-overexpressing drug-resistant cell line MCF-7/Flv1000 was cultured in the medium with 1 μM of flavopiridol. The mutated ABCG2-overexpressing drug resistant cell line MCF-7/ADVP3000 was maintained in the medium with 5 μg/mL of verapamil and 3 μg/mL of doxorubicin. Another G482 mutant ABCG2-overexpressing drug resistant cell line, S1-M1-80, was maintained in the medium with 80 μM of mitoxantrone. The drug-resistant cell line S1/Flv5000, which does not express ABCG2, was also generated from S1 by increasing the amount of flavopiridol and was maintained in the medium with 5 μM of flavopiridol. All the cell lines were grown as adherent monolayers in flasks with DMEM (Dulbecco's modified Eagle's medium) culture medium (Hyclone Co.) containing 10% bovine serum at 37° C. in a humidified atmosphere of 5% CO2.
  • Example 2.1.c MTT Cytotoxicity Data
  • Cells in 96-well plates were preincubated with or without the reversal agents for 1 h and then different concentrations of chemotherapeutic drugs were added into designated wells. After 68 h of incubation, MTT solution (4 mg/mL) was added to each well, and the plate was further incubated for 4 h, allowing viable cells to change the yellow-colored MTT into dark-blue formazan crystals. Subsequently the medium was discarded, and 100 mL of DMSO was added into each well to dissolve the formazan crystals. The absorbance was determined at 570 nm by an OPSYS MR Microplate Reader from DYNEX Technologies, Inc.
  • Example 2.1.d Paclitaxel and Mitoxantrone Accumulation
  • Cells in 24-well plates were preincubated with or without the reversal agents for 1 h at 37° C., then incubated with 0.1 μM [3H]-paclitaxel or 0.2 μM [3H]-mitoxantrone for 2 h in the presence or absence of the reversal agents at 37° C. After washing 3 times with ice-cold PBS, the cells were trypsinized and lysed in 10 mM lysis buffer (pH 7.4, containing 1% Triton X-100 and 0.2% SDS). Each sample was placed in scintillation fluid and radioactivity was measured in a Packard TRI-CARB 1900CA liquid scintillation analyzer from Packard Instrument Company, Inc.
  • Example 2.1.e Flow Cytometric Assays
  • Flow cytometric assays were carried out as follows. The cells were trypsinized and then resuspended in complete media (phenol red-free Iscove's modified Eagle's medium, IMEM, with 10% fetal calf serum) containing 250 nM BODIPY-prazosin alone or with various concentrations of the inhibitors for 30 min at 37° C. Cells were then washed once in cold complete medium and then incubated for another 1 h at 37° C. in substrate-free media with or without the same described concentrations of the inhibitors to generate the efflux histograms. Subsequently, cells were washed twice with cold DPBS (Dulbecco's PBS) and placed on ice in a dark environment until ready for analysis. Cells were analyzed on a FACSort flow cytometer equipped with a 488-nm argon laser. For all samples, at least 10,000 events were collected. Cell debris was eliminated by gating on forward versus side scatter, and dead cells were excluded on the basis of propidium iodide staining.
  • Example 2.1.f In Vitro Transport Assays
  • Transport assays were carried out using the rapid filtration method as described in Chen et al. (2003) Cancer Res. 63:4048-54. Membrane vesicles were incubated with various concentrations of inhibitors for 1 h on ice, and then transport reactions were carried out at 37° C. for 10 min in a total volume of 50 μL of medium (membrane vesicles 10 μg, 0.25 M sucrose, 10 mM Tris-HCl, pH 7.4, 10 mM MgCl2, 4 mM ATP or AMP, 10 mM phosphocreatine, 100 μg/mL creatine phosphokinase, and 0.25 μM [3H]-E217βG, or 0.5 μM [3H]-methotrexate). Reactions were stopped by the addition of 3 mL of ice-cold stop solution (0.25 M sucrose, 100 mM NaCl, and 10 mM Tris-HCl, pH 7.4). During the rapid filtration step, samples were passed through 0.22 μm GVWP filters (Millipore) presoaked in the stop solution. The filters were washed three times with 3 mL of ice-cold stop solution. Radioactivity was measured by liquid scintillation counting.
  • Example 2.1.g ATPase Assay of ABCB1 and ABCG2
  • The vanadate (Vi)-sensitive ATPase activity of ABCB1 or ABCG2 in the membrane vesicles of HIGH FIVE™ insect cells was measured as described in Ambudkar et al., supra. The membrane vesicles (100 μg of protein/mL) were incubated in ATPase assay buffer (50 mM MES, pH 6.8, 50 mM KCl, 5 mM sodium azide, 2 mM EGTA, 2 mM dithiothreitol, 1 mM ouabain, and 10 mM MgCl2) with or without 0.3 mM vanadate at 37° C. for 5 min, and then incubated with different concentrations of drugs at 37° C. for 3 min. The ATPase reaction was incubated by the addition of 5 mM Mg-ATP. After incubating at 37° C. for 20 min, the reactions were stopped by adding 0.1 mL of 5% SDS solution. The liberated inorganic phosphate was measured as described in Shukla et al. (2006) Biochemistry 45:8940-51. In the inhibition assays, the decrease in maximum Vi-sensitive ABCB1 or ABCG2 activity by sildenafil was measured in the presence of verapamil at 50 μM or FTC at 10 μM, respectively.
  • Example 2.1.h Photoaffinity Labeling of ABCB1 and ABCG2 with [125I]-IAAP
  • The photoaffinity labeling of ABCB1 and ABCG2 with [125I]-IAAP was conducted as described in Sauna et al., supra. The crude membranes from HIGH FIVE™ insect cells expressing ABCB1 and MCF7/FLV1000 cells expressing R482 ABCG2 (50 μg of protein) were incubated at room temperature with different concentrations of drugs in 50 mM Tris-HCl (pH 7.5) with [125I]-IAAP (5-7 nM) for 5 min under subdued light. The samples were photo-cross-linked by exposure to a 365 nm UV light for 10 min at room temperature. ABCB1 and ABCG2 were immunoprecipitated using BXP-21 antibodies as described in Shukla et al., supra. Both ABCB1 and ABCG2 samples were subjected to SDS-PAGE in a 7% Trisacetate NuPAGE gel, the gel was dried, and exposed to Bio-Max MR film (Eastman Kodak Co.) at −70° C. for 8 to 12 h. The radioactivity incorporated into the ABCB1 or ABCG2 band was quantified using the STORM 860 PhosphorImager System and ImageQuaNT (Molecular Dynamics).
  • Example 2.1.i Ligand Structure Preparation
  • Sildenafil and IAAP structures were built using the fragment dictionary of Maestro 9.0 and energy minimized by Macromodel program v9.7 (2009; Schrödinger, Inc.) using the OPLS-AA force field (Jorgensen et al., supra) with the steepest descent followed by a truncated Newton conjugate gradient protocol. Partial atomic charges were computed using the OPLS-AA force field. The low-energy 3D structures of sildenafil and IAAP were generated with the following parameters present in LigPrep v2.3: different protonation states at physiologic pH, all possible tautomers and ring conformations.
  • Example 2.1.j Protein Structure Preparation
  • The X-ray crystal structure of ABCB1 in apoprotein state (PDB ID: 3G5U) and in complex with inhibitors QZ59-RRR (PDB ID: 3G6O) and QZ59-SSS (PDB ID: 3G61) obtained from the RCSB Protein Data Bank were used to build the homology model of human ABCB1. Aller et al., supra. Homology modeling was carried out using the default parameters of Prime v2.1 as implemented in Maestro 9.0. The input file for the amino acid sequence of human ABCB1 in the Prime structure prediction application was obtained as a FASTA file (UniProt Accession number P08183.3) extracted from www.uniprot.org. The cocrystal structures of ABCB1 from the mouse model in complex with QZ59-RRR and QZ59-SSS inhibitors were used as templates for modeling site-1 and site-2, respectively; whereas apoprotein ABCB1 was used as a template for modeling site-3 and site-4. The resultant alignment of human ABCB1 and mouse ABCB1 sequences produced 87% sequence identity and 93% similarity. On the resultant alignment built using default parameters, side chains were optimized and residues were minimized. The initial structure thus obtained was refined by means of default parameters mentioned in protein preparation facility implemented in Maestro v9.0 and Impact program v5.5 (2009; Schrödinger, Inc.), in which the protonation states of residues were adjusted to the dominant ionic forms at pH 7.4. The refined human ABCB1 homology model was used further to generate four different receptor grids by selecting QZ59-RRR (site-1) and QZ59-SSS (site-2) bound ligands, all amino acid residues known to contribute to verapamil binding (site-3), and two residues known to be common to three previous sites (site-4), as shown in Table 11.
  • Example 2.1.k Docking Protocol
  • All docking calculations were done using the “Extra Precision” (XP) mode of Glide program v5.5 (2009; Schrödinger, Inc.) and the default parameters. The top-scoring pose-ABCB1 complex was then subjected to energy minimization using Macromodel program v9.7 utilizing the OPLS-AA force field (Jorgensen et al., supra), and used for graphical analysis. All computations were carried out on a Dell Precision 470n dual processor with the Linux OS (Red Hat Enterprise WS 4.0).
  • Example 2.1.l Statistical Analysis
  • All experiments were repeated at least three times and the differences were determined by the Student's t test. The statistical significance was determined at p<0.05.
  • Example 2.2 Sildenafil Sensitizes ABCB1- and ABCG2-Overexpressing Cells to Chemotherapeutic Drugs
  • First, the sensitivity of ABCB1-, ABCG2-, and ABCC1-overexpressing cells to sildenafil was assessed. The results of the MTT assay showed that sildenafil did not inhibit the growth of any of the cell lines used at concentrations of up to 50 μM (not shown). However, sildenafil increased the sensitivity of ABCB1-overexpressing drug-resistant cells to substrate drugs. The IC50 values of ABCB1 substrates colchicine, vinblastine, and paclitaxel were much higher in ABCB1-overexpressing cells KB-C2 and KB-V1 than parent KB-3-1 cells, but sildenafil decreased the IC50 values of these drugs in KB-C2 and KB-V1 cells (Table 8). At 2.5 μM, sildenafil moderately increased the sensitivity (in KB-C2 and KB-V1 cells) to all three drugs, and at 10 μM, sildenafil increased the sensitivity with efficacy comparable to that of the equivalent concentration of verapamil. Neither sildenafil nor verapamil altered the cytotoxicity of the three drugs in parental KB-3-1 cells. The IC50 values of cisplatin, which is not a substrate for ABCB1 and exhibited equal sensitivity in KB-3-1, KB-C2, and KB-V1 cells, were not affected by either sildenafil or verapamil in these three cell lines.
  • The effects of sildenafil on ABCC1- and ABCG2-mediated drug resistance were determined. In the ABCC1-overexpressing KB-CV60 cells, sildenafil at 10 μM showed no significant reduction in the IC50 value of vincristine, a known ABCC1 substrate (not shown).
  • It has been reported that mutations at amino acid 482 in ABCG2 alter the substrate and antagonist specificity of ABCG2 (Robey et al. (2003) Br. J. Cancer 89:1971-78; Honjo et al. (2001) Cancer Res. 61:6635-39); therefore, the reversing effects of sildenafil on both wild type (R482) and mutant (R482G and R482T) ABCG2-overexpressing cells were determined. Compared with parental MCF-7 cells, MCF-7/Flv1000 and MCF-7/AdVp3000 cells exhibited high levels of resistance to ABCG2 substrates flavopiridol, mitoxantrone, and SN-38, but not to non-ABCG2 substrate cisplatin (Table 9). Similarly, the IC50 values for flavopiridol, mitoxantrone, and SN-38 in S1-M1-80 were significantly higher than in parental S1 cells. At 50 μM, sildenafil decreased the IC50 values for flavopiridol, mitoxantrone, and SN-38 in the ABCG2-expressing cell lines down to levels observed when cytotoxicity assays were carried out in the presence of the known specific ABCG2 inhibitor FTC at 2.5 μM. However, the IC50 values of these ABCG2 substrate drugs were not significantly different in the parental cells or the S1/FLV5000 cells (which do not express ABCG2, but also are resistant to flavopiridol in the presence or absence of sildenafil). Furthermore, when the non-ABCG2 substrate cisplatin was used, its IC50 values were not affected by either sildenafil or FTC in any of the cell lines. A similar phenomenon was observed in both wild-type and mutant ABCG2-transfected HEK293 cells (Table 10). Representative cell survival curves (FIGS. 8-11) show that the survival curves in the presence of sildenafil were shifted remarkably to the left in the ABCB1- or ABCG2-overexpressing cells. Based on the above results, it appears that sildenafil significantly inhibits ABCB1-mediated drug efflux and partially reverses ABCG2-mediated efflux.
  • TABLE 8
    The Reversal Effect of Sildenafil and Verapamil on ABCB1-Mediated Resistancea
    IC50 ± SDb (μM)
    Compounds KB-3-1 KB-C2 KB-V1
    Colchicine  0.0057 ± 0.0015 (1.0)c  4.292 ± 1.493 (747.5)  0.240 ± 0.035 (41.8)
    + Sildenafil (2.5 μM) 0.0062 ± 0.0013 (1.1)  0.721 ± 0.236 (125.5)  0.059 ± 0.035 (10.3)
    + Sildenafil (5 μM) 0.0058 ± 0.0021 (1.0)  0.163 ± 0.031 (28.4) 0.052 ± 0.022 (9.1)
    + Sildenafil (10 μM) 0.0051 ± 0.00148 (0.9)   0.060 ± 0.004 (10.5) 0.044 ± 0.024 (7.6)
    + Verapamil (2.5 μM) 0.0054 ± 0.0016 (0.9)  0.264 ± 0.132 (46.0)  0.168 ± 0.036 (29.2)
    + Verapamil (5 μM) 0.0047 ± 0.0021 (0.8)  0.101 ± 0.026 (17.6)  0.081 ± 0.037 (14.1)
    + Verapamil (10 μM) 0.0032 ± 0.0001 (0.6) 0.055 ± 0.009 (9.6) 0.055 ± 0.038 (9.6)
    Vinblastine  0.0452 ± 0.0010 (1.0)c  0.464 ± 0.234 (10.3)  5.427 ± 3.362 (120.1)
    + Sildenafil (2.5 μM) 0.0445 ± 0.0114 (1.0) 0.139 ± 0.086 (3.1) 0.295 ± 0.211 (6.5)
    + Sildenafil (5 μM) 0.0443 ± 0.0076 (1.0) 0.092 ± 0.085 (2.0) 0.131 ± 0.091 (2.9)
    + Sildenafil (10 μM) 0.0437 ± 0.0173 (1.0) 0.036 ± 0.016 (0.8) 0.084 ± 0.049 (1.9)
    + Verapamil (2.5 μM) 0.0400 ± 0.0148 (0.9) 0.123 ± 0.109 (2.7) 0.238 ± 0.085 (5.3)
    + Verapamil (5 μM) 0.0250 ± 0.0038 (0.6) 0.056 ± 0.035 (1.2) 0.107 ± 0.015 (2.4)
    + Verapamil (10 μM) 0.0222 ± 0.0002 (0.5) 0.038 ± 0.021 (0.8) 0.057 ± 0.013 (1.3)
    Paclitaxel  0.0056 ± 0.0006 (1.0)c  4.380 ± 0.802 (788.5)   5.997 ± 1.952 (1079.6)
    + Sildenafil (2.5 μM) 0.0059 ± 0.0014 (1.1)  0.056 ± 0.027 (10.0)  0.293 ± 0.337 (52.7)
    + Sildenafil (5 μM) 0.0057 ± 0.0010 (1.0) 0.024 ± 0.016 (4.4)  0.144 ± 0.171 (25.8)
    + Sildenafil (10 μM) 0.0056 ± 0.0021 (1.0) 0.013 ± 0.007 (2.4)  0.071 ± 0.040 (12.8)
    + Verapamil (2.5 μM) 0.0048 ± 0.0021 (0.9) 0.049 ± 0.039 (8.9)  0.237 ± 0.308 (42.7)
    + Verapamil (5 μM) 0.0042 ± 0.0011 (0.7) 0.012 ± 0.003 (2.1) 0.055 ± 0.044 (9.9)
    + Verapamil (10 μM) 0.0034 ± 0.0007 (0.6) 0.009 ± 0.004 (1.7) 0.042 ± 0.017 (7.5)
    Cisplatin   1.745 ± 0.161 (1.0)c 1.726 ± 0.083 (1.0) 1.756 ± 0.178 (1.0)
    + Sildenafil (2.5 μM)  1.637 ± 0.088 (0.9) 1.884 ± 0.320 (1.1) 1.835 ± 0.071 (1.1)
    + Sildenafil (5 μM)  1.601 ± 0.030 (0.9) 1.838 ± 0.206 (1.1) 1.865 ± 0.006 (1.1)
    + Sildenafil (10 μM)  1.628 ± 0.157 (0.9) 1.651 ± 0.104 (0.9) 1.811 ± 0.081 (1.0)
    + Verapamil (2.5 μM)  1.729 ± 0.231 (1.0) 1.790 ± 0.250 (1.0) 1.784 ± 0.165 (1.0)
    + Verapamil (5 μM)  1.623 ± 0.001 (0.9) 1.762 ± 0.142 (1.0) 1.747 ± 0.056 (1.0)
    + Verapamil (10 μM)  1.457 ± 0.007 (0.8) 1.738 ± 0.042 (1.0) 1.797 ± 0.106 (1.0)
    aCell survival was determined by MTT assay as described in the Example 2.1.c.
    bData are expressed as means ± SD of at least three independent experiments carried out in triplicate.
    cFold-resistance values (values in parentheses) were calculated by dividing all IC50 values obtained for colchicine, vinblastine, paclitaxel, and cisplatin by the respective IC50 value for colchicine, vinblastine, paclitaxel, and cisplatin in the KB-3-1 cells in the absence of a reversal agent.
  • TABLE 9
    The Reversal Effect of Sildenafil and FTC on ABCG2-Mediated Resistance in Drug-Selected Cell Linesa
    IC50 ± SDb (μM)
    S1/ MCF-7/ MCF7/
    Compounds S1 FLV5000 S1-M1-80 MCF-7 Flv1000 ADVP3000
    Flavopiridol 0.0961 ± 0.0096 6.2046 ± 0.4631 0.6992 ± 0.0470 0.3443 ± 0.0021  6.8556 ± 0.4794  3.9005 ± 0.4139
     (1.0)c (64.9)  (7.3)  (1.0)c (19.9)  (11.3) 
    +Sildenafil 0.0854 ± 0.0291 5.4169 ± 0.1866 0.1310 ± 0.0192 0.3445 ± 0.0342  3.3609 ± 0.2038  2.0516 ± 0.2546
    (10 μM) (0.9) (56.4)  (1.4) (1.0) (9.8) (6.0)
    +Sildenafil 0.0767 ± 0.0292 5.1281 ± 0.3575 0.0951 ± 0.0351 0.3605 ± 0.0445  0.4531 ± 0.0151  0.5682 ± 0.0321
    (50 μM) (0.8) (53.4)  (1.0) (1.0) (1.3) (1.7)
    +FTC 0.1239 ± 0.0388 5.6540 ± 0.1843 0.0948 ± 0.0148 0.3441 ± 0.0148  0.2777 ± 0.0197  0.4166 ± 0.0147
    (2.5 μM) (1.3) (58.8)  (1.0) (1.0) (0.8) (1.2)
    Mitoxantrone 0.3796 ± 0.0416 1.1218 ± 0.1437 223.60 ± 8.60  7.6649 ± 0.1934 511.35 ± 16.71 279.25 ± 6.03 
     (1.0)c (3.0) (559.3)   (1.0)c (66.7)  (36.4) 
    +Sildenafil 0.1917 ± 0.0006 0.4212 ± 0.1297 113.65 ± 12.80  2.6792 ± 0.0243 127.21 ± 2.62  96.77 ± 3.01
    (10 μM) (0.5) (1.1) (284.3)  (0.3) (16.6)  (12.6) 
    +Sildenafil 0.1085 ± 0.0344 0.3469 ± 0.0330 32.95 ± 2.33  1.3434 ± 0.0843 36.93 ± 2.12 24.39 ± 3.66
    (50 μM) (0.3) (0.9) (82.4)  (0.2) (4.8) (3.2)
    +FTC 0.3119 ± 0.0310 0.9986 ± 0.0218 3.55 ± 1.91 1.4795 ± 0.2029 18.45 ± 1.63 13.82 ± 0.67
    (2.5 μM) (0.8) (2.6) (8.9) (0.2) (2.4) (1.8)
    SN-38 0.2449 ± 0.0170 0.3442 ± 0.0874 3.5533 ± 1.3962 1.2218 ± 0.2626 22.827 ± 2.001 14.301 ± 1.617
     (1.0)c (1.4) (14.4)   (1.0)c (18.7)  (11.3)
    +Sildenafil 0.2488 ± 0.0598 0.1647 ± 0.0063 1.4886 ± 0.5598 0.6009 ± 0.0071 10.639 ± 1.537  3.861 ± 0.173
    (10 μM) (1.0) (0.7) (6.0) (0.5) (8.7) (3.2)
    +Sildenafil 0.2070 ± 0.0244 0.1584 ± 0.0151 0.4924 ± 0.0281 0.3137 ± 0.0346  4.214 ± 0.277  1.784 ± 0.086
    (50 μM) (0.8) (0.6) (2.0) (0.3) (3.4) (1.5)
    +FTC 0.2808 ± 0.0280 0.2933 ± 0.0265 0.1014 ± 0.0310 0.2993 ± 0.0272  2.580 ± 0.132  0.198 ± 0.049
    (2.5 μM) (1.1) (1.2) (0.4) (0.2) (2.1) (0.2)
    Cisplatin 14.70 ± 2.23  53.45 ± 5.80  18.00 ± 2.59  34.41 ± 1.76  35.12 ± 1.86 31.12 ± 3.20
     (1.0)c (3.6) (1.2)  (1.0)c (1.0) (0.9)
    +Sildenafil 15.39 ± 2.94  56.27 ± 1.93  20.08 ± 1.43  34.76 ± 0.80  34.43 ± 1.32 30.72 ± 3.79
    (10 μM) (1.0) (3.8) (1.4) (1.0) (1.0) (0.9)
    +Sildenafil 15.90 ± 3.35  56.88 ± 4.19  20.64 ± 2.24  32.27 ± 1.70  34.47 ± 5.58 28.01 ± 4.65
    (50 μM) (1.1) (3.9) (1.4) (1.0) (1.0) (0.8)
    +FTC 13.27 ± 0.52  47.35 ± 4.48  15.30 ± 1.38  33.24 ± 1.41  35.02 ± 1.04 31.13 ± 0.33
    (2.5 μM) (0.9) (3.2) (1.0) (1.0) (1.0) (0.9)
    aCell survival was determined by MTT assay as described in the Example 2.1.c.
    bData are expressed as means ± SD of at least three independent experiments performed in triplicate.
    cFold-resistance values (values in parentheses) were calculated by dividing all IC50 values obtained for flavopiridol, mitoxantrone, SN-38, and cisplatin by the respective IC50 value for flavopiridol, mitoxantrone, SN-38, and cisplatin in the S1 cells in the absence of a reversal agent.
  • TABLE 10
    The Reversal Effect of Sildenafil and FTC on ABCG2-Mediated Resistance in ABCG2-Transfected Cell Linesa
    IC50 ± SDb (μM)
    Compounds HEK293/pcDNA3 HEK/ABCG2-G2 HEK/ABCG2-R5 HEK/ABCG2-T7
    Flavopiridol  0.1640 ± 0.0171 (1.0)c 0.3976 ± 0.0910 (2.4) 0.2821 ± 0.0148 (1.7) 0.7578 ± 0.0423 (4.6)
    + Sildenafil (10 μM) 0.1493 ± 0.0059 (0.9) 0.1216 ± 0.0112 (0.7) 0.1670 ± 0.0107 (1.0) 0.2256 ± 0.0108 (1.4)
    + Sildenafil (50 μM) 0.1599 ± 0.0180 (1.0) 0.1122 ± 0.0117 (0.7) 0.1466 ± 0.0030 (0.9) 0.1916 ± 0.0316 (1.2)
    + FTC (2.5 μM) 0.1276 ± 0.0033 (0.8) 0.0988 ± 0.0386 (0.6) 0.1643 ± 0.0236 (1.0) 0.1643 ± 0.0296 (1.0)
    Mitoxantrone  0.0356 ± 0.0030 (1.0)c  1.1042 ± 0.6023 (31.0)  0.4574 ± 0.3924 (12.8)  0.8424 ± 0.1858 (23.7)
    + Sildenafil (10 μM) 0.0115 ± 0.0007 (0.3) 0.0875 ± 0.0045 (2.5) 0.1142 ± 0.0004 (3.2) 0.1168 ± 0.0118 (3.3)
    + Sildeuafil (50 μM) 0.0057 ± 0.0006 (0.2) 0.0429 ± 0.0061 (1.2) 0.0784 ± 0.0207 (2.2) 0.0418 ± 0.0020 (1.2)
    + FTC (2.5 μM) 0.0315 ± 0.0092 (0.9) 0.0696 ± 0.0054 (2.0) 0.0420 ± 0.0156 (1.2) 0.0919 ± 0.0208 (2.6)
    SN-38  0.0058 ± 0.0002 (1.0)c 0.2042 ± 0.1362 (2.3)  0.1282 ± 0.0882 (22.1)  0.1171 ± 0.0704 (20.2)
    + Sildenafil (10 μM) 0.0034 ± 0.0010 (0.6) 0.0101 ± 0.0021 (1.7) 0.0175 ± 0.0018 (3.0) 0.0488 ± 0.0186 (8.4)
    + Sildenafil (50 μM) 0.0019 ± 0.0003 (0.3) 0.0044 ± 0.0100 (0.8) 0.0063 ± 0.0029 (1.1) 0.0139 ± 0.0025 (2.4)
    + FTC (2.5 μM) 0.0036 ± 0.0092 (0.8) 0.0050 ± 0.0009 (1.9) 0.0081 ± 0.0051 (1.4) 0.0082 ± 0.0023 (1.4)
    Cisplatin  1.6300 ± 0.1697 (1.0)c 1.2305 ± 0.0841 (0.8) 1.3265 ± 0.2001 (0.8) 1.8435 ± 0.0233 (1.1)
    + Sildenafil (10 μM) 1.5640 ± 0.0156 (1.0) 1.1371 ± 0.1577 (0.7) 1.3855 ± 0.0955 (0.9) 1.8115 ± 0.0403 (1.1)
    + Sildenafil (50 μM) 2.0180 ± 0.4285 (1.2) 1.1993 ± 0.1429 (0.7) 1.9820 ± 0.0438 (1.2) 1.9680 ± 0.0665 (1.2)
    + FTC (2.5 μM) 1.1535 ± 0.0488 (0.7)  1.0466 ± 0.028 (0.6) 1.3495 ± 0.2072 (0.8) 1.9530 ± 0.0834 (1.2)
    aCell survival was determined by MTT assay as described in the Example 2.1.c.
    bData are expressed as means ± SD of at least three independent experiments carried out in triplicate.
    cFold-resistance values (values in parentheses) were calculated by dividing all IC50 values obtained for flavopiridol, mitoxantrone, SN-38, and cisplatin by the respective IC50 value for flavopiridol, mitoxantrone, SN-38, and cisplatin in the HEK293/pcDNA3 cells in the absence of a reversal agent.
  • Example 2.3 Sildenafil Increases Accumulation of [3H]-Paclitaxel in ABCB1-Overexpressing Cells, and [3H]-Mitoxantrone and BODIPY-Prazosin in ABCG2-Overexpressing Cells
  • To investigate the potential mechanism by which sildenafil sensitizes ABCB1- and ABCG2-overexpressing cells to chemotherapeutic drugs, the effect of sildenafil on the accumulation of chemotherapeutic drugs in ABCB1- or ABCG2-overexpressing cells was examined. Intracellular [3H]-paclitaxel was measured in ABCB1-overexpressing cells in the presence or absence of sildenafil, and the results are shown in FIG. 4A. After 2 h of incubation, the intracellular levels of [3H]-paclitaxel in ABCB1-overexpressing KB-C2 and KB-V1 cells were significantly lower than that of the parental KB-3-1 cells. Sildenafil at 10 μM significantly increased the intracellular level of [3H]-paclitaxel, close to the effect of verapamil at 10 μM in KB-C2 and KB-V1 cells. Neither sildenafil nor verapamil altered the intracellular levels of [3H]-paclitaxel in parental KB-3-1 cells. Similarly, the intracellular levels of [3H]-mitoxantrone were measured in the ABCG2-overexpressing cells in the presence or absence of sildenafil (FIG. 4B-D). The intracellular levels of [3H]-mitoxantrone in cells expressing either wild-type or mutant ABCG2 were significantly less than that in parental cells. In the presence of sildenafil, either at 10 or 50 μM, all ABCG2-overexpressing cell lines displayed elevated intracellular [3H]-mitoxantrone levels, and intracellular levels of [3H]-mitoxantrone increased with increasing concentrations of sildenafil. However, the effects of sildenafil at 50 μM were less than those observed for FTC at 2.5 μM. Neither sildenafil nor FTC affected the intracellular levels of [3H]-mitoxantrone in parental cells.
  • In addition, the effect of sildenafil on the accumulation of a known fluorescent substrate of ABCG2 in the ABCG2-overexpressing cells was evaluated. Sildenafil at 50 μM enhanced the accumulation of BODIPY-prazosin in cells expressing either wild-type or mutant ABCG2, but demonstrated weaker enhancement than that of FTC at 2.5 μM. Representative histograms for BODIPY-prazosin are shown in FIGS. 5A-D. Taken together, these data are in agreement with the cytotoxicity results and suggest that sildenafil is able to inhibit the efflux function of ABCB1 and ABCG2, leading to the significant increase of intracellular accumulation of [3H]-paclitaxel in ABCB1-overexpressing cells and [3H]-mitoxantrone as well as BODIPY-prazosin in ABCG2-overexpressing cells.
  • Example 2.4 Sildenafil Inhibits Transport of E217βG and Methotrexate by ABCG2
  • To assess the potency of sildenafil as an in vitro inhibitor of ABCG2, the ability of sildenafil to inhibit the transport activity of ABCG2 was analyzed using the chemotherapeutic drug substrate [3H]-methotrexate and the physiologic substrate [3H]-E217βG. In Shi et al. ((2007) Cancer Res. 97:11012-20), it was shown that only wild-type ABCG2 was able to transport methotrexate and E217βG by the in vitro transport system. Thus, membrane vesicles prepared from HEK293/pcDNA3.1 and ABCG2-482-R5 cells were used (FIGS. 5E-F). Sildenafil significantly inhibited the rates of both methotrexate and E217βG uptake in the membrane vesicles of wild-type ABCG2 in a concentration-dependent manner, but its inhibitory effect was weaker than that of FTC at the same concentration. FTC also moderately decreased the uptake rates of both methotrexate and E217βG in the membrane vesicles of HEK293/pcDNA3.1, but sildenafil did not. These in vitro transport results suggest that sildenafil is able to directly inhibit the transport function of E217βG and methotrexate in wild-type ABCG2-expressing cells.
  • Example 2.5 Sildenafil Stimulates ATPase Activity and Affects the Photoaffinity Labeling of ABCB1 and ABCG2 with [125I]-IAAP
  • Generally, the substrates of ABC transporters stimulate their ATPase activity, and among the reversal agents, some (e.g., verapamil) stimulate the activity, whereas others (e.g., cyclosporine A) inhibit ATP hydrolysis (Ambudkar et al. (1999) Annu. Rev. Pharmacol Toxicol. 39:361-98). To assess the effect of sildenafil on the ATPase activity of ABCB1 and ABCG2, the membrane vesicles of HIGH FIVE™ insect cells overexpressing ABCB1 or ABCG2 were used in the presence of various concentrations of sildenafil under conditions that suppressed the activity of other major membrane ATPases. As shown in FIGS. 6A and 6B, sildenafil at the indicated concentrations potently stimulated the ATPase activity of ABCB1, but mildly stimulated the ATPase activity of ABCG2. The concentrations of sildenafil required for 50% stimulation of ATPase activity of ABCB1 and ABCG2 were 5-10 and 0.25-0.5 μM, respectively. These results indicate that sildenafil may be a substrate for ABCB1 and ABCG2.
  • The photoaffinity analogue of prazosin, [125I]-IAAP, which is transported by both ABCB1 and ABCG2, has been widely used to determine the binding regions of ABCB1 and ABCG2 that interact with substrates and inhibitors. To test whether sildenafil interacts at the prazosin binding site of ABCB1 or ABCG2, the ability of sildenafil to prevent photolabeling of ABCB1 and ABCG2 with [125I]-IAAP was examined by using the membrane vesicles from HIGH FIVE™ insect cells transfected with ABCB1 or ABCG2. As shown in FIGS. 6C and 6D, sildenafil dose dependently inhibited the photoaffinity labeling of ABCB1 or ABCG2 with [125I]-IAAP. The 50% inhibition of the photoaffinity labeling of ABCB1 and ABCG2 with [125I]-IAAP by sildenafil was observed at sildenafil concentrations of 10 and 100 μM, respectively. The ABCB1 inhibitors cyclosporine A (at 10 μM) and verapamil (at 20 μM) inhibited the [125I]-IAAP photolabeling of ABCB1 up to about 95% and 65%, respectively, and the ABCG2 inhibitor FTC (at 10 μM) inhibited the [125I]-IAAP photolabeling of ABCG2 to about 40%, compared with membranes incubated with no inhibitor. Thus, these data suggest that sildenafil competes with IAAP to bind both ABCB1 and ABCG2, and that sildenafil interacts with the transmembrane regions of both transporters.
  • Example 2.6 Model for Sildenafil Binding to ABCB1
  • PDE5A homology model of human ABCB1 based on the mouse (Mdr3) ABCB1-QZ59-RRR cocrystal structure was used to predict sildenafil-ABCB1 binding conformation. A human ABCB1 homology model (FIG. 7A) was then used to dock sildenafil using Glide docking software to investigate its potential binding mode. Three binding sites were reported in the crystal structure of mouse Mdr3 as represented by ABCB1-QZ59-RRR (site-1), ABCB1-QZ59-SSS (site-2), and ABCB1-verapamil (site-3). Aller et al., supra. To identify a favorable binding site on ABCB1 for sildenafil, docking experiments using these sites were carried out. Because the photoaffinity labeling data suggested that sildenafil displaces IAAP in a dose-dependent manner, IAAP was also docked to these sites for comparison. These data also indicated that sildenafil and IAAP share the same binding site on ABCB1 (i.e., site-1). Binding energy data for the docked poses of sildenafil and IAAP were compared at each of the binding sites (Table 11). On the basis of binding energy data analysis, both ligands (sildenafil and IAAP) were found to bind most favorably within the QZ59-RRR binding site of ABCB1. Bound conformation of sildenafil in context of site-1 is addressed below.
  • TABLE 11
    Binding Energies of Sildenafil and IAAP in Their Ionized States within
    Each of the Predicted Binding Sites of ABCB1
    Glide score kcal/mol
    Ligands Site-1a Site-2b Site-3c Site-4d
    Figure US20140288078A1-20140925-C00004
    −8.48 −5.72 −5.20 −3.21
    Figure US20140288078A1-20140925-C00005
    −8.89 −5.79 −4.54 −5.16
    a-Site represented by bound QZ59-RRR.
    b-Site represented by bound ligand QZ59-SSS.
    c-Verapamil binding site.
    d-Site grid generated using residues Phe728 and Val982, which are known to be common to the above three sites.
  • The XP-Glide-predicted binding mode of sildenafil shows the importance of hydrophobic interactions within the large drug binding cavity of ABCB1 (FIG. 7B). The N-methylpiperazine (D-ring) of sildenafil was found to be stabilized by hydrophobic contacts with the side chains of Met69 of transmembrane (TM) 1 and Phe336, Leu339, and Ile340 of TM6. The C-ring along with its ethoxy substituent enters into favorable hydrophobic interactions with residues Phe72 of TM1, Leu975 and Phe978 of TM12. The A-ring (along with its methyl and propyl substituents) and the B-ring forms hydrophobic interactions with the side chains of Phe728, Ala729, and Phe732 of TM7 and Val982 of TM12. In addition to the hydrophobic contacts, sildenafil also seems to be stabilized by electrostatic interactions with key residues Tyr953 of TM11 and Tyr307 of TM5. For example, the sulfonyl oxygen atom enters into a hydrogen bonding interaction with the hydroxyl group of Tyr953 (—SO2—HO-Tyr953), whereas the carbonyl oxygen atom present in the B-ring is located at a distance of 3.9 Å from the side chain hydroxyl group of Tyr307. Transmembrane domain numbering is as reported previously (Aller et al., supra).
  • Example 3 Effect of Sildenafil on Antineoplastic Drug Treatment of Tumors in a Mouse Model Example 3.1 Materials and Methods Example 3.1.a Materials
  • Paclitaxel (as antineoplastic drug) and saline (as vehicle) were purchased from Sigma Chemical Company. Sildenafil was purified from 100 mg VIAGRA® tablets as described in Example 2.1.a.
  • Example 3.1.b Cell Lines and Experimental Animals
  • KB-3-1 cells and the ABCB1/Pgp-overexpressing drug-resistant cell line KB-C2 cells were obtained as described in Example 2.1.b.
  • Six-to-eight-week old male athymic NCR nude mice from Taconic (Hudson, N.Y.) were used for all xenografts. The mice were provided with sterilized food and water. The animal protocol was reviewed and approved by the Institutional Animal Care and Use Committee at St. John's University.
  • Example 3.1.c Mouse Tumor Xenografts
  • KB-3-1 or KB-C2 cells were grown in flasks, harvested and implanted (at 2.5×106 cells in 0.2 mL for KB-3-1 cells and 4.0×106 cells in 0.2 mL for KB-C2 cells) subcutaneously under the shoulder in the nude mice. When the tumors reached 0.5 cm, the mice were randomized into four groups, six mice per group: (1) vehicle, (2) sildenafil (given at 75 mg/kg p.o.), (3) paclitaxel (given at 18 mg/kg i.p.), and (4) paclitaxel and sildenafil, all treated every three days for a total of six administrations. The mice were observed for 2-3 weeks, and the tumor volumes estimated every three days from the perpendicular diameters (A and B) according to the following formula: V=π/6×((A+B)/2)3 (see, e.g., Dai et al. (2008) 68:7509-14).
  • Mice were euthanized when the tumor weight exceeded 1 gram in the control group, and the tumors were excised and weighed.
  • Example 3.2 Coadministration of Sildenafil and Paclitaxel Leads to Significant Decrease in Tumor Weight and Volume in KB-C2 Mouse Xenografts
  • The efficacy of sildenafil in blocking MDR in vivo was evaluated using mouse tumor xenografts. As demonstrated in FIGS. 12 and 13, there was a significant reduction in tumor weight and volume in mice treated with both sildenafil and paclitaxel in comparison with mice treated with paclitaxel alone.
  • Similar results were obtained from experiments on mouse xenografts of human lung cancer cell line H460/MX20 (which is an ABCG2-overexpressing cell line derived from parental H460 cells).
  • Example 4 Effects of Sildenafil, Vardenafil, and Tadalafil on ABCC10/MRP7-Mediated Multidrug Resistance Example 4.1 Materials and Methods Example 4.1.a Reagents
  • Sildenafil, vardenafil, and tadalafil were purchased from Toronto Research Chemicals Inc. (Ontario, Canada). Cepharanthine was provided by Daiichi Sankyo Pharmaceutical Co. Ltd (Tokyo, Japan). Dulbecco's modified Eagle's medium (DMEM), fetal bovine serum (FBS), penicillin/streptomycin and trypsin 0.25% were products of Hyclone (Logan, Utah). Monoclonal antibody 14C10 (against GAPDH) was acquired from Cell Signaling Technology, Inc. (Danvers, Mass.). Polyclonal antibody D-19 (against MRP7) was obtained from Santa Cruz Biotechnology Inc (Santa Cruz, Calif.). Alexa Flour 488 donkey anti-goat secondary antibody for immunocytochemistry was purchased from Molecular Probes (Eugene, Oreg.). [3H]-paclitaxel (46.5 Ci/mmol) was purchased from Moravek Biochemicals Inc (Brea, Calif.). Paclitaxel, docetaxel, vinblastine, DMSO, MTT, and other chemicals were purchased from Sigma Chemical (St. Louis, Mo.).
  • Example 4.1.b Cell Lines and Cell Culture
  • The MRP7 cDNA was generously provided by Dr. Gary Kruh (University of Illinois, Chicago, Ill.) and inserted into the pcDNA3.1 expression vector. The MRP7 expression vector and parental plasmid were introduced into HEK293 cells by electroporation as previously described (Chen et al. (2003) Mol. Pharmacol. 63:351-58). Individual colonies were selected in medium containing G418 (1 mg/ml) and cultured for further analysis. All the cell lines were grown as adherent monolayers in flasks with Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% bovine serum, 100 units/ml penicillin and 100 mg/ml streptomycin in a humidified incubator containing 5% CO2 at 37° C.
  • Example 4.1.c Cytotoxicity Assay
  • MTT colorimetric assay was performed to analyze the drug sensitivity as previously described (Shi et al. (2007) Cancer Res. 67:11012-20). HEK293-pcDNA3.1 and HEK/MRP7 cells were seeded into 96-well plates in triplicate at 6000 cells/well. After incubation in DMEM supplemented with 10% bovine serum at 37° C. for 24 h, three different nontoxic concentrations of sildenafil, vardenafil, and tadalafil (1.25, 2.5, and 5 μM) were added into plates 1 h prior to the addition of the substrates of MRP7 (paclitaxel, docetaxel, and vinblastine).
  • After drug incubation of 68 h, 20 μl MTT solution (4 mg/ml) was added into each well. The plates were further incubated for 4 h, then the medium was discarded, and 100 μl DMSO was added into each well to dissolve the formazan crystals. The absorbance was determined at 570 nm by an OPSYS Microplate Reader from DYNEX Technologies, Inc. (Chantilly, Va.). The degree of resistance was calculated by dividing the IC50 values (concentrations required to inhibit growth by 50%) for the HEK/MRP7 cells by those of the parental HEK293-pcDNA3.1 cells. The Bliss method was used to calculate the IC50 values according to survival curves.
  • Example 4.1.d [3H]-Paclitaxel Accumulation and Efflux
  • The effect of PDE5 inhibitors on the intracellular accumulation of paclitaxel in HEK293-pcDNA3.1 and HEK/MRP7 cells was measured using [3H]-paclitaxel. HEK293-pcDNA3.1 and HEK/MRP7 cells were trypsinized and four aliquots from each cell line were suspended in the medium, aliquots were preincubated with medium-only (control), sildenafil, vardenafil, or tadalafil (5 μM each) at 37° C. for 2 h, then incubated with 0.1 μM [3H]-paclitaxel for another 2 h. For efflux studies, the cells were treated the same as in the drug accumulation study, and then washed three times with ice-cold PBS, and suspended in fresh medium with or without PDE5 inhibitors. Aliquots were evenly collected at various time points (0, 30, 60, and 120 min). Samples from both accumulation and efflux experiments were washed with ice-cold PBS three times and placed in scintillation fluid, and the radioactivity was measured in a Packard TRI-CARB® 1900CA liquid scintillation analyzer from Packard Instrument Company, Inc. (Downers Grove, Ill.).
  • Example 4.1.e Preparation of Total Cell Lysates and Immunoblotting Analysis
  • To determine the effect of PDE5 inhibitors on the expression of MRP7, HEK/MRP7 cells were incubated with 5 μM sildenafil, vardenafil, or tadalafil for different time periods (0, 24, 48 and 72 h), then harvested and rinsed twice with cold PBS. The total cell lysates were collected with radioimmunoprecipitation assay (RIPA) buffer (1×PBS, 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS, 100 μg/ml phenylmethylsulfonyl fluoride, 10 μg/ml aprotinin, 10 μg/ml leupeptin) for 30 min with occasional rocking followed by centrifugation at 12,000 rpm at 4° C. for 15 min.
  • The protein concentration was determined by a bicinchoninic acid (BCATM)-based protein assay (Thermo Scientific, Rockford, Ill.). Equal amounts of total cell lysates (40 μg of protein) were resolved by 4-12% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and electrophoretically transferred onto PVDF membranes. After being incubated in blocking solution containing 5% skim milk in TBST buffer (10 mM Tris-HCL, pH 8.0, 150 mM NaCl and 0.1% Tween 20) at room temperature for 1 h, the membranes were immunoblotted overnight with primary antibodies to anti-MRP7 (1:200 dilution) and anti-GAPDH (1:1000 dilution) at 4° C.
  • Subsequently, the membranes were washed three times for 15 min with TBST buffer and incubated at room temperature for 2 h with horseradish peroxide (HRP)-conjugated secondary antibody (1:2000 dilution). The protein-antibody complex was detected by the enhanced Phototope TM-HRP Detection Kit (Cell Signaling Technology, Inc.) and exposed to Kodak medical X-ray processor (Eastman Kodak Co.). The protein expression was quantified by Scion Image software (Scion Co., Frederick, Md.).
  • Example 4.1.f Immunofluorescence Analysis
  • HEK/MRP7 cells (1×104) were seeded in 24 well plates and cultured overnight. Sildenafil, vardenafil, or tadalafil at 5 μM were added into the wells and then cultured at 37° C. for 72 h in a humidified incubator containing 5% CO2. Cells were washed with PBS and fixed with 4% paraformaldehyde for 15 min at room temperature and then rinsed with PBS three times. Nonspecific reaction was blocked with 1% BSA for 1 h at room temperature. A polyclonal antibody D19 against MRP7 (1:200) was added and incubated overnight. Cells were then incubated with Alexa Flour® 488 donkey anti-goat IgG (1:2000) for 1 h at room temperature. DAPI was used for nuclear staining. Immunofluorescent images were taken with an inverted microscope (model IX70; Olympus, Center Valley, Pa.) with IX-FLA fluorescence and CCD camera.
  • Example 4.1.g Statistical Analysis
  • All experiments were repeated at least three times, and the differences were determined by using the Student's t-test. The statistical significance was determined at p<0.05.
  • Example 4.2 Effects of PDE5 Inhibitors on the Sensitivity of Antineoplastic Drugs in HEK293-pcDNA3.1 and HEK/MRP7 Cells
  • In order to analyze the reversal efficacy of PDE5 inhibitors, the cytotoxicities of antineoplastic drugs (paclitaxel, docetaxel, or vinblastine) alone and in combination with a PDE5 inhibitor (sildenafil, vardenafil, or tadalafil) were investigated at nontoxic concentrations (1.25, 2.5, and 5 μM) in HEK293-pcDNA3.1 and HEK/MRP7 cells. As shown in Table 12 and FIG. 14, HEK/MRP7 cells compared to parental HEK293-pcDNA3.1 cells exhibited a significant resistance to various MRP7 substrates such as paclitaxel, docetaxel, and vinblastine, which is consistent with previous reports (e.g., Shi et al. (2011) Cancer Res. 71:3029-41). Sildenafil, vardenafil, and tadalafil dose-dependently decreased the IC50 values of the above-mentioned MRP7 substrates for HEK/MRP7 cells.
  • Tadalafil showed the least reversal effect of the three PDE5 inhibitors. Cepharanthine, a known MRP7 inhibitor used as a positive control at 2.5 μM, completely reversed the resistance of HEK/MRP7 cells to paclitaxel, docetaxel, and vinblastine. In contrast, sildenafil, vardenafil, or tadalafil did not significantly reverse the resistance of HEK/MRP7 cells to cisplatin, a nonsubstrate of MRP7 (Table 12, FIG. 14). In the parental HEK293-pcDNA3.1, the IC50 values of paclitaxel, docetaxel, and vinblastine in the presence or absence of sildenafil, vardenafil, or tadalafil showed no significant difference (Table 12, FIG. 14).
  • TABLE 12
    The effects of PDE5 inhibitors on the sensitivity of HEK293-pcDNA3.1 and
    HEK/MRP7 cells to paclitaxel, docetaxel, vinblastine and cisplatin.
    IC50 ± SDa (nM)
    Compounds HEK 293-pcDNA-3.1 HEK/MRP7
    Paclitaxel 11.64 ± 1.33 (1.0)b 107.18 ± 11.25 (9.21)
    + Sildenafil 1.25 μM 10.69 ± 1.04 (0.92) 45.47 ± 3.78** (3.91)
    + Sildenafil 2.5 μM  9.96 ± 0.92 (0.86) 28.35 ± 2.41** (2.44)
    + Sildenafil 5 μM  9.38 ± 0.86 (0.81) 13.37 ± 2.07** (1.15)
    + Vardenafil 1.25 μM 10.85 ± 1.27 (0.93) 34.85 ± 3.36** (2.99)
    + Vardenafil 2.5 μM  9.63 ± 0.89 (0.83) 19.86 ± 2.61** (1.71)
    + Vardenafil 5 μM  9.14 ± 1.01 (0.79) 12.39 ± 1.54** (1.06)
    + Tadalafil 1.25 μM 12.43 ± 0.95 (1.07)  93.78 ± 6.23 (8.06)
    + Tadalafil 2.5 μM 11.41 ± 1.22 (0.98)  81.25 ± 7.16* (6.98)
    + Tadalafil 5 μM 10.84 ± 0.77 (0.93) 68.36 ± 5.83** (5.87)
    + Cepharanthine 2.5 μM  8.97 ± 1.18 (0.77) 11.81 ± 0.82** (1.01)
    Docetaxel 5.73 ± 0.65 (1.0)   64.81 ± 5.19 (11.31)
    + Sildenafil 1.25 μM  5.35 ± 0.47 (0.93) 38.29 ± 3.75** (6.68)
    + Sildenafil 2.5 μM  5.28 ± 0.41 (0.92) 22.37 ± 2.97** (3.90)
    + Sildenafil 5 μM  4.74 ± 0.56 (0.83)  7.36 ± 0.83** (1.28)
    + Vardenafil 1.25 μM 5.72 ± 0.38 (1.0) 31.85 ± 3.62** (5.56)
    + Vardenafil 2.5 μM  4.88 ± 0.55 (0.85) 17.95 ± 2.57** (3.13)
    + Vardenafil 5 μM  4.59 ± 0.39 (0.80)  6.84 ± 0.79** (1.19)
    + Tadalafil 1.25 μM  6.26 ± 0.43 (1.09)   62.48 ± 5.03 (10.91)
    + Tadalafil 2.5 μM  6.35 ± 0.59 (1.11)  55.21 ± 5.87 (9.64)
    + Tadalafil 5 μM  5.86 ± 0.54 (1.02)  47.85 ± 3.98* (8.35)
    + Cepharanthine 2.5 μM 4.07 ± 0.52* (0.71)  5.88 ± 0.41** (1.03)
    Vinblastine 11.19 ± 1.23 (1.0)   56.31 ± 4.61 (5.03)
    + Sildenafil 1.25 μM 10.96 ± 0.95 (0.98) 36.19 ± 2.48** (3.23)
    + Sildenafil 2.5 μM 10.27 ± 0.82 (0.92) 25.92 ± 3.04** (2.32)
    + Sildenafil 5 μM  9.69 ± 0.97 (0.87) 14.37 ± 1.19** (1.28)
    + Vardenafil 1.25 μM 10.73 ± 1.11 (0.96) 33.72 ± 2.89** (3.01)
    + Vardenafil 2.5 μM 10.32 ± 1.03 (0.92) 21.45 ± 1.94** (1.92)
    + Vardenafil 5 μM  9.48 ± 0.98 (0.85) 12.39 ± 1.05** (1.11)
    + Tadalafil 1.25 μM 11.84 ± 0.88 (1.06)  51.28 ± 4.85 (4.58)
    + Tadalafil 2.5 μM 11.51 ± 1.19 (1.03)  47.95 ± 4.11 (4.29)
    + Tadalafil 5 μM 10.66 ± 1.08 (0.95)  40.54 ± 3.92* (3.62)
    + Cepharanthine 2.5 μM  8.92 ± 1.07 (0.80) 11.35 ± 0.93** (1.01)
    Cisplatin 1574.26 ± 84.95 (1.0)   1552.22 ± 74.39 (0.99) 
    + Sildenafil 5 μM 1730.35 ± 63.89 (1.10)   1629.48 ± 81.76 (1.04) 
    + Vardenafil 5 μM 1681.45 ± 102.34 (1.07)   1714.60 ± 71.43 (1.09) 
    + Tadalafil 5 μM 1746.87 ± 75.02 (1.11)   1757.12 ± 109.15 (1.12) 
    + Cepharanthine 2.5 μM 1677.38 ± 59.32 (1.07)   1635.30 ± 62.78 (1.04) 
    aIC50: concentration that inhibited cell survival by 50%. Data are means ± SD of at least three independent experiments performed in triplicate.
    bFold-resistance was determined by dividing the IC50 values of HEK/MRP7 cells by the IC50 values of HEK293-pcDNA3.1 cells in the absence or presence of sildenafil, vardenafil, tadalafil or verapamil.
    *represents p < 0.05;
    **represents p < 0.01
  • Example 4.3 PDE5 Inhibitors Increase the Intracellular Accumulation of [3H]-Paclitaxel in HEK/MRP7 Cells
  • As further confirmation of the effects of PDE5 inhibitors on the drug efflux function of MRP7, intracellular accumulation of [3H]-paclitaxel studies were performed. The intracellular concentration of [3H]-paclitaxel in HEK/MRP7 cells was significantly lower (28.5%) than that in parental HEK293-pcDNA3.1 cells (100%, FIG. 15). After the cells were incubated with either sildenafil, vardenafil, or tadalafil at 5 μM for 2 h, the intracellular accumulation of [3H]-paclitaxel in HEK/MRP7 cells was significantly increased by 3.3-, 3.7- and 2.1-fold, respectively, as compared to 2.5 μM of cepharanthine as a positive control (3.8-fold; FIG. 15). Neither PDE5 inhibitors nor cepharanthine significantly affected the intracellular levels of [3H]-paclitaxel in HEK293-pcDNA3.1 cells (FIG. 15).
  • Example 4.4 PDE5 Inhibitors Inhibit the Efflux of [3H]-Paclitaxel Mediated by MRP7 in HEK/MRP7 Cells
  • To ascertain whether the increase in the intracellular [3H]-paclitaxel accumulation in the presence of sildenafil, vardenafil, or tadalafil was due to the inhibition of [3H]-paclitaxel efflux by MRP7 (ABCC10), a time course study was designed to measure intracellular [3H]-paclitaxel levels in the presence of sildenafil, vardenafil, or tadalafil. As shown in FIG. 16, HEK/MRP7 cells significantly extruded a higher percentage of intracellular [3H]-paclitaxel than that in HEK293-pcDNA3.1 cells. However, in the presence of sildenafil, vardenafil, or tadalafil at 5 μM, there were significant decreases in the efflux of intracellular [3H]-paclitaxel at different time periods (0, 30, 60, and 120 min) from HEK/MRP7 cells, but not from the parental HEK293-pcDNA3.1 cells. The intracellular accumulation of [3H]-paclitaxel at 0 min was set as 100%, and at 30, 60 and 120 min, the percentages were 62.43%, 39.47%, and 23.44%, respectively, of the accumulated [3H]-paclitaxel that remained in HEK/MRP7 cells in the absence of PDE5 inhibitors. When HEK/MRP7 cells were incubated with sildenafil, the percentage of the intracellular [3H]-paclitaxel at 30, 60 and 120 min increased significantly to 90.62%, 82.14%, and 62.35%, respectively (FIG. 16A). Vardenafil significantly increased the percentages of the intracellular [3H]-paclitaxel at 30, 60 and 120 min to 95.34%, 93.58%, and 63.92%, respectively (FIG. 16B). Tadalafil at 30, 60 and 120 min increased significantly the percentage of [3H]-paclitaxel accumulation to 76.44%, 63.41%, and 46.48%, respectively (FIG. 16C). Sildenafil and vardenafil were more potent than tadalafil, which is consistent with the results in colorimetric growth assay and [3H]-paclitaxel accumulation experiments.
  • Example 4.5 PDE5 Inhibitors do not Alter the Expression of MRP7
  • Reversal of MRP7-mediated MDR can be achieved by either altering MRP7 expression or inhibiting MRP7 function. To evaluate the effects of sildenafil, vardenafil, and tadalafil on MRP7 expression, HEK/MRP7 cells were treated with sildenafil, vardenafil, or tadalafil at 5 μM for 0, 24, 48, and 72 h and the MRP7 expression levels were examined by Western blot analysis. The results shown in FIG. 17A indicated that PDE5 inhibitors do not significantly alter the protein expression levels of MRP7 in HEK/MRP7 cells.
  • Example 4.6 PDE5 Inhibitors do not Alter the Localization of MRP7
  • Presumably, the ABC transporters could be downregulated if they were translocated or dislodged from the plasma membrane to a cytosolic region. To rule out this possibility, an immunofluorescence assay was performed to examine whether the location of MRP7 was altered after treatment with a PDE5 inhibitor. As shown in FIG. 17, there was no alteration of MRP7 protein localization in plasma membranes after the treatment with sildenafil, vardenafil, or tadalafil at 5 μM for 72 h. The Western blotting (FIG. 17A) and immunocytochemical (FIG. 17B) experiments suggested that all three PDE5 inhibitors do not alter the expression and/or localization of the MRP7 transporter in HEK/MRP7 cells.
  • EQUIVALENTS
  • Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents of the specific embodiments of the invention described herein. Such equivalents also are intended to be encompassed by the following claims.

Claims (2)

1. A method of treating multidrug resistance in a cancer cell in a subject in need thereof by inhibiting ABCB1 transporter activity, comprising administering to the subject a therapeutically effective amount of a phosphodiesterase 5 (PDE5) inhibitor.
2-21. (canceled)
US14/215,982 2011-03-28 2014-03-17 Use of phosphodiesterase inhibitors for treating multidrug resistance Abandoned US20140288078A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/215,982 US20140288078A1 (en) 2011-03-28 2014-03-17 Use of phosphodiesterase inhibitors for treating multidrug resistance

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161465948P 2011-03-28 2011-03-28
US201161501984P 2011-06-28 2011-06-28
US13/432,315 US8673914B2 (en) 2011-03-28 2012-03-28 Use of phosphodiesterase inhibitors for treating multidrug resistance
US14/215,982 US20140288078A1 (en) 2011-03-28 2014-03-17 Use of phosphodiesterase inhibitors for treating multidrug resistance

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/432,315 Continuation US8673914B2 (en) 2011-03-28 2012-03-28 Use of phosphodiesterase inhibitors for treating multidrug resistance

Publications (1)

Publication Number Publication Date
US20140288078A1 true US20140288078A1 (en) 2014-09-25

Family

ID=46928026

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/432,315 Expired - Fee Related US8673914B2 (en) 2011-03-28 2012-03-28 Use of phosphodiesterase inhibitors for treating multidrug resistance
US14/215,982 Abandoned US20140288078A1 (en) 2011-03-28 2014-03-17 Use of phosphodiesterase inhibitors for treating multidrug resistance

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US13/432,315 Expired - Fee Related US8673914B2 (en) 2011-03-28 2012-03-28 Use of phosphodiesterase inhibitors for treating multidrug resistance

Country Status (1)

Country Link
US (2) US8673914B2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017139623A1 (en) * 2016-02-10 2017-08-17 Immunomedics, Inc. Combination of abcg2 inhibitors with sacituzumab govitecan (immu-132) overcomes resistance to sn-38 in trop-2 expressing cancers

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8673914B2 (en) * 2011-03-28 2014-03-18 St. John's University Use of phosphodiesterase inhibitors for treating multidrug resistance
EP3024457A4 (en) 2013-07-26 2017-06-28 Update Pharma Inc. Compositions to improve the therapeutic benefit of bisantrene
EP3936145A1 (en) * 2014-07-31 2022-01-12 The University Of Western Australia A method for the identification of immunotherapy-drug combinations using a network approach
EP3267986B1 (en) * 2015-03-13 2022-09-14 Indiana University Research & Technology Corporation Pde5 inhibitors for use in reducing renal cyst size and/or preventing renal cyst growth
CN106565599A (en) * 2016-06-20 2017-04-19 中国药科大学 2-aminomethylpyridylnicotinamides and preparation method and application thereof
MX2019007276A (en) * 2016-12-22 2019-11-11 Mavupharma Inc Compositions and methods of enhancing or augmenting type i ifn production.
WO2019157125A1 (en) * 2018-02-07 2019-08-15 L.E.A.F. Holdings Group Llc Alpha polyglutamated methotrexate and uses thereof
CA3144770A1 (en) * 2019-07-15 2021-01-21 Biolab Sanus Farmaceutica Ltda Stereoisomers of the compound 3-(benzo[d][1,3]dioxol-5-yl)-7-(1-hydroxypropan-2-yl)-1-(1h-indol-3-yl)-6,7-dihydro-3h-oxazol[3,4-a]pyrazine-5,8-dione and use thereof as an antitumor agent and phosphodiesterase enzyme inhibitor
CN110464726A (en) * 2019-08-20 2019-11-19 中国人民解放军第四军医大学 Application of the Tadalafei as protein arginine transmethylase PRMT5 inhibitor
CN111701009B (en) * 2020-05-18 2022-10-11 湖南科技学院 Polypeptide ABC transporter inhibitor XH-14C and application thereof
CN113082210B (en) * 2021-03-09 2023-04-18 广州白云山医药集团股份有限公司白云山制药总厂 Tumor chemotherapy pharmaceutical composition

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8673914B2 (en) * 2011-03-28 2014-03-18 St. John's University Use of phosphodiesterase inhibitors for treating multidrug resistance

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR930702292A (en) 1990-11-06 1993-09-08 알렌 제이. 스피겔 Quinazolin derivatives for enhancing tumor therapeutic activity
CA2238283C (en) 1997-05-30 2002-08-20 Cell Pathways, Inc. Method for identifying compounds for inhibition of neoplastic lesions, pharmaceutical compositions from such compounds and uses of such compounds and compositions for treating neoplastic lesions
US6703400B2 (en) 2001-02-23 2004-03-09 Schering Corporation Methods for treating multidrug resistance
US6861431B2 (en) 2001-03-23 2005-03-01 The Board Of Trustees Of The University Of Illinois Compounds capable of modulating the activity of multidrug transporters and therapeutic use of the same
US6906064B2 (en) 2002-06-10 2005-06-14 W. Joseph Thompson Method for treating a patient with neoplasia using Iressa
JP2009512677A (en) 2005-10-19 2009-03-26 ランバクシー ラボラトリーズ リミテッド Composition of phosphodiesterase type IV inhibitor
CA2645633A1 (en) 2006-04-05 2007-11-01 Novartis Ag Combinations of therapeutic agents for treating cancer
EP2328910B1 (en) 2008-06-04 2014-08-06 Synergy Pharmaceuticals Inc. Agonists of guanylate cyclase useful for the treatment of gastrointestinal disorders, inflammation, cancer and other disorders

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8673914B2 (en) * 2011-03-28 2014-03-18 St. John's University Use of phosphodiesterase inhibitors for treating multidrug resistance

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017139623A1 (en) * 2016-02-10 2017-08-17 Immunomedics, Inc. Combination of abcg2 inhibitors with sacituzumab govitecan (immu-132) overcomes resistance to sn-38 in trop-2 expressing cancers
CN108601841A (en) * 2016-02-10 2018-09-28 免疫医疗公司 The combination of ABCG2 inhibitor and SACITUZUMAB GOVITECAN (IMMU-132) overcome the resistance to SN-38 in the cancer for expressing TROP-2
US10954305B2 (en) 2016-02-10 2021-03-23 Immunomedics, Inc. Combination of ABCG2 inhibitors with sacituzumab govitecan (IMMU-132) overcomes resistance to SN-38 in Trop-2 expressing cancers

Also Published As

Publication number Publication date
US8673914B2 (en) 2014-03-18
US20120252816A1 (en) 2012-10-04

Similar Documents

Publication Publication Date Title
US8673914B2 (en) Use of phosphodiesterase inhibitors for treating multidrug resistance
Shi et al. Sildenafil reverses ABCB1-and ABCG2-mediated chemotherapeutic drug resistance
US10265279B2 (en) Combinations of LSD1 inhibitors for use in the treatment of solid tumors
US11325892B2 (en) Compounds and methods for treating cancer
Fung et al. High-throughput screening of small molecules identifies hepcidin antagonists
US20210212989A1 (en) Modulators of gtpases and their use
Ding et al. The phosphodiesterase-5 inhibitor vardenafil is a potent inhibitor of ABCB1/P-glycoprotein transporter
US10207988B2 (en) Methods and compositions for treatment of cancer and autoimmune disease
US20180085371A1 (en) Combination of panobinostat and ruxolitinib in the treatment of cancer such as a myeloproliferative neoplasm
Zabludoff et al. AZD7762, a novel checkpoint kinase inhibitor, drives checkpoint abrogation and potentiates DNA-targeted therapies
CN108348520A (en) MDM2 inhibitor and a combination thereof
Chen et al. Osimertinib (AZD9291) enhanced the efficacy of chemotherapeutic agents in ABCB1-and ABCG2-overexpressing cells in vitro, in vivo, and ex vivo
WO2020211847A1 (en) Application of tegaserod in preparation of anti-tumor drugs
US10888568B2 (en) Pharmaceutical composition for treatment of cancer using phenothiazine
Patel et al. Suppression of ABCG2 mediated MDR in vitro and in vivo by a novel inhibitor of ABCG2 drug transport
Tang et al. CEP-33779 antagonizes ATP-binding cassette subfamily B member 1 mediated multidrug resistance by inhibiting its transport function
US20130224306A1 (en) Method for treating a pulmonary hypertension condition without companion diagnosis
CA3191363A1 (en) Pharmaceutical combination and tumor treatment
WO2021216814A1 (en) Btk inhibitors to treat pulmonary distress in covid-19 patients
WO2023246940A1 (en) Method of treating a cancer through suppression of growth of cancer stem cells, and downregulation of wnt pathway
US20220193056A1 (en) Compositions and methods of treating pik3ca helical domain mutant cancers
US10478437B2 (en) Combination therapy to enhance the anticancer efficacy of platinum drugs
WO2021195706A1 (en) New dynamin inhibitors and uses
Zhang Preclinical pharmacology of the MDM2 antagonist nutlin-3a
WO2024015641A1 (en) Small molecule for treatment of cancer of the appendix

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION