EP1251840A2 - Method for using potassium channel activation for delivering a medicant to an abnormal brain region and/or a malignant tumor - Google Patents

Method for using potassium channel activation for delivering a medicant to an abnormal brain region and/or a malignant tumor

Info

Publication number
EP1251840A2
EP1251840A2 EP01905141A EP01905141A EP1251840A2 EP 1251840 A2 EP1251840 A2 EP 1251840A2 EP 01905141 A EP01905141 A EP 01905141A EP 01905141 A EP01905141 A EP 01905141A EP 1251840 A2 EP1251840 A2 EP 1251840A2
Authority
EP
European Patent Office
Prior art keywords
nonoate
potassium channel
medicant
agent
channel activator
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.)
Withdrawn
Application number
EP01905141A
Other languages
German (de)
French (fr)
Inventor
Keith L. Black
Nagendra S. Ningaraj
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.)
Cedars Sinai Medical Center
Original Assignee
Cedars Sinai Medical Center
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
Priority claimed from US09/491,500 external-priority patent/US7018979B1/en
Application filed by Cedars Sinai Medical Center filed Critical Cedars Sinai Medical Center
Publication of EP1251840A2 publication Critical patent/EP1251840A2/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/88Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/04Nitro compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid, pantothenic acid
    • A61K31/198Alpha-aminoacids, e.g. alanine, edetic acids [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/282Platinum compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/295Iron group metal compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4245Oxadiazoles
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/475Quinolines; Isoquinolines having an indole ring, e.g. yohimbine, reserpine, strychnine, vinblastine
    • 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
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/655Azo (—N=N—), diazo (=N2), azoxy (>N—O—N< or N(=O)—N<), azido (—N3) or diazoamino (—N=N—N<) compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/69Boron compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • This invention relates to the medical arts In particular, it relates to a method of enhancing the delivery of a medicant across abnormal microvasculature to a tissue requiring treatment
  • Pathologic neovascularization i e , the proliferation or development of new blood vessels, is essential for the growth and spread of primary, secondary and metastatic malignant tumors It is known that certain properties of the new capillaries and arterioles constituting the neomicrovasculature in solid tumors differ from those of normal microvasculature (J
  • Neomicrovasculature induced by angiogenic factors from malignant cells was reported to possess altered pharmacological reactivity to some vasoconstricting agents, compared with neomicrovasculature that was not induced by neoplastic cells (S P Andrade and W T Beraldo, Pharmacological reactivity of neoplastic and non-neoplastic associated neovasculature to vasoconstrictors, Int J Exp. Pathol 79(6) 425-32 [1998])
  • combretastatin A-4 was shown to cause vascular damage and occlusion selectively in the blood vessels of malignant tumors compared to normal blood vessels (G G Dark et al, Combretastatin A-4, an agent that displays potent and selective toxicity toward tumor vasculature, Cancer Res 57(10) 1829-34 [1997], D J Chaplin et al.
  • Anti-vascular approaches to solid tumour therapy evaluation of combretastatin A4 phosphate, Anticancer Res 19(1 A) 189-95 [1999])
  • Monoclonal antibodies have been directed to antigens and antigemc combinations specific to endothelial cells of pathologic neovasculature, such as vascular cell adhesion molecule
  • VCAM phosphatidylse ⁇ ne
  • PS phosphatidylse ⁇ ne
  • PSMA prostate-specific membrane antigen
  • the blood-brain barrier is a transvascular permeability barrier thought to result from the interendothelial tight junctions formed by the cerebrovascular endothelial cells of brain capillaries and arterioles in both normal and abnormal brain tissue, the maintenance of the blood-brain barrier possibly involves endogenous nitric oxide production and a cyclic GMP-dependent mechanism (Liu, S M and Sundqvist, T , Nitric oxide and cGMP regulate endothelial permeability and F-actin distribution in hydrogen peroxide-treated endothelial cells, Exp Cell Res 235(1) 238-44 [1997])
  • the blood-brain barrier protects the brain from changes in the composition of the systemic blood supply (e g , in electrolytes) or from blood-borne macromolecules, such as immunoglobulins or other
  • Neuwelt et al. used an intracarotid injection of hypertonic mannitol to osmotically disrupt the blood-brain barrier They reported that this enhanced the uptake by brain tissue of inactivated HSV-1 particles that were administered immediately afterward by intracarotid bolus injection (E A Neuwelt et al., Delivery of ultraviolet-inactivated 35S- herpesvirus across an osmotically modified blood-brain barrier, J Neurosurg 74(3) 475-79 [1991], Also, S E Doran etal, Gene expression from recombinant viral vectors in the central nervous system after blood-brain barrier disruption, Neurosurgery 36(5) 965-70 [1995], G Nilaver et al, Delivery of herpesvirus and adenovirus to nude rat intracerebral tumors after osmotic blood-brain barrier disruption, Proc Natl Acad Sci USA 92(21) 9829-33 [1995])
  • LTC sub 4 Intracarotid infusion of leukot ⁇ ene C sub 4 (LTC sub 4) selectively increases the permeability in brain tumor capillaries without affecting the permeability in normal brain capillaries
  • the effect of LTC sub 4 on brain tumor capillaries is, however, limited to small molecules and it can only slightly increase the permeability of those small molecules in abnormal brain tissue relative to normal Accordingly, LTC. sub 4 does not significantly increase the delivery of some larger water soluble molecules to brain tumors or other abnormalities
  • vasoactive nanopeptide bradykinin and agonists or polypeptide analogs thereof have been injected intravenously to increase blood-brain barrier permeability to co-administered neuropharmaceutical or diagnostic agents
  • bradykinin (B Malfroy-Camine, Method for increasing blood-brain barrier permeability by administering a bradyki m agonist of blood-brain barrier permeability, U S Patent No 5,112,596. J W Kozarich et al, Increasing blood brain barrier permeability with permeabihzer pephdes, U S Patent No 5,268,164) Intracarotid infusion of bradykinin will selectively increase permeability 2- to 12-fold in brain tumor and ischemic brain capillaries for molecules ranging in molecular weight from 100 to 70,000 Daltons (Inamura, T et al, Bradykinin selectively opens blood-tumor barrier in experimental brain tumors, J Cereb Blood Flow Metab 14(5) 862-70 [1994]) Bradykinin does not increase permeability in the normal blood brain barrier except at very high doses (Wirth, K et al, DesArg9-D-Arg[Hyp3,Thi5,D-T ⁇ c7,0 ⁇ c8
  • a method for selectively delivering to abnormal brain tissue a neuropharmaceutical agent (e g , 5-fluorouracil, cisplatin, methotrexate, or monoclonal antibodies) or a diagnostic agent (e g , technicium-99 glucoheptonate, gallium-EDTA, and ferrous magnetic or iodinated contrasting agents) employed intracarotid infusion of bradykinin, or a bradykinin analog, such as RMP-7, the bradykinin or bradykinin analog was administered approximately contemporaneously with the agent (K L Black, Method for selective opening of abnormal brain tissue capillaries, U S Patent Nos 5,527,778 and 5,434,137) Enhanced transvascular delivery of HSV-derived viral particles to malignant cells in
  • the calcium-activated potassium channel (K ⁇ ) is an important regulator of blood vessel tone (Nelson MT, Quayle JM Physiological roles and properties of potassium channels in arterial smooth muscle, Am J Physiol 268(4 Pt 1) C799-822[1995], Bang, L et al, Nitroglycerin-mediated vasorelaxatwn is modulated by endothelial calcium-activated potassium channels, Cardiovasc Res 43(3) 772-78 [1999])
  • the KCa channel is ubiquitously distributed in tissues as and subunits Its activity is triggered by depolarization and enhanced by an increase in cytosolic calcium di-cation (Ca 2+ ) A local increase in Ca 2+ is sensed by the extremely sensitive brain -subunit of the K Ca , directed towards the cytoplasm in the cell, that allows a significant potassium cation flux through these channels Under conditions when intracellular cyclic 3', 5' adenosine monophosphate (cAMP) concentration rises in vascular end
  • Minoxidil sulfate and chromakalim are reported to be activators of K ATP (A D Wickenden et al, Comparison of the effects of the K(+)-channel openers cromaka m and minoxidil sulphate on vascular smooth muscle, Br J Pharmacol, 103(1) 1148-52 [1991])
  • guanosine 3',5'-cyclic monophosphate commonly known as cyclic GMP (cGMP)
  • cGMP cyclic GMP
  • effector proteins (1) cGMP-dependent protein kinases, which mediate protein phosphorylation, (2) cGMP-gated ion channel protein kinases, which mediate cation influx across the plasma membrane, and (3) phosphodiesterases, which mediate cyclic nucleotide catabo sm
  • cGMP-dependent protein kinases which mediate protein phosphorylation
  • cGMP-gated ion channel protein kinases which mediate cation influx across the plasma membrane
  • phosphodiesterases which mediate cyclic nucleotide catabo sm
  • cGMP guanylyl cyclase
  • a nitric oxide-activated enzyme Patel, A I and Diamond, J , Activation of guanosine 3 ',5 '-cyclic monophosphate (cGMP)-dependent protein kinase in rabbit aorta by nitroglycerin and sodium nitroprusside, !
  • nitric oxide participates in the regulation of microvascular tone (Joo, F et al, Regulation of the macromolecular transport in the brain microvessels: the role of cyclic GMP, Brain Res 278(1-2) 165-74 1983])
  • glial tumors and ischemic tissue are more immunopositive for nNOS and eNOS relative to normal brain (Cai, Z et al , Prenatal hypoxia-ischemia alters expression and activity of nitric oxide synthase in young rat brain and causes learning deficits, Brain Res Bull 49(5) 359-65 [1999], Nakano, S et al, Increased brain tumor microvessel permeability after intracarotid bradykinm infusion is mediated by nitric oxide, Cancer Research, 56 4027-4031 [1996], Faraci, F M et al, Role of soluble guanylate cyclase in dilator responses of the cerebral microcirculation, Brain Res 821
  • cGMP-dependent protein kinases PKG or cGK
  • ATP-dependent phosphorylation directly or indirectly activate calcium-dependent potassium channels
  • cGMP -dependent protein kinase activates Ca-activated K channels in cerebral artery smooth muscle cells, Am J Physiol 265 [Cell Physiol 34) C299-C303 [1993], Fukao, M et al, Cyclic GMP -dependent protein kinase activates cloned BK Ca channels expressed in mammalian cells by direct phosphorylation at serine 1072, 1 Biol Chem 274(16) 10927-35 [1999], Becker, E M etal, The vasodilator-stimulated phosphoprotein (VASP): target ofYC-1 and nitric oxide effects in human and rat platelets, !
  • VASP vasodilator-stimulated phosphoprotein
  • nitric oxide can activate K ⁇ by both cGMP-dependent and cGMP-independent mechanisms (Chen, C H et al, Nitric oxide activates Ca 2* -activated Kf channels in cultured bovine adrenal chromaffin cells, Neurosci Lett 248(2) 127-29 [1998], Vaali, K et al, Relaxing effects of NO donors on guinea pig trachea in vitro are mediated by calcium-sensitive potassium channels, J Pharmacol Exp Ther 286(1) 110-14 [1998], Sobey,
  • Bradykinin is thought to increase [Ca 2 ⁇ and thus may activate K ⁇ channels While some other known activators of IC ⁇ do not act as vasodilators, for example, 1,3-dihydro-l- [2-hydroxy-5-(trifluoromethyl)phenyl]-5-(trifluoromethyl)-2H-benzimidazol-2-one (NS- 1619,
  • Kc a may play an important role in vasodilatation mediated by vasodilators, such as bradykinin, nitric oxide donors, cyclic guanosine monophosphate (cGMP), and guanylyl cyclase activators (Berg T , Koteng O , Signaling pathways in bradykinin- and nitric oxide-induced hypotension in the normotensive rat; role of Kf -channels, Br J Pharmacol ,121(6) 1113-20 [1997], Bolotina, V M et al.
  • vasodilators such as bradykinin, nitric oxide donors, cyclic guanosine monophosphate (cGMP), and guanylyl cyclase activators
  • Nitric oxide directly activates calcium-dependent potassium channels in vascular smooth muscle, Nature 368(6474) 850-3 [1994], Robertson, B E , et al, cGMP -dependent protein kinase activates Ca-activated K channels in cerebral artery smooth muscle cells, Am J Physiol 265(1 Pt 1) C299-303 [1993], Sobey, C G et al, Mechanisms of bradykinin-induced cerebral vasodilatation in rats.
  • Bradykinins action as a powerful vasodilator is disadvantageous when using bradykinin to open the blood-brain barrier to therapeutic anticancer agents Bradykinin or its analogs may adversely lower blood pressure, reduce cerebral blood flow, or contribute to brain edema in some patients (E g , A M Butt, Effect of inflammatory agents on electrical resistance across the blood-brain barrier inpial microvessels of anesthetized rats, Brain Res 696(1-2) 145-50 [1995]) In addition, bradykinin constricts smooth muscle and stimulates pain receptors Consequently,
  • the present invention relates to a method of delivering a medicant to an abnormal brain region in a mammalian subject, including a human
  • the method includes administering to the subject a potassium channel activator (i e , activator of calcium- or ATP-dependent potassium channels [K Ca or K ATP ])
  • Potassium channel activators include direct agonists (other than bradykinin or bradykinin analogs), such as NS-1619 or minoxidil
  • Potassium channel activators also include compounds that indirectly activate potassium channels, for example nitric oxide, nitric oxide donors, and other activators of guanylyl cyclase Activators of cyclic GMP-dependent protein kinase, which activates calcium-dependent potassium channels, are also included
  • the potassium channel activator is administered to the subject under conditions and in an amount sufficient to increase the permeability to the medicant of a capillary or arteriole delivering blood to cells of the abnormal brain region in the subject Simultaneously or
  • the present invention also relates to a method of delivering a medicant to a malignant tumor in the brain or anywhere in the body of a mammalian subject
  • the method involves administering to the subject a potassium channel activator, such as a potassium channel agonist other than bradykinin or a bradykinin analog, under conditions and in an amount sufficient to increase the permeability to the medicant of a capillary or arteriole delivering blood to cells of the malignant tumor in the subject Simultaneously or substantially simultaneously with the potassium channel activator the medicant is administered to the subject, and it is delivered selectively to the malignant cells compared to non-malignant cells by virtue of the potassium channel activator
  • the inventive method is useful in treating any kind of malignant tumor by increasing the selectivity of drug delivery to neoplastic tissue, thereby minimizing damage to non-malignant tissue from medicants, including cytotoxic chemotherapeutic agents, and focusing the therapeutic or diagnostic action of the agents
  • this invention also directed to a method of treating a
  • the present invention also relates to a pharmaceutical composition that comprises a combination of a potassium channel activator, other than bradykinin or a bradykinin analog, formulated in a pharmaceutically acceptable solution together with a medicant for delivery by intravascular infusion or bolus injection into a mammal, such as a human
  • a pharmaceutical composition that comprises a combination of a potassium channel activator, other than bradykinin or a bradykinin analog, formulated in a pharmaceutically acceptable solution together with a medicant for delivery by intravascular infusion or bolus injection into a mammal, such as a human
  • a pharmaceutical composition is useful in practicing the inventive methods
  • the invention also relates to a kit for enhancing the delivery of a medicant to an abnormal brain region and/or to a malignant tumor
  • IBTX iberiotoxin
  • ODQ a selective inhibitor of soluble guanylyl cyclase, lH-[l,2,4]oxadiazolo[4,3-a]quinozalin-l-one
  • Figure 2 shows the enhancing effect of the nitric oxide donor diethylamine-NONOate (DEA/NO) on blood-tumor barrier permeability to Evans blue stain in malignant RG2 glioma tissue in Wistar rats
  • Figure 2A shows a brain section from a rat treated with phosphate buffered saline (PBS only control)
  • Figure 2B shows a brain section from a DEA/NO-treated rat
  • Figure 4 shows transendothelial vesicular transport in a RG2 tumor capillary RG2 tumor-bearing brain sections from Wistar rats were prepared after a 15-minute intracarotid infusion with PBS (0 8 mL)(Figure 4A, 54,000x magnification), bradykinin (10 ⁇ g min "1 kg "x )( Figure 4B, 87,000x magnification), or NS-1619 (5 3 ⁇ g min "1 kg “1 , Figure 4C, 87,000x magnification)
  • TEM analysis of the tumor center section shows that K Ca activators bradykinin and NS-1619 increase pinocytotic transport vesicles in the endothelial cytoplasm
  • Figure 5 shows that a potassium channel activator (bradykinin) increases transport of horse radish peroxidase (HRP) from the tumor capillary lumen through endothelial pinocytotic vesicles
  • HRP horse radish peroxidase
  • FIG 7 shows an increase in the unidirectional transport constant caused by the K ATP activator minoxidil sulfate and the K Ca activator bradykinin
  • Figure 8 shows that bradykinin-induced increase in K, was not attenuated by glibenclamide, while IBTX significantly decreased the effect caused by bradykinin
  • Figure 9 shows that the minoxidil sulfate-induced increase in K, was attenuated by glibenclamide, while IBTX failed to block the effect
  • Figure 10 illustrates increased micro vascular permeability in an ischemic brain region as a result of treatment with a potassium channel activator K, is shown for bradykinin-treated (black bars) and control animals (gray bars) in paired histograms, tissues tested were (left to right) caudate putamen, cortex, caudate putamen contralateral side, and cortex contralateral side
  • Figure 10A shows that blood-brain barrier permeability after one hour of ischemia was not affected by bradykinin treatment compared to the vehicle-treated group (MCA occlusion for
  • Figure 10B shows that bradykinin significantly increased permeability in ischemic infarct penumbra after 2 hours of MCA occlusion, followed by 1 hour of reperfusion (Inset autoradiograph)
  • Figure 11 shows that potassium activators enhance permeability of microvasculature in ischemic brain tissue No change in blood-brain barrier permeability at the infarct brain tissue
  • Figure 12A shows the enhancing effect of NS-1619 on blood-tumor barrier permeability to [ 14 C] -aminoisobutyric acid (AIB) tracer (left) compared to the effect on blood-brain barrier permeability in normal brain tissue adjacent (middle) and contralateral (right) to malignant RG2 glioma tissue in Wistar rats
  • Figure 12B shows the enhancing effect of minoxidil sulfate on blood-tumor barrier permeability to [ 14 C]ATB tracer (left) compared to the effect on blood-brain barrier permeability in normal brain tissue adjacent (middle) and contralateral (right) to malignant RG2 ghoma tissue in Wistar rats
  • Figure 14 shows specific inhibition by iberiotoxin (IBTX, 2 3 ⁇ g kg “1 min “1 ) of the permeability increasing effect of NS-1619 (26 5 ⁇ g kg “1 min “1 )
  • the K was determined in RG2 tumor-bearing Wistar rats using [ 14 C]AIB with NS-1619 (26 5 ⁇ g kg "1 min "1 ) with or without
  • Figure 15 shows intense over-expression of K Ca as indicated by anti-K Ca immunostain of glioma tissue (Fig 15B), compared to normal contralateral brain tissue (Fig 15 A) Magnification is lOOx
  • Figure 16 shows intense over-expression of K ATP as indicated by anti-K ATP (anti-Kir 6 2) immunostain of RG2 (Fig 16C) or C6 (Fig 16D) glioma tissue from tumor centers, compared to normal brain tissue (Fig 16 A) and RG2 tumor periphery (Fig 16B) Magnification is lOOx
  • inventive methods are useful for selectively delivering a medicant to abnormal brain regions and/or malignant tumors in mammalian subjects
  • the methods involve administering to the mammalian subject a potassium channel activator, other than bradykinin or a bradykinin analog, under conditions and in an amount sufficient to increase the permeability to the medicant of a capillary or arteriole delivering blood to cells of the abnormal brain region and/or to malignant cells of a malignant tumor present in the subject
  • the increase in permeability ranges from at least 2-to 6-fold, compared to controls without the administration of a potassium channel activator.
  • the relative increase in permeability tends to be greater for large molecular weight medicants (e g., about 10,000 to 250,000 Daltons) than for smaller molecular weight substances (e g , about 50-200 Daltons)
  • the abnormal brain regions include regions of brain tissue physiologically directly affected by a physical or biochemical injury, for example, Alzheimers disease, Parkinsonism, trauma, infection, stroke, brain ischemia, or regions of neoplastic growth within the brain, such as benign or malignant brain tumor tissues
  • the present invention is also useful for selectively delivering a medicant to a malignant tumor in the brain or to a tumor elsewhere in the body of a mammalian subject.
  • the inventive technology is useful in the treatment of all kinds of solid malignant tumors, including gliomas, glioblastomas, oligodendrogliomas, astrocytomas, ependymomas, primitive neuroectodermal tumors, atypical meningiomas, malignant meningiomas, neuroblastomas, sarcomas, melanomas, lymphomas, or carcinomas.
  • the tumor to be treated can be contained in the skull, brain, spine, thorax, lung, peritoneum, prostate, ovary, uterus, breast, stomach, liver, bowel, colon, rectum, bone, lymphatic system, skin, or in any other organ or tissue of the subject.
  • inventive methods are useful in treating any mammal, including a human, non-human primate, canine, feline, bovine, porcine or ovine mammal, as well as in a small mammal such as a mouse, rat, gerbil, hamster, or rabbit.
  • the potassium channel activator is an activator of either a calcium-activated potassium channel (K Ca ) of any conductance level, whether of large, intermediate, or small conductance, or of an ATP-sensitive potassium channel (K ATP ). Included are direct agonists of K Ca , such as 1 ,3-dihydro- 1 -[2-hydroxy-5-(trifluoromethyl)phenyl]-5-(trifluoromethyl)-
  • K Ca calcium-activated potassium channel
  • K ATP ATP-sensitive potassium channel
  • potassium channel activators include compounds that indirectly activate potassium channels, for example activators of soluble guanylyl (i.e., guanylate) cyclase, such as nitric oxide, nitric oxide donors, metalloporphyrins (e.g., zinc or tin protoporphyrin IX), YC-1 (a benzyl indazole derivative), or guanylyl cyclase activating proteins (GCAPs).
  • activators of soluble guanylyl (i.e., guanylate) cyclase such as nitric oxide, nitric oxide donors, metalloporphyrins (e.g., zinc or tin protoporphyrin IX), YC-1 (a benzyl indazole derivative), or guanylyl cyclase activating proteins (GCAPs).
  • activators of soluble guanylyl (i.e., guanylate) cyclase such as
  • minoxidil 2,4-diamino-6-piperidino pyramidine-3 -oxide
  • pinacidil ((+/-)-N-cyano-N'-4-pyridinyl-N"-(l,2,2-trimethyl propyl)- guanidine
  • a preferred potassium channel activator is nitric oxide gas, which is fully permeable across biological membranes.
  • Inhalable nitric oxide gas can be administered to the subject by mask in a controlled gas mixture as is known in the art. (E.g., Kieler- Jensen, N. et al.
  • the concentration in the gas mixture of nitric oxide (NO) is preferably about 1 to 100 ppm NO, more preferably about 4 to 80 ppm NO, and most preferably about 20 to 40 ppm
  • Nitric oxide donors are compounds that produce NO-related physiological activity when applied to biological systems
  • NO-donors can mimic an endogenous NO-related response or substitute for an endogenous NO deficiency
  • NO-donors can mimic an endogenous NO-related response or substitute for an endogenous NO deficiency
  • NO-donors can mimic an endogenous NO-related response or substitute for an endogenous NO deficiency
  • NO-donors can mimic an endogenous NO-related response or substitute for an endogenous NO deficiency
  • NO + , NO 0 , or NO " NO
  • the redox state of NO makes a substantial difference to the NO donors reactivity towards other biomolecules, the profile of by-products, and the bioresponse (Feelisch, M , The use of nitric oxide donors in pharmacological studies, Naunyn-Schmiedebergs Arch Pharmacol 358 113-22 [1998])
  • Some classes of NO donors require enzymatic cat
  • nitric oxide donors include organic nitrate compounds, which are nitric acid esters of mono- and polyhydric alcohols Typically, these have low water solubility, and stock solutions are prepared in ethanol or dimethyl sulfoxide (DMSO) Examples are glyceryl trinitrate (GTN) or nitroglycerin (NTG), pentaerythrityl tetranitrate (PETN). isosorbide dinitrate (ISDN), and isosorbide 5-mononitrate (IS-5-N) Administration of organic nitrates can be done intravenously, intraperitoneally, intramuscularly, transdermally, or in the case of PETN, ISDN, NTG, and IS-5-N, orally
  • GTN glyceryl trinitrate
  • NTG nitroglycerin
  • PETN pentaerythrityl tetranitrate
  • ISDN isosorbide dinitrate
  • IS-5-N isosorbide 5-mononitrate
  • S-nitrosothiol compounds including S-nitroso-N-ace1yl-D,L-per ⁇ ciUamine (SNAP), S-nitrosoglutathione (SNOG), S-nitrosoalbumin,
  • S-nitrosocysteine S-nitrosothiol compounds are particularly light-sensitive, but stock solutions kept on ice and in the dark are stable for several hours, and chelators such as EDTA can be added to stock solutions to enhance stability Administration is preferably by an intravenous or intra-arterial delivery route
  • nitric oxide donors include sydnonimine compounds, such as molsidomine (N-ethoxycarbonyl-3-morpholino-sydnonimine), linsidomine (SLN-1, 3 -morpholino-sydnonimine or 3 -morpholinylsydnoneimine or
  • a most preferred nitric oxide donor is chosen from among the so-called NONOate compounds
  • the NONOates are adducts of NO with nucleophilic residues (X " ), such as an amine or sulfite group, in which an NO dimer is bound to the nucleophilic residue via a nitrogen atom to form a functional group of the structure X[-N(O)NO] "
  • X nucleophilic residues
  • the NONOates typically release NO at predictable rates largely unaffected by biological reactants, and NO release is thought to be by acid-catalyzed dissociation with the regeneration of X " and NO This property is particularly useful in accordance with the inventive methods of selectively delivering a medicant, because abnormal brain regions and malignant tumors can typically be relatively hypoxic and possess a relatively low ambient pH (e g , pH 6 5-7 0), which concentrates release of NO selectively in the microvasculature of the abnormal brain region or malignant tumor
  • NONOates include most preferably diethylamine-NONOate (DEA/NO, N-Ethylethanamine l , l -Diethyl-2-hydroxy-2-nitrosohydrazine (1 1 ) or l-[N,N-diethylamino]diazen-l-ium-l,2-diolate)
  • Other preferred NONOates include diethylene triamine-NONOate(DETA/NO, 2,2'-Hydroxynitrosohydrazino]bis-ethanamine), spermine-NONOate (SPER/NO, N-(4-[-l-(3-Aminopropyl)-2-hydroxy-2-nitrosohydrazino] butyl)-l,3-propanediamine), propylamino-propylamine-NONOate (PAPA/NO, 3-(2-Hydroxy-2-nitroso-l-propylhydrazino)-l-propanamine or (Z)-l-[N-
  • NONOates are preferably administered intravenously or intra-arterially
  • Nitric oxide donors have different potencies (Ferraro, R et al, Comparative effects of several nitric oxide donors on intracellular cyclic GMP levels in bovine chromaffin cells: correlation with nitric oxide production, Br J Pharmacol 127(3) 779-87 [1999])
  • DEA/NO is among the most potent nitric oxide donors, with a half-life of about 2 to 4 minutes, less potent are PAPA/NO (t 1/2 about 15 minutes), SPER/NO (t 1/2 about 34-40 minutes), even less potent
  • Stock solutions of NO donors are preferably made up freshly before use (at the appropriate pH for each particular NO donor), chilled on ice, and protected from light (e g , by the use of darkened glass vials wrapped in aluminum foil), although organic nitrates can be stored for months to years if the vial is properly sealed
  • final dilutions are prepared in pharmaceutically acceptable buffer and the final pH of the NO donor-containing buffer is checked for physiological suitability, especially when strongly acidic (e g , hydrochlo ⁇ de salts) or alkaline (e g , NONOates) stock solutions are used
  • the product of NO exposure time and NO concentration largely determines the quality and magnitude of the biological response to exogenously supplied NO Short-lived NO donors, such as DEA/NO, are most preferably admimstered by continuous infusion rather than by bolus to avoid delivering only a short burst of NO
  • potassium channel activators include activators of any endogenous species of cyclic GMP-dependent protein kinase (PKG or cGK), that activates a potassium channel directly (e g , by directly phosphorylating K Ca ) or indirectly (e g , by phosphorylating another regulatory protein that directly modulates Kc a activity) Included are activators of cGK I, cGK II, or other isoforms of cGMP-dependent protein kinase (e g , Smolenski, A et al. [1998])
  • PKG activators include, but are not limited to, octobromo-cyclic GMP (8Br-cGMP) and dibutyryl cyclic GMP
  • useful potassium channel activators are pharmaceutically acceptable molecular conjugates or salt forms that still have activity as potassium channel activators as defined herein
  • An example is minoxidil sulfate, but other pharmaceutically acceptable salts comprise anions other than sulfate, such as chloride, carbonate, bicarbonate, nitrate, or the like
  • Other embodiments of pharmaceutically acceptable salts contain cations, such as sodium, potassium, magnesium, calcium, ammonium, or the like
  • useful potassium channel activators are hydrochloride salts
  • the potassium channel activator employed in the inventive methods is one other than the vasodilator bradykinin (Arg-Pro-Pro-Gly-Phe-Ser-Pro-Phe-Arg), or a polypeptide bradykinin analog, such as receptor mediated permeabilizer (RMP)-7 or A7 (e g , Kozarich et ⁇ /, U S Patent No 5,268,164 and PCT Application No WO 92/18529)
  • RMP receptor mediated permeabilizer
  • Other analogs of bradykinin include related peptide structures which exhibit the same properties as bradykinin but have modified amino acids or peptide extensions on either terminal end of the peptide
  • Examples of bradykinin analogs include [phe sup 8 (CH sub 2 NH) Arg sup 9 ]-bradykinin, Nacetyl [phe sup.8 (CH sub 2 ⁇ NH ⁇ Arg sup 9 ] bradykinin and desArg9-bradykinin
  • an effective amount of YC-1 for humans is about 15 to about 45 micrograms per kg body mass, and for nitric oxide donors generally about 15 to about 45 micrograms per kg body mass
  • the optimal amount for each individual for any particular potassium channel activator can be determined by routine means involving close physiological monitoring over the delivery period
  • the dose can be administered in a bolus injection, but is preferably administered by infusion over a period of one to thirty minutes, and most preferably during a period of one to fifteen minutes
  • a dose rate of about 0 75 to about 100 ⁇ g kg "1 min "1 is most suitable.
  • the medicant is administered simultaneously or substantially simultaneously with the potassium channel activator, and the medicant is delivered by the blood stream selectively to the abnormal brain region and/or to the malignant cells compared to normal brain tissue or non-malignant cells
  • Simultaneously means that the medicant is administered contemporaneously or concurrently with the potassium channel activator
  • Substantially simultaneously means that the medicant is administered within about one hour after the potassium channel activator is last administered, preferably within about 30 minutes after, and most preferably, is administered simultaneously with the potassium channel activator
  • substantially simultaneously means that the medicant is administered within about 30 minutes before, and preferably within about 15 minutes before the potassium channel activator is first administered
  • the methods of delivering a medicant to an abnormal brain region and/or to a malignant tumor in a mammalian subject are effective in selectively delivering any medicant across the microvascular of an abnormal brain region and/or malignant tumor
  • the medicant is a drug, i e , a chemotherapeutic agent
  • chemotherapeutic agents including therapeutic cytotoxic agents (e g , cisplatin, carboplatin, methotrexate, 5-fluorouracil, amphotericin), naked DNA expression vectors, therapeutic proteins, therapeutic oligonucleotides or nucleotide analogs, interferons, cytokines, or cytokine agonists or antagonists, adrenergic agents, anticonvulsants, anti-trauma agents, or any neuropharmaceutical agent used to treat or prevent an injury or disorder of the brain
  • Chemotherapeutic agents also include ischemia-protective drugs such as N-methyl-D-aspartate (NMD A)
  • TGF transforming growth factors
  • the medicant also includes anticancer chemotherapeutic agents
  • anticancer chemotherapeutic agents are cytotoxic agents, such as 5-fluorouracil, cisplatin, carboplatin, methotrexate, daunorubicin, doxorubicin, vincristine, vinblastine, or a cytotoxic alkylating agent, such as, but not limited to, busulfan (1,4-butanediol dimethanesulphonate, Myleran, Glaxo
  • the anticancer chemotherapeutic agents are particularly useful in practicing the method of selectively delivering a medicant to a malignant tumor, in the brain or in any other tissue of the body, and in the method of treating a malignant tumor in a human subject
  • Medicants also include any therapeutic viral particle, for example an adenovirus-derived or herpes simplex virus (HSV)-derived viral vector for delivering genetic material to a cellular target in vivo
  • Medicants also include diagnostic agents, such as imaging or contrast agents, for example, radioactively labeled substances (e g , [ 99 Tc]-glucoheptonate), gallium-labeled imaging agents (e.g , gallium-EDTA), ferrous magnetic, fluorescent, luminescent, or iodinated contrast agents
  • any of the afore-mentioned medicants having anticancer activity can also be used in practicing the method of selectively delivering a medicant to a malignant tumor or the method of treating a malignant tumor in a human
  • the medicant can be a molecular substance having a molecular weight between about 50 Daltons and about 250 kD Or it can be a particle, such a s a viral particle, having a diameter between about 50 to 250 nanometers
  • the medicant can be, but is preferably not, an agent that is highly lipid soluble and thus inherently able to penetrate cell membranes, for example nitrosourea.
  • the amount of medicant that is employed is within a conventional dose range for each medicant, however by practicing the inventive method, the increased transvascular permeability afforded can provide a greater selective therapeutic effect per dose or permit a lower effective dose to be used, if desired, for example to lessen systemic toxic effects from anti-cancer medication in a particular subject.
  • the medicant is administered by any appropriate method that can deliver it to the blood stream. Typically, this is by intravenous, intramuscular, or intra-arterial (including intracarotid) injection or infusion. However, for some applications other acceptable delivery routes can be used as long as the dose of medicant enters the blood stream substantially simultaneously with the potassium channel activator.
  • Examples include ingestion (e.g., of a powder, suspension, solution, emulsion, tablet, capsule or caplet); subcutaneous injection; stereotactic injection; or transdermal or transmucosal delivery by adhesive patch, suppository or gel for delivery through the skin, mucosa or epithelium of the mouth including the sublingual epithelium, the rectum, or the vaginal epithelium.
  • ingestion e.g., of a powder, suspension, solution, emulsion, tablet, capsule or caplet
  • subcutaneous injection e.g., of a powder, suspension, solution, emulsion, tablet, capsule or caplet
  • stereotactic injection e.g., stereotactic injection
  • transdermal or transmucosal delivery by adhesive patch, suppository or gel for delivery through the skin, mucosa or epithelium of the mouth including the sublingual epithelium, the rectum, or the vaginal epithelium.
  • the medicant is administered together with the potassium channel activator in a pharmaceutical composition of the present invention.
  • the inventive pharmaceutical composition comprises a combination of a potassium channel activator, other than bradykinin or a bradykinin analog, as described above, formulated in a pharmaceutically acceptable solution together with a medicant, as described above, for delivery by intravascular infusion or bolus injection into a mammal, such as a human.
  • the solution is thus suitably balanced, osmotically (e.g., about 0.15 M saline) and with respect to pH, typically between pH 7.2 and 7.5; preferably the solution further comprises a buffer, such as a phosphate buffer (e.g., in a phosphate buffered saline solution).
  • the solution is formulated to deliver a dose rate of about 0.075 to 1500 micrograms of potassium channel activator per kilogram body mass in a pharmaceutically acceptable fluid volume over a maximum of about thirty minutes.
  • the solution is preferably formulated to deliver a dose rate of about 0.075 to 150 micrograms of potassium channel activator per kilogram body mass in a pharmaceutically acceptable fluid volume over a period of up to about thirty minutes
  • the invention also relates to a kit for enhancing the delivery of a medicant to an abnormal brain region and/or to a malignant tumor
  • the kit is an assemblage of materials or components, including a potassium channel activator, other than bradykinin or a bradykinin analog, as described above
  • the kit contains instructions for using the potassium channel activator to enhance the permeability of abnormal microvascular, including neomicrovasculature, to a medicant in general, or alternatively, to a particular medicant
  • the kit also contains other components, such as a particular medicant in any pharmaceutically acceptable formulation, or paraphernalia for injection or infusion, for example syringes, infusion lines, clamps, and/or infusion bags/bottles, which can contain a pharmaceutically acceptable infusible formulation of the potassium channel activator with or without a particular medicant also contained therein
  • the materials or components assembled in the kit can be provided to the practitioner stored in any convenient and suitable ways that preserve their operability and utility
  • the components
  • RG2 Malignant Cell Line and Tumor Implantation
  • RG2 cells derived from a rat glioma are kept frozen until use, then are thawed and maintained in a monolayer culture in F12 medium with 10% calf serum In some experiments C6 glioma cells were used
  • the Wistar rats (approximately 140-160 g body weight) were anesthetized with intra-peritoneal ketamine (50 mg/kg), and glial cells (1 x 10 5 ) were implanted into the right hemisphere, but not the contralateral hemisphere, by intracerebral injection suspended in 5 ⁇ L F12 medium (1-2% methylcellulose) by a Hamilton syringe
  • the implantation coordinates were 3 -mm lateral to the bregma and 4 5 mm
  • Nitric oxide donors included sodium 2-(N,N-diethylamino)-diazenolate-2-oxide (DEA/NO), a nitric oxide-donor with a half-life of 2 1 min or PAPA/NO ([Z]-l-[N-[3-Amino propyo]-N-[N-propyo amino] diazen-l-ium-l,2-diolate, (Alexis Corp ), a nitric oxide-donor with a half-life of 15 minutes DEA/NO or PAPA/NO were dissolved in PBS and administered to RG2 glioma-bearing rats to determine permeability (K,) of [ 14 C]-AIB without affecting the physiological parameters [ 14 C]-AIB was administered intravenously as a
  • K Unidirectional Transport Constant
  • K The unidirectional transfer constant K, for [ 14 C] ⁇ - aminoisobutyric acid (AIB) was measured in normal tissue and tumor tissue as an indicator of permeability across the blood-tumor and blood-brain barriers Quantitative autoradiography was used to obtain K, values ( ⁇ L g "1 min "1 ) The initial rate for blood-to-brain transfer was calculated using a previously described equation (Ohno, K, et al, Lower limits of cerebrovascular permeability to nonelectrolytes in the conscious rat, Am J Physiol 235(3) H299-307, [1978], Inamura, T , et al. , Bradykinin selectively opens blood-tumor barrier in experimental brain tumors, J.
  • NS-1619 was dissolved in 25% ethanol and diluted with PBS to obtain various concentrations for infusion NS-1619 was administered by intracarotid infusion (dose rates 0, 13, 26 5, 53, 80, 100 and 110 ⁇ g kg "1 min "1 , all at 53 3 ⁇ L/min) to RG2 glioma-bearing rats to determine a dose that produces increased permeability (K,) of [ 14 C]-AIB, which was administered intravenously K, was determined as described above Physiological parameters were monitored during the experiments
  • the unidirectional transport constant (K,) for [ 14 C]-AIB to the tumor center and tissue surrounding tumor was determined after intracarotid infusion of bradykinin (10 ⁇ g min "1 kg "1 for 15 min) or minoxidil sulfate (26 6 ⁇ g min "1 kg "1 for 15 min) or co-infusion of bradykinin with minoxidil sulfate for 15 min Physiological parameters were monitored during the experiments
  • the selective K ATP inhibitor, glibenclamide 13 3 ⁇ g min "1 kg "1 for 15 min was used to determine whether inhibition of K ATP channel blocks bradykinin-inducible or minoxidil sulfate-inducible increases in blood-tumor barrier permeability
  • RG2 glioma cells were implanted into the right hemisphere of rat brains as described above Seven days after implantation, regional permeability study was performed by intracarotid infusion of octobromo-cyclic GMP (8Br-cGMP, 16 7 ⁇ g kg "1 min "1 ), an activator of cyclic GMP-dependent protein kinase (PKG), without or with PKG inhibitors
  • Rat brain tissue was prepared for TEM analysis by the following procedures After experiments involving intracarotid infusion of potassium channel activators, inhibitors, horse radish peroxidase (HRP), and/or vehicle control buffers, rats were infusion-fixed from the heart first with PBS (50-100 mL) followed with 1 0% glutaraldehyde (250 mL) The brain tissue was cut cronally to expose the tumor region, and the region of interest was selected, sliced into small pieces of about 1 mm thickness, and was immersion-fixed in 1 0% glutaraldehyde, at 4°C for 2 hours The sample was rinsed in 5% sucrose / 0 1 M PBS at 4°C, overnight, with continuous shaking and changes of the solution The samples then were immersion-fixed with 1% OsO4 at 4°C for 2 hours with continuous shaking The sample was then dehydrated with an increasing concentration (50-100%) of ethanol at 4°C for 15 minutes with constant shaking and frequent changes of solution Samples
  • the density of the vesicles was determined for three vessels selected from each rat (each treatment group contained 5-6 rats) by taking from each blood vessel four electron micrographs as a test zone at high magnification
  • the test zones were selected randomly, such as at 3, 6, 9, and 12 o'clock on the EM screen
  • the areas of the test zones were measured, and the number of vesicles was counted by a person who was not aware of the background of the micrographs
  • Vesicular density was expressed as the number of vesicles per square micrometer of cytoplasm
  • the proportion of the total vesicular area to the cytoplasmic area was also determined using the same micrographs total vesicular area was measured and the proportion to the cytoplasm which included those vesicles was calculated and expressed as percentage
  • the mean diameter of the vesicles was also calculated
  • Temporary Middle Cerebral Artery fMCA Temporary Middle Cerebral Artery fMCA occlusion MCA occlusion was carried out as described by Liu, Y et al, The time course of glucose metabolism in rat cerebral ischemia with middle cerebral artery occlusion-reperfuswn model and the effect ofMK-80, Neurological Research 18 (6) 505-508 (1996) with slight modifications Briefly, the right MCA was occluded temporarily with a silicone rubber cylinder inserted from the bifurcation of the intracarotid artery after ligation of ipsilateral common and external carotid arteries The cylinder is made of 4-0 nylon 17 mm long surgical thread, coated with silicone mixed with a hardener to grade the diameter of the distal 5 mm to 0 25-0 30 mm The thread was inserted into the intracarotid artery through the external carotid artery near the bifurcation and ligated at the point of insertion The silicone rubber cylinder
  • Evans blue dye was injected intravenously after 50 minutes of reperfusion (five minutes after commencing administration of potassium channel activator and/or inhibitor) Ten minutes after injection of Evans blue, the rat was perfused with 200 mL PBS through the heart to wash off excess Evans blue dye from the brain microvessels
  • Nitric oxide donors also increased the permeability of the blood-tumor barrier
  • Figure 2 shows that RG2 glioma tissue was stained in vivo by Evans blue stain (MW 960 82) in DEA/NO-treated Wistar rats (Figure 2B) compared to PBS-treated controls ( Figure 2 A) These results were comparable to the results obtained by in vivo staining by Evans blue using NS-1619 or YC-1 (data not shown).
  • IBTX alone at the dose investigated did not affect the brain-tumor barrier permeability of abnormal capillaries.
  • Kc a channel protein was immunolocalized using a specific antibody as described above.
  • HRP horse radish peroxidase
  • Tissue 3 Infusion 33 Density of the vesicles Total area of the vesicles (%) c (number/m2) b
  • BG basal ganglia
  • PBS phosphate buffer saline vehicle control
  • BK PBS + bradykinin
  • b Values are means SD
  • n number of rats
  • Q Values are means SD
  • n number of capillaries d
  • Potassium channel activators increase permeability in abnormal brain capillaries of ischemic brain region.
  • Figure 10A shows that the permeability of brain microvasculature was not responsive to a potassium channel activator after only one hour of MCA occlusion. This indicates that properties of the microvasculature are changed by prolonged ischemia as the tissue becomes increasingly abnormal.

Abstract

Disclosed are methods of selectively delivering a medicant to an abnormal brain region and/or to a malignant tumor in a mammalian subject, including a human. A medicant is administered simultaneously or substantially simultaneously with a calcium- or ATP-dependent potassium channel [KCa or KATP] activator (other than bradykinin or a bradykinin analog), such as a direct potassium channel agonist or an indirect potassium channel activator, such as an activator of soluble guanylyl cyclase (e.g., nitric oxide or a nitric oxide donor) or an activator of cyclic GMP-dependent protein kinase, whereby the medicant is delivered selectively to the cells of the abnormal brain region and/or to the tumor, compared to normal tissues. Thus, among the disclosures is a method of treating a malignant tumor in a human subject. Also disclosed are pharmaceutical compositions that combine a potassium channel activator together with a medicant and a kit for enhancing the delivery of a medicant to an abnormal brain region and/or to a malignant tumor.

Description

METHOD FOR USING POTASSIUM CHANNEL ACTIVATION
FOR DELIVERING A MEDICANT TO AN ABNORMAL BRAIN REGION
AND/OR A MALIGNANT TUMOR
BACKGROUND OF THE INVENTION
Throughout the application various publications are referenced in parentheses The disclosures of these publications in their entireties are hereby incorporated by reference in the application in order to more fully describe the state of the art to which this invention pertains
1 THE FIELD OF THE INVENTION
This invention relates to the medical arts In particular, it relates to a method of enhancing the delivery of a medicant across abnormal microvasculature to a tissue requiring treatment
2 DISCUSSION OF THE RELATED ART
Pathologic neovascularization, i e , the proliferation or development of new blood vessels, is essential for the growth and spread of primary, secondary and metastatic malignant tumors It is known that certain properties of the new capillaries and arterioles constituting the neomicrovasculature in solid tumors differ from those of normal microvasculature (J
Denekamp et al, Vasculature and microenvironmental gradients: the missing links in novel approaches to cancer therapy? ', Adv Enzyme Regul 38 281-99 [1998]) Neomicrovasculature induced by angiogenic factors from malignant cells was reported to possess altered pharmacological reactivity to some vasoconstricting agents, compared with neomicrovasculature that was not induced by neoplastic cells (S P Andrade and W T Beraldo, Pharmacological reactivity of neoplastic and non-neoplastic associated neovasculature to vasoconstrictors, Int J Exp. Pathol 79(6) 425-32 [1998])
A number of proposed cancer treatments have been based on differences between neomicrovasculature and normal microvasculature For example, combretastatin A-4 was shown to cause vascular damage and occlusion selectively in the blood vessels of malignant tumors compared to normal blood vessels (G G Dark et al, Combretastatin A-4, an agent that displays potent and selective toxicity toward tumor vasculature, Cancer Res 57(10) 1829-34 [1997], D J Chaplin et al. , Anti-vascular approaches to solid tumour therapy: evaluation of combretastatin A4 phosphate, Anticancer Res 19(1 A) 189-95 [1999]) Monoclonal antibodies have been directed to antigens and antigemc combinations specific to endothelial cells of pathologic neovasculature, such as vascular cell adhesion molecule
(VCAM)-l, phosphatidylseπne (PS), the glycoprotein endosialin, and prostate-specific membrane antigen (PSMA), with the aim of selectively inducing thrombosis in neovasculature (E g , S Ran et al. , Infarcation of solid Hodgkins tumors in mice by antibody-directed targeting of tissue factor to tumor vasculature, Cancer Res 58(20) 4646-53 [1998], I Ohizumi et al, Antibody-based therapy targeting tumor vascular endothelial cells suppresses solid tumor growth in rats, Biochem Biophys Res Commun 236(2) 493-96 [1997], S S Chang et al, Fve different antiprostate-specific membrane antigen (PSMA) antibodies confirm PSMA expression in tumor-associated neovasculature, Cancer Res 59(13) 3192-98 [1999], W J Rettig et al, Identification of endosialin, a cell surface glycoprotein of vascular endothelial cells in human cancer, Proc Natl Acad Sci USA 89(22) 10832-36 [1992]) But taken alone, shutting down blood flow through the neomicrovasculature to malignant tumors may not necessarily result in stopping tumor growth, because actively proliferating populations of neoplastic cells at the periphery of solid tumors may have access to blood supplied by normal microvasculature (E g , D J Chaplin et al. [1999]) Consequently, other conventional and novel therapeutic modalities will continue to be of value in the treatment of malignant, solid tumors However, the efficacy of novel therapeutic agents, including cytotoxic chemotherapeutic agents, monoclonal antibodies, cytokines, effector cells, and viral particles has been limited by their ability to reach their targets in vivo in adequate quantities (E g , R K Jain, Vascular and interstitial barriers to delivery of therapeutic agents in tumors, Cancer Metastasis Rev 9(3) 253-66 [1990]) An important limiting factor is the low permeability to macromolecules and viral particles of neomicrovasculature supplying the tumors
This problem of microvascular permeability is especially acute with respect to malignant tumors of the central nervous system These malignancies are usually fatal, despite recent advances in the areas of neurosurgical techniques, chemotherapy and radiotherapy In particular, there are no standard therapeutic modalities that can substantially alter the prognosis for patients with malignant tumors of the brain, cranium, and spinal cord For example, high mortality rates persist for patients diagnosed with malignant medulloblastomas, malignant meningiomas, malignant neurofibrosarcomas and malignant gliomas, which are characterized by infiltrative tumor cells throughout the brain Although intracranial tumor masses can be debulked surgically, treated with palliative radiation therapy and chemotherapy, the survival associated with intracranial tumors, for example, a glioblastoma, is typically measured in months The development of new therapeutic modalities against solid brain tumors largely depends on transvascular delivery of the potential therapeutic agent
Transvascular delivery of chemotherapeutic agents and viral particles to tumor cells or other abnormal brain tissue is hampered by the blood-brain barrier, particularly the blood-tumor barrier found in brain tumors The blood-brain barrier is a transvascular permeability barrier thought to result from the interendothelial tight junctions formed by the cerebrovascular endothelial cells of brain capillaries and arterioles in both normal and abnormal brain tissue, the maintenance of the blood-brain barrier possibly involves endogenous nitric oxide production and a cyclic GMP-dependent mechanism (Liu, S M and Sundqvist, T , Nitric oxide and cGMP regulate endothelial permeability and F-actin distribution in hydrogen peroxide-treated endothelial cells, Exp Cell Res 235(1) 238-44 [1997]) The blood-brain barrier protects the brain from changes in the composition of the systemic blood supply (e g , in electrolytes) or from blood-borne macromolecules, such as immunoglobulins or other polypeptides, and prevents the transvascular delivery of many exogenously supplied pharmaceutical agents to brain tissues
The treatment of brain tissue abnormalities, such as tumors, often involves the use of pharmaceutical agents with a significant toxicity of their own, making it highly desirable to be able to preferentially direct such agents to the abnormal or malignant tissue While, there has been a great deal of interest in developing techniques which are capable of opening the blood-brain barrier to allow transport of pharmaceutical agents to the brain Few of these methods are capable of selectively opening the blood-brain barrier only in the abnormal brain tissue while leaving the blood-brain barrier in the normal brain tissue intact
For example, Neuwelt et al. used an intracarotid injection of hypertonic mannitol to osmotically disrupt the blood-brain barrier They reported that this enhanced the uptake by brain tissue of inactivated HSV-1 particles that were administered immediately afterward by intracarotid bolus injection (E A Neuwelt et al., Delivery of ultraviolet-inactivated 35S- herpesvirus across an osmotically modified blood-brain barrier, J Neurosurg 74(3) 475-79 [1991], Also, S E Doran etal, Gene expression from recombinant viral vectors in the central nervous system after blood-brain barrier disruption, Neurosurgery 36(5) 965-70 [1995], G Nilaver et al, Delivery of herpesvirus and adenovirus to nude rat intracerebral tumors after osmotic blood-brain barrier disruption, Proc Natl Acad Sci USA 92(21) 9829-33 [1995])
Intracarotid infusion of leukotπene C sub 4 (LTC sub 4) selectively increases the permeability in brain tumor capillaries without affecting the permeability in normal brain capillaries The effect of LTC sub 4 on brain tumor capillaries is, however, limited to small molecules and it can only slightly increase the permeability of those small molecules in abnormal brain tissue relative to normal Accordingly, LTC. sub 4 does not significantly increase the delivery of some larger water soluble molecules to brain tumors or other abnormalities
The vasoactive nanopeptide bradykinin and agonists or polypeptide analogs thereof (e.g , receptor-mediated permeabilizers [RMPs]) have been injected intravenously to increase blood-brain barrier permeability to co-administered neuropharmaceutical or diagnostic agents
(B Malfroy-Camine, Method for increasing blood-brain barrier permeability by administering a bradyki m agonist of blood-brain barrier permeability, U S Patent No 5,112,596. J W Kozarich et al, Increasing blood brain barrier permeability with permeabihzer pephdes, U S Patent No 5,268,164) Intracarotid infusion of bradykinin will selectively increase permeability 2- to 12-fold in brain tumor and ischemic brain capillaries for molecules ranging in molecular weight from 100 to 70,000 Daltons (Inamura, T et al, Bradykinin selectively opens blood-tumor barrier in experimental brain tumors, J Cereb Blood Flow Metab 14(5) 862-70 [1994]) Bradykinin does not increase permeability in the normal blood brain barrier except at very high doses (Wirth, K et al, DesArg9-D-Arg[Hyp3,Thi5,D-Tιc7,0ιc8]bradykmιn (desArg!0-[Hoel40]) is a potent bradykinin Bl receptor antagonist, Eur J Pharmacol 205(2) 217-18 [1991]) Opemng of the blood-tumor barrier by bradykinin is transient, lasting 15 to 20 minutes (Inamura et al. [1994]) After opening of abnormal brain capillaries with bradykinin, the capillaries become refractory to the bradykinin effect for up to 60 minutes (Inamura et al. [1994]) A method for selectively delivering to abnormal brain tissue a neuropharmaceutical agent (e g , 5-fluorouracil, cisplatin, methotrexate, or monoclonal antibodies) or a diagnostic agent (e g , technicium-99 glucoheptonate, gallium-EDTA, and ferrous magnetic or iodinated contrasting agents) employed intracarotid infusion of bradykinin, or a bradykinin analog, such as RMP-7, the bradykinin or bradykinin analog was administered approximately contemporaneously with the agent (K L Black, Method for selective opening of abnormal brain tissue capillaries, U S Patent Nos 5,527,778 and 5,434,137) Enhanced transvascular delivery of HSV-derived viral particles to malignant cells in the brains of rats was also achieved by disrupting the blood-brain barrier with bradykinin or RMP-7 (N G Rainov, Selective uptake of viral and monocrystalline particles delivered intra-arterially to experimental brain neoplasms, Hum Gene Ther 6(12) 1543-52 [1995], N G Rainov et al , Long-term survival in a rodent brain tumor model by bradykinin-enhanced intra-arterial delivery of a therapeutic herpes simplex virus vector, Cancer Gene Ther 5(3) 158-62 [1998], F H Barnett et al, Selective delivery of herpes virus vectors to experimental brain tumors using RMP-7, Cancer Gene Ther 6(1) 14-20 [1999])
The calcium-activated potassium channel (K^ ) is an important regulator of blood vessel tone (Nelson MT, Quayle JM Physiological roles and properties of potassium channels in arterial smooth muscle, Am J Physiol 268(4 Pt 1) C799-822[1995], Bang, L et al, Nitroglycerin-mediated vasorelaxatwn is modulated by endothelial calcium-activated potassium channels, Cardiovasc Res 43(3) 772-78 [1999]) The KCa channel is ubiquitously distributed in tissues as and subunits Its activity is triggered by depolarization and enhanced by an increase in cytosolic calcium di-cation (Ca2+) A local increase in Ca2+ is sensed by the extremely sensitive brain -subunit of the KCa, directed towards the cytoplasm in the cell, that allows a significant potassium cation flux through these channels Under conditions when intracellular cyclic 3', 5' adenosine monophosphate (cAMP) concentration rises in vascular endothelium (e g hypoxia), ATP-sensitive potassium channels (KATP) may also play a role (J E Brian et al, Recent insights into the regulation of cerebral circulation, Clin Exp Pharmacol Physiol. 23(6-7) 449-57 [1996]) Minoxidil sulfate and chromakalim are reported to be activators of KATP (A D Wickenden et al, Comparison of the effects of the K(+)-channel openers cromaka m and minoxidil sulphate on vascular smooth muscle, Br J Pharmacol, 103(1) 1148-52 [1991])
Intimately connected with the regulation of potassium channels is guanosine 3',5'-cyclic monophosphate, commonly known as cyclic GMP (cGMP), an important signal transducing molecule, which mediates the regulation of three main classes of effector proteins (1) cGMP-dependent protein kinases, which mediate protein phosphorylation, (2) cGMP-gated ion channel protein kinases, which mediate cation influx across the plasma membrane, and (3) phosphodiesterases, which mediate cyclic nucleotide catabo sm (Lohse, M J et al, Pharamacology of NO:cGMP signal transduction, Naunyn-Schmiedebergs Arch Pharmacol 358 111-12 [1998], Smolenski, A et al, Functional analysis of cGMP -dependent protein kinases I and II as mediators of NO/cGMP effects, Naunyn-Schmiedebergs Arch Pharmacol 358 134-39 [1998], He, P et al , cGMP modulates basal and activated microvessel permeability independently of [Ca2+]ι, Am J Physiol 274(6 Pt 2) H1865-74 [1998], Holschermann, H et al. , Dual role ofcGMP in modulation of macromolecule permeability of aortic endothelial cells, Am J Physiol 272(1 Pt 2) H91-98 [1997])
The production of cGMP from GTP is catalyzed by soluble guanylyl cyclase, a nitric oxide-activated enzyme (Patel, A I and Diamond, J , Activation of guanosine 3 ',5 '-cyclic monophosphate (cGMP)-dependent protein kinase in rabbit aorta by nitroglycerin and sodium nitroprusside, ! Pharmacol Exp Ther 283(2) 885-93 [1997], Patel, A I et al , Activation of guanosine 3 ',5 '-cyclic monophosphate (cGMP) -dependent protein kinase in rat vas deferens and distal colon is not accompanied by inhibition of contraction, J Pharmacol Exp Ther 283(2) 894-900 [1997])
There is also evidence that nitric oxide participates in the regulation of microvascular tone (Joo, F et al, Regulation of the macromolecular transport in the brain microvessels: the role of cyclic GMP, Brain Res 278(1-2) 165-74 1983]) For example, glial tumors and ischemic tissue are more immunopositive for nNOS and eNOS relative to normal brain (Cai, Z et al , Prenatal hypoxia-ischemia alters expression and activity of nitric oxide synthase in young rat brain and causes learning deficits, Brain Res Bull 49(5) 359-65 [1999], Nakano, S et al, Increased brain tumor microvessel permeability after intracarotid bradykinm infusion is mediated by nitric oxide, Cancer Research, 56 4027-4031 [1996], Faraci, F M et al, Role of soluble guanylate cyclase in dilator responses of the cerebral microcirculation, Brain Res 821(2) 368-73 [1999]) Further, the pretreatment of glioma-beanng rats with the NOS inhibitor, L-NAME, significantly reduces bradykinin-induced permeability (Moncada, S et al, Endogenous nitric oxide: physiology, pathology and clinical relevance, Eur J Clin Invest 21(4)361-74 [1991], Sugita, M et al., Nitric oxide and cyclic GMP attenuate sensitivity of the tumor barrier to bradykinin, Neurological Research 20 559-563 [1998])
In turn, one class of enzymes that is activated by cGMP (and cAMP) is cGMP-dependent protein kinases (PKG or cGK), which through enzymatic ATP-dependent phosphorylation, directly or indirectly activate calcium-dependent potassium channels (Robertson, B E et al. , cGMP -dependent protein kinase activates Ca-activated K channels in cerebral artery smooth muscle cells, Am J Physiol 265 [Cell Physiol 34) C299-C303 [1993], Fukao, M et al, Cyclic GMP -dependent protein kinase activates cloned BKCa channels expressed in mammalian cells by direct phosphorylation at serine 1072, 1 Biol Chem 274(16) 10927-35 [1999], Becker, E M etal, The vasodilator-stimulated phosphoprotein (VASP): target ofYC-1 and nitric oxide effects in human and rat platelets, ! Cardiovasc Pharmacol 35(3) 390-97 [2000]) There is also evidence that nitric oxide can activate K^ by both cGMP-dependent and cGMP-independent mechanisms (Chen, C H et al, Nitric oxide activates Ca2* -activated Kf channels in cultured bovine adrenal chromaffin cells, Neurosci Lett 248(2) 127-29 [1998], Vaali, K et al, Relaxing effects of NO donors on guinea pig trachea in vitro are mediated by calcium-sensitive potassium channels, J Pharmacol Exp Ther 286(1) 110-14 [1998], Sobey,
C G and Faraci, F M , Inhibitory effect of 4-amιnopyrιdιne on responses of the basilar artery to nitric oxide, Br J Pharmacol 126(6) 1437-43 [1999], Kurtz, A et al. , Mode of nitric oxide action on the renal vasculature, Acta Physiol Scand 168(1) 41-45 [2000])
Treatments directed to the use of potassium channel activators or agonists have been taught for disorders including hypertension, cardiac and cerebral ischemia, nicotine addiction,bronchial constriction, and neurodegenerative diseases, but not particularly for the treatment of malignant tumors (Erhardt et al. , Potassium channel activators/openers, U S Patent No 5,416,097, Schohe-Loop et al, 4, 4 '-bridged bιs-2, 4-dιamιnoquιnazolιnes, U S Patent No 5,760,230, Sit et al, 4-aryl-3-hydroxyquιnolιn-2-one derivatives as ion channel modulators, U S Patent No 5, 922,735, Garcia et al, Biologically active compounds, U S Patent No 5,399,587, Cherksey, Potassium channel activating compounds and methods of use thereof, U S Patent No 5,234,947)
Bradykinin is thought to increase [Ca2^ and thus may activate K^ channels While some other known activators of IC^ do not act as vasodilators, for example, 1,3-dihydro-l- [2-hydroxy-5-(trifluoromethyl)phenyl]-5-(trifluoromethyl)-2H-benzimidazol-2-one (NS- 1619,
M Holland et al, Effects of the BKCa channel activator, NS1619, on rat cerebral artery smooth muscle, Br J Pharmacol , 117(1) 119-29 [1996]), evidence is accumulating that Kca may play an important role in vasodilatation mediated by vasodilators, such as bradykinin, nitric oxide donors, cyclic guanosine monophosphate (cGMP), and guanylyl cyclase activators (Berg T , Koteng O , Signaling pathways in bradykinin- and nitric oxide-induced hypotension in the normotensive rat; role of Kf -channels, Br J Pharmacol ,121(6) 1113-20 [1997], Bolotina, V M et al. , Nitric oxide directly activates calcium-dependent potassium channels in vascular smooth muscle, Nature 368(6474) 850-3 [1994], Robertson, B E , et al, cGMP -dependent protein kinase activates Ca-activated K channels in cerebral artery smooth muscle cells, Am J Physiol 265(1 Pt 1) C299-303 [1993], Sobey, C G et al, Mechanisms of bradykinin-induced cerebral vasodilatation in rats. Evidence that reactive oxygen species activate K+ channels, Stroke 28(11) 2290-4, discussion 2295 [1997], C G Sobey and F M Faraci, Effect of nitric oxide and potassium channel agonists and inhibitors on basilar artery diameter, Am J Physiol 272(1 Pt 2) H256-62 [1997], Hardy, P et al, A major role for prostacyclin in nitric oxide-induced ocular vasorelaxatwn in the piglet, Circ Res 83(7) 721-29
[1998], Bychkov R et al, Calcium-activated potassium channels and nitrate-induced vasodilatwn in human coronary arteries, J Pharmacol Exp Ther 285(1) 293-98 [1998], Armstead, W M , Contribution qfKCa Channel activation to hypoxic cerebrovasodilatwn does not involve NO, Brain Res 799(1) 44-48 [1998]) Bradykinins action as a powerful vasodilator is disadvantageous when using bradykinin to open the blood-brain barrier to therapeutic anticancer agents Bradykinin or its analogs may adversely lower blood pressure, reduce cerebral blood flow, or contribute to brain edema in some patients (E g , A M Butt, Effect of inflammatory agents on electrical resistance across the blood-brain barrier inpial microvessels of anesthetized rats, Brain Res 696(1-2) 145-50 [1995]) In addition, bradykinin constricts smooth muscle and stimulates pain receptors Consequently, there is still a definite need to maximize the effectiveness of a wide variety of therapeutic agents through enhanced selective transvascular delivery to malignant tumors, including those of the central nervous system, and/or to other abnormal brain regions These and other benefits the present invention provides as described herein
SUMMARY OF THE INVENTION
The present invention relates to a method of delivering a medicant to an abnormal brain region in a mammalian subject, including a human The method includes administering to the subject a potassium channel activator (i e , activator of calcium- or ATP-dependent potassium channels [KCa or KATP]) Potassium channel activators include direct agonists (other than bradykinin or bradykinin analogs), such as NS-1619 or minoxidil Potassium channel activators also include compounds that indirectly activate potassium channels, for example nitric oxide, nitric oxide donors, and other activators of guanylyl cyclase Activators of cyclic GMP-dependent protein kinase, which activates calcium-dependent potassium channels, are also included The potassium channel activator is administered to the subject under conditions and in an amount sufficient to increase the permeability to the medicant of a capillary or arteriole delivering blood to cells of the abnormal brain region in the subject Simultaneously or substantially simultaneously with the potassium channel activator, the medicant is administered, so that the medicant is delivered selectively to the cells of the abnormal region compared to normal brain regions, due to the increased permeability of capillaries and arterioles supplying the abnormal brain region The method is particularly valuable in the treatment of physical or biochemical brain injuries caused by trauma, infection, stroke, ischemia, and, particularly, malignant brain tumors, for which survival rates are notoriously poor
The present invention also relates to a method of delivering a medicant to a malignant tumor in the brain or anywhere in the body of a mammalian subject The method involves administering to the subject a potassium channel activator, such as a potassium channel agonist other than bradykinin or a bradykinin analog, under conditions and in an amount sufficient to increase the permeability to the medicant of a capillary or arteriole delivering blood to cells of the malignant tumor in the subject Simultaneously or substantially simultaneously with the potassium channel activator the medicant is administered to the subject, and it is delivered selectively to the malignant cells compared to non-malignant cells by virtue of the potassium channel activator The inventive method is useful in treating any kind of malignant tumor by increasing the selectivity of drug delivery to neoplastic tissue, thereby minimizing damage to non-malignant tissue from medicants, including cytotoxic chemotherapeutic agents, and focusing the therapeutic or diagnostic action of the agents Thus, this invention, also directed to a method of treating a malignant tumor in a human subject, offers enhanced prospects of survival to cancer patients, with fewer harmful side effects The selectivity of the methods is based on the role of calcium-and ATP-dependent potassium transporters (channels) in mediating the permeability of microvasculature to various drugs, macromolecules, and viral particles, combined with the greater number of calcium- and ATP-dependent potassium channels present in abnormal brain vasculature or tumor neomicrovasculature compared to normal microvasculature
The present invention also relates to a pharmaceutical composition that comprises a combination of a potassium channel activator, other than bradykinin or a bradykinin analog, formulated in a pharmaceutically acceptable solution together with a medicant for delivery by intravascular infusion or bolus injection into a mammal, such as a human The pharmaceutical composition is useful in practicing the inventive methods
The invention also relates to a kit for enhancing the delivery of a medicant to an abnormal brain region and/or to a malignant tumor These and other advantages and features of the present invention will be described more fully in a detailed description of the preferred embodiments which follows
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 shows the response to the soluble guanylyl cyclase activator YC-1 (2 66 μg min"1 kg"1, n = 6) in the unidirectional transfer constant K, for [14C]α-aminoisobutyric acid in malignant RG2 glioma tissue in Wistar rats, compared to DMSO + saline control (n = 6), YC-1
+ iberiotoxin (IBTX, 0 2 μg IBTX min"1 kg"1, n = 3), or YC-1 + a selective inhibitor of soluble guanylyl cyclase, lH-[l,2,4]oxadiazolo[4,3-a]quinozalin-l-one (ODQ) (2 66 μg min"1 kg"1, n = 4) treatments for 15 minutes each K, (μL/g/min) in the tumor core (gray bars), tumor-adjacent tissue taken about 3 mm from the tumor core (white bars), and normal contralateral brain tissue (black bars) is shown
Figure 2 shows the enhancing effect of the nitric oxide donor diethylamine-NONOate (DEA/NO) on blood-tumor barrier permeability to Evans blue stain in malignant RG2 glioma tissue in Wistar rats Figure 2A shows a brain section from a rat treated with phosphate buffered saline (PBS only control) Figure 2B shows a brain section from a DEA/NO-treated rat
Figure 3 shows the response to DEA/NO (2 66 μg min"1 kg"1, n = 3) in the unidirectional transfer constant K, for [14C]α-aminoisobutyric acid in malignant RG2 glioma tissue in Wistar rats, compared to PBS control, DEA/NO + iberiotoxin (IBTX, 0 2 μg IBTX min"1 kg"1, n = 3), or PAPA/NO (2 66 μg min"1 kg'1, n = 3) treatments for 15 minutes each K, (μL/g/min ) in tumor centers (black bars), tumor-adjacent tissue taken about 3 mm from the tumor core (white bars), and normal contralateral brain tissue (cross-hatched bars) is shown
Figure 4 shows transendothelial vesicular transport in a RG2 tumor capillary RG2 tumor-bearing brain sections from Wistar rats were prepared after a 15-minute intracarotid infusion with PBS (0 8 mL)(Figure 4A, 54,000x magnification), bradykinin (10 μg min"1 kg"x)(Figure 4B, 87,000x magnification), or NS-1619 (5 3 μg min"1 kg"1, Figure 4C, 87,000x magnification) TEM analysis of the tumor center section shows that KCa activators bradykinin and NS-1619 increase pinocytotic transport vesicles in the endothelial cytoplasm Figure 5 shows that a potassium channel activator (bradykinin) increases transport of horse radish peroxidase (HRP) from the tumor capillary lumen through endothelial pinocytotic vesicles Figure 5 A (8,700x magnification) and Figure 5B (21,000x magnification) show vascular and tumor tissue from a rat in the PBS control group Figure 5C (8,700x magnification) and Figure 5D (21,000x magnification) show vascular and tumor tissue from a rat treated with the potassium channel activator bradykinin Figure 6 illustrates that a potassium channel activator (bradykinin) induces vesicular transport of HRP in RG2 tumor cells Figure 6A (8,700x magnification) shows tumor tissue from a rat in the PBS control, Figure 6B (10,800x magnification) and Figure 6C (21,000x magnification) show tumor tissue from a bradykinin-treated rat
Figure 7 shows an increase in the unidirectional transport constant caused by the KATP activator minoxidil sulfate and the KCa activator bradykinin Figure 8 shows that bradykinin-induced increase in K, was not attenuated by glibenclamide, while IBTX significantly decreased the effect caused by bradykinin
Figure 9 shows that the minoxidil sulfate-induced increase in K, was attenuated by glibenclamide, while IBTX failed to block the effect Figure 10 illustrates increased micro vascular permeability in an ischemic brain region as a result of treatment with a potassium channel activator K, is shown for bradykinin-treated (black bars) and control animals (gray bars) in paired histograms, tissues tested were (left to right) caudate putamen, cortex, caudate putamen contralateral side, and cortex contralateral side Figure 10A shows that blood-brain barrier permeability after one hour of ischemia was not affected by bradykinin treatment compared to the vehicle-treated group (MCA occlusion for
1 hour, followed by 1 hour of reperfusion, Inset autoradiograph) Figure 10B shows that bradykinin significantly increased permeability in ischemic infarct penumbra after 2 hours of MCA occlusion, followed by 1 hour of reperfusion (Inset autoradiograph)
Figure 11 shows that potassium activators enhance permeability of microvasculature in ischemic brain tissue No change in blood-brain barrier permeability at the infarct brain tissue
(significantly less uptake of Evans blue) was observed in a rat subjected to temporary ischemia (2 hours MCA occlusion and 1 hour of reperfusion, top and bottom left panels) Considerable enhancement of Evans blue uptake was seen in ischemic brain tissue in bradykinin-treated (10 μg min^kg"1 for 15 min, top middle panel) or NS-1619-treated (1 5 μg min"1 kg"1 for 15 min, lower middle panel) rats compared to PBS vehicle-treated (top and bottom left panels)
However, co-infusion with IBTX (0 2 μg min"1 kg"1 for 15 min) attenuated the increased uptake of Evans blue induced by bradykinin (top right panel) or NS-1619 (bottom right panel)
Figure 12A shows the enhancing effect of NS-1619 on blood-tumor barrier permeability to [14C] -aminoisobutyric acid (AIB) tracer (left) compared to the effect on blood-brain barrier permeability in normal brain tissue adjacent (middle) and contralateral (right) to malignant RG2 glioma tissue in Wistar rats
Figure 12B shows the enhancing effect of minoxidil sulfate on blood-tumor barrier permeability to [14C]ATB tracer (left) compared to the effect on blood-brain barrier permeability in normal brain tissue adjacent (middle) and contralateral (right) to malignant RG2 ghoma tissue in Wistar rats Figure 13 shows a dose-response to NS-1619 in the unidirectional transfer constant K, for [14C] -aminoisobutyric acid in malignant RG2 glioma tissue in Wistar rats K, = L/g/min
Figure 14 shows specific inhibition by iberiotoxin (IBTX, 2 3 μg kg"1 min"1) of the permeability increasing effect of NS-1619 (26 5 μg kg"1 min"1) The K, was determined in RG2 tumor-bearing Wistar rats using [14C]AIB with NS-1619 (26 5 μg kg"1 min"1) with or without
IBTX (2 3 μg kg"1 min"1), for 15 minutes The results are compared with PBS, pH 7 4 with or without 5% ethanol
Figure 15 shows intense over-expression of KCa as indicated by anti-KCa immunostain of glioma tissue (Fig 15B), compared to normal contralateral brain tissue (Fig 15 A) Magnification is lOOx
Figure 16 shows intense over-expression of KATP as indicated by anti-KATP (anti-Kir 6 2) immunostain of RG2 (Fig 16C) or C6 (Fig 16D) glioma tissue from tumor centers, compared to normal brain tissue (Fig 16 A) and RG2 tumor periphery (Fig 16B) Magnification is lOOx
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS The inventive methods are useful for selectively delivering a medicant to abnormal brain regions and/or malignant tumors in mammalian subjects The methods involve administering to the mammalian subject a potassium channel activator, other than bradykinin or a bradykinin analog, under conditions and in an amount sufficient to increase the permeability to the medicant of a capillary or arteriole delivering blood to cells of the abnormal brain region and/or to malignant cells of a malignant tumor present in the subject The increase in permeability ranges from at least 2-to 6-fold, compared to controls without the administration of a potassium channel activator. The relative increase in permeability tends to be greater for large molecular weight medicants (e g., about 10,000 to 250,000 Daltons) than for smaller molecular weight substances (e g , about 50-200 Daltons) The abnormal brain regions include regions of brain tissue physiologically directly affected by a physical or biochemical injury, for example, Alzheimers disease, Parkinsonism, trauma, infection, stroke, brain ischemia, or regions of neoplastic growth within the brain, such as benign or malignant brain tumor tissues The present invention is also useful for selectively delivering a medicant to a malignant tumor in the brain or to a tumor elsewhere in the body of a mammalian subject. The inventive technology is useful in the treatment of all kinds of solid malignant tumors, including gliomas, glioblastomas, oligodendrogliomas, astrocytomas, ependymomas, primitive neuroectodermal tumors, atypical meningiomas, malignant meningiomas, neuroblastomas, sarcomas, melanomas, lymphomas, or carcinomas. The tumor to be treated can be contained in the skull, brain, spine, thorax, lung, peritoneum, prostate, ovary, uterus, breast, stomach, liver, bowel, colon, rectum, bone, lymphatic system, skin, or in any other organ or tissue of the subject.
The inventive methods are useful in treating any mammal, including a human, non-human primate, canine, feline, bovine, porcine or ovine mammal, as well as in a small mammal such as a mouse, rat, gerbil, hamster, or rabbit.
The potassium channel activator is an activator of either a calcium-activated potassium channel (KCa) of any conductance level, whether of large, intermediate, or small conductance, or of an ATP-sensitive potassium channel (KATP). Included are direct agonists of KCa, such as 1 ,3-dihydro- 1 -[2-hydroxy-5-(trifluoromethyl)phenyl]-5-(trifluoromethyl)-
2H-benzimidazol-2-one (NS-1619) or l-ethyl-2-benzimidazolinone (1-EBIO).
Also included among useful potassium channel activators are compounds that indirectly activate potassium channels, for example activators of soluble guanylyl (i.e., guanylate) cyclase, such as nitric oxide, nitric oxide donors, metalloporphyrins (e.g., zinc or tin protoporphyrin IX), YC-1 (a benzyl indazole derivative), or guanylyl cyclase activating proteins (GCAPs). (See, e.g., Koesling, D., Modulators of soluble guanylyl cyclase, Naunyn-Schmiedebergs Arch. Pharmacol.358: 123-126 [1998]).
Examples of useful potassium channel activators that are KATP agonists include minoxidil (2,4-diamino-6-piperidino pyramidine-3 -oxide; insoluble in water, soluble in ethanol 29 mg/mL), pinacidil ((+/-)-N-cyano-N'-4-pyridinyl-N"-(l,2,2-trimethyl propyl)- guanidine; insoluble in water, soluble in ethanol 14 mg/mL]), (+)-cromakalim, (-)-cromakalim or levcromakalim, (+/-)-cromakalim, or diazoxide.
A preferred potassium channel activator is nitric oxide gas, which is fully permeable across biological membranes. Inhalable nitric oxide gas can be administered to the subject by mask in a controlled gas mixture as is known in the art. (E.g., Kieler- Jensen, N. et al. , Inhaled nitric oxide in the evaluation of heart transplant candidates with elevated pulmonary vascular resistance, J Heart Lung Transplant 13(3) 366-75 [1994], Rajek, A et al, Inhaled nitric oxide reduces pulmonary vascular resistance more than prostaglandin E(l) during heart transplantation, Anesth Analg 90(3) 523-30 [2000], Solina, A et al, A comparison of inhaled nitric oxide and milrinone for the treatment of pulmonary hypertension in adult cardiac surgery patients, J Cardiothorac Vase Anesth 14(1) 12-17 [2000], Fullerton, D A et al, Effective control of pulmonary vascular resistance with inhaled nitric oxide after cardiac operation, J Thorac Cardiovasc Surg 111(4) 753-62, discussion 762-3 [1996]) The concentration in the gas mixture of nitric oxide (NO) is preferably about 1 to 100 ppm NO, more preferably about 4 to 80 ppm NO, and most preferably about 20 to 40 ppm NO The gas mixture also contains appropriate concentrations of oxygen and nitrogen and/or other inert gases, such as carbon dioxide, helium or argon Optionally, gaseous anesthetics, such as nitrous oxide (N2O), xenon, and halogenated volatile anesthetics (HVAs), e g , halothane, sevoflurane, and isoflurane, are also included in the gas mixture when general anesthesia is indicated General anesthesia is indicated, for example, when administration of the potassium channel activator (and/or the medicant or chemotherapeutic agent) is by intracarotid infusion, general anesthesia is typically not required using intravenous or other delivery routes The skilled practitioner is aware of evidence that HVAs can inhibit soluble guanylyl cyclase activity (See, Masaki, E , Halogenated volatile anesthetics inhibit carbon monoxide-stimulated soluble guanylyl cyclase activity in rat brain, Acta Anaesthesiol Scand 44(3) 321-25 [2000], Masaki
E and Kondo I, Methylene blue, a soluble guanylyl cyclase inhibitor, reduces the sevoflurane minimum alveolar anesthetic concentration and decreases the brain cyclicguanosine monophosphate content in rats, Anesth Analg 89(2) 484-89 [1999]) Consequently, an HVA is not the preferred choice of inhalable anesthesia for use with a guanylyl cyclase activator in accordance with the method
Nitric oxide donors are compounds that produce NO-related physiological activity when applied to biological systems Thus, NO-donors can mimic an endogenous NO-related response or substitute for an endogenous NO deficiency The skilled artisan is aware that in biological systems there are at least three redox states of NO that can be released by various NO donors (NO+, NO0, or NO"), all of which are encompassed by the terms "nitric oxide" or "NO" for purposes of the present invention The redox state of NO makes a substantial difference to the NO donors reactivity towards other biomolecules, the profile of by-products, and the bioresponse (Feelisch, M , The use of nitric oxide donors in pharmacological studies, Naunyn-Schmiedebergs Arch Pharmacol 358 113-22 [1998]) Some classes of NO donors require enzymatic catalysis, while others produce NO non-enzymatically, some NO donors require reduction, for example by thiols, and some oxidation, in order to release NO
Preferred examples of nitric oxide donors include organic nitrate compounds, which are nitric acid esters of mono- and polyhydric alcohols Typically, these have low water solubility, and stock solutions are prepared in ethanol or dimethyl sulfoxide (DMSO) Examples are glyceryl trinitrate (GTN) or nitroglycerin (NTG), pentaerythrityl tetranitrate (PETN). isosorbide dinitrate (ISDN), and isosorbide 5-mononitrate (IS-5-N) Administration of organic nitrates can be done intravenously, intraperitoneally, intramuscularly, transdermally, or in the case of PETN, ISDN, NTG, and IS-5-N, orally
Other preferred examples are S-nitrosothiol compounds, including S-nitroso-N-ace1yl-D,L-perύciUamine (SNAP), S-nitrosoglutathione (SNOG), S-nitrosoalbumin,
S-nitrosocysteine S-nitrosothiol compounds are particularly light-sensitive, but stock solutions kept on ice and in the dark are stable for several hours, and chelators such as EDTA can be added to stock solutions to enhance stability Administration is preferably by an intravenous or intra-arterial delivery route Other preferred examples of nitric oxide donors include sydnonimine compounds, such as molsidomine (N-ethoxycarbonyl-3-morpholino-sydnonimine), linsidomine (SLN-1, 3 -morpholino-sydnonimine or 3 -morpholinylsydnoneimine or
5-amino-3mo holinyl-l,2,3-oxadiazolium, e g , chloride salt), and pirsidomine (CAS 936) Stock solutions are typically prepared in DMSO or DMF, and are stable at 4CC to room temperature, if protected from light Linsidomine is highly water soluble and stable in acidic solution in deoxygenated distilled water, adjusted to about pH 5, for an entire day At physiological pH, SIN-1 undergoes rapid non-enzymatic hydrolysis to the open ring form SIN-1 A, also a preferred nitric oxide donor, which is stable at pH 7 4 in the dark Administration is preferably by an intravenous or intra-arterial delivery route Also useful as nitric oxide donors are iron nitrosyl compounds, such as sodium nitroprusside (SNP, sodium pentacyanonitrosyl ferrate(II)) Aqueous stock solutions are preferably made freshly in deoxygenated water before use and kept in the dark, stability of stock solutions is enhanced at pH 3-5 Inclusion in the delivery buffer of a physiologically compatible thiol, such as glutathione, can enhance release of NO SNP is administered by intravenous infusion, and the skilled practitioner is aware that long-term use is precluded by the release of five equivalents of toxic CN-per mole SNP as NO is released
A most preferred nitric oxide donor is chosen from among the so-called NONOate compounds The NONOates are adducts of NO with nucleophilic residues (X"), such as an amine or sulfite group, in which an NO dimer is bound to the nucleophilic residue via a nitrogen atom to form a functional group of the structure X[-N(O)NO]" The NONOates typically release NO at predictable rates largely unaffected by biological reactants, and NO release is thought to be by acid-catalyzed dissociation with the regeneration of X" and NO This property is particularly useful in accordance with the inventive methods of selectively delivering a medicant, because abnormal brain regions and malignant tumors can typically be relatively hypoxic and possess a relatively low ambient pH (e g , pH 6 5-7 0), which concentrates release of NO selectively in the microvasculature of the abnormal brain region or malignant tumor
NONOates include most preferably diethylamine-NONOate (DEA/NO, N-Ethylethanamine l , l -Diethyl-2-hydroxy-2-nitrosohydrazine (1 1 ) or l-[N,N-diethylamino]diazen-l-ium-l,2-diolate) Other preferred NONOates include diethylene triamine-NONOate(DETA/NO, 2,2'-Hydroxynitrosohydrazino]bis-ethanamine), spermine-NONOate (SPER/NO, N-(4-[-l-(3-Aminopropyl)-2-hydroxy-2-nitrosohydrazino] butyl)-l,3-propanediamine), propylamino-propylamine-NONOate (PAPA/NO, 3-(2-Hydroxy-2-nitroso-l-propylhydrazino)-l-propanamine or (Z)-l-[N-(3-aminopropyl)-N-
(n-propyl)amino]diazen-l-ium-l,2-diolate), MAHMA-NONOate (MAHMA/NO,
6-(2 -Hydroxy- 1 -methyl-2- nitrosohydrazino)-N-methyl- 1 -hexanamine), dipropylenetriamine- NONOate (DPTA/NO, 3,3'- (Hydroxynitrosohydrazino)bis-l-propanamine), PIPERAZI/NO, proli-NONOate (PROLI/NO, 1 -([2-carboxylato]pyrrolidin- 1 -yl)diazen- 1 -mm- 1 ,2-diolate- methanol, e g , disodium salt), SULFO-NONOate (SULFO/NO, hydroxydiazenesulfonic acid 1 -oxide, e g , diammonium salt), the sulfite NONOate (SULFI/NO), and Angelis salt (OXI/NO)
Almost all NONOate compounds are highly soluble in water, and aqueous stock solutions are prepared in cold deoxygenated 1 to 10 mM NaOH (preferably about pH 12) just prior to use Alkaline stock solutions are stable for several hours if kept on ice in the dark The characteristeic UN absorbance of NONOates can be used for spectrophotometric quantification of NONOate in aqueous solutions NONOates are preferably administered intravenously or intra-arterially Nitric oxide donors have different potencies (Ferraro, R et al, Comparative effects of several nitric oxide donors on intracellular cyclic GMP levels in bovine chromaffin cells: correlation with nitric oxide production, Br J Pharmacol 127(3) 779-87 [1999]) For example, DEA/NO is among the most potent nitric oxide donors, with a half-life of about 2 to 4 minutes, less potent are PAPA/NO (t1/2 about 15 minutes), SPER/NO (t1/2 about 34-40 minutes), even less potent are DETA NO (t1/2 about 20 hours) and SNAP (t1/2 about 33 to 41 hours, although this can be shortened in the presence of a physiological reductant such as glutathione) SNP is also a potent NO donor (See, Ferrero et al [1999], Salom, J B et al, Relaxant effects of sodium nitroprusside and NONOates in rabbit basilary artery, Pharmacol 57(2) 79-87 [1998], Salom, J B et al. , Comparative relaxant effects of the NO donors sodium nitroprusside, DEA NO and SPER/NO in rabbit carotid arteries, Gen Pharmacol 32(1) 75-79
[1999], Salom, J B et al, Relaxant effects of sodium nitroprusside and NONOates in goat middle cerebral artery: delayed impairment by global ischemia-reperfusion, Nitric Oxide 3(1) 85-93 [1999], Kimura, M et al, Responses of human basilar and other isolated arteries to novel nitric oxide donors, J Cardiovac Pharmacol 32(5) 695-701 [1998]) Consequently, effective concentrations or doses of NONOates or other NO donors will vary over the preferred dose ranges for potassium channel activators described herein
Stock solutions of NO donors are preferably made up freshly before use (at the appropriate pH for each particular NO donor), chilled on ice, and protected from light (e g , by the use of darkened glass vials wrapped in aluminum foil), although organic nitrates can be stored for months to years if the vial is properly sealed Preferably, immediately before administration to the subject, final dilutions are prepared in pharmaceutically acceptable buffer and the final pH of the NO donor-containing buffer is checked for physiological suitability, especially when strongly acidic (e g , hydrochloπde salts) or alkaline (e g , NONOates) stock solutions are used The product of NO exposure time and NO concentration largely determines the quality and magnitude of the biological response to exogenously supplied NO Short-lived NO donors, such as DEA/NO, are most preferably admimstered by continuous infusion rather than by bolus to avoid delivering only a short burst of NO
Also included among potassium channel activators are activators of any endogenous species of cyclic GMP-dependent protein kinase (PKG or cGK), that activates a potassium channel directly (e g , by directly phosphorylating KCa) or indirectly (e g , by phosphorylating another regulatory protein that directly modulates Kca activity) Included are activators of cGK I, cGK II, or other isoforms of cGMP-dependent protein kinase (e g , Smolenski, A et al. [1998]) Useful examples of PKG activators include, but are not limited to, octobromo-cyclic GMP (8Br-cGMP) and dibutyryl cyclic GMP
Included among useful potassium channel activators are pharmaceutically acceptable molecular conjugates or salt forms that still have activity as potassium channel activators as defined herein An example is minoxidil sulfate, but other pharmaceutically acceptable salts comprise anions other than sulfate, such as chloride, carbonate, bicarbonate, nitrate, or the like Other embodiments of pharmaceutically acceptable salts contain cations, such as sodium, potassium, magnesium, calcium, ammonium, or the like Other embodiments of useful potassium channel activators are hydrochloride salts
However, the potassium channel activator employed in the inventive methods is one other than the vasodilator bradykinin (Arg-Pro-Pro-Gly-Phe-Ser-Pro-Phe-Arg), or a polypeptide bradykinin analog, such as receptor mediated permeabilizer (RMP)-7 or A7 (e g , Kozarich et α/, U S Patent No 5,268,164 and PCT Application No WO 92/18529) Other analogs of bradykinin include related peptide structures which exhibit the same properties as bradykinin but have modified amino acids or peptide extensions on either terminal end of the peptide Examples of bradykinin analogs include [phe sup 8 (CH sub 2 NH) Arg sup 9 ]-bradykinin, Nacetyl [phe sup.8 (CH sub 2 ~NH~Arg sup 9 ] bradykinin and desArg9-bradykinin In accordance with the inventive methods, the potassium channel activator is administered by intravenous or intra-arterial injection or infusion For treating an abnormal brain region, such as an intracranial tumor, the potassium channel activator is preferably administered by intracarotid infusion Except where otherwise noted, the amount of potassium channel activator to be administered to the mammalian subject ranges from 0 075 to 1500 micrograms per kilogram body mass For humans the range of 0 075 to 150 micrograms per kilogram body mass is preferred As the skilled practitioner is aware, the physiological responses of individual patients to treatment with particular potassium channel activators will vary. For example, generally an effective amount of YC-1 for humans is about 15 to about 45 micrograms per kg body mass, and for nitric oxide donors generally about 15 to about 45 micrograms per kg body mass However, the optimal amount for each individual for any particular potassium channel activator can be determined by routine means involving close physiological monitoring over the delivery period
The dose can be administered in a bolus injection, but is preferably administered by infusion over a period of one to thirty minutes, and most preferably during a period of one to fifteen minutes For example, in rats, a dose rate of about 0 75 to about 100 μg kg"1 min"1 is most suitable. At dose rates above about 100 μg kg"1 min"1 a concomitant fall in blood pressure has sometimes been observed In humans, effective dose rates are about 0 075 to about 15 μg kg"1 min"1, with cautious monitoring of blood pressure being advised The practitioner skilled in the art is also cautious in regulating the total infusion volume, rate of liquid infusion, and electrolyte balance to avoid adverse physiological effects related to these Some potassium channel activators, such as NS-1619, minoxidil, minoxidil sulfate, pinacidil, or diazoxide are not easily dissolved in water; in preparing these agents for administration, a suitable and pharmaceutically acceptable solvent, such as ethanol, can be used to dissolve the potassium channel activator prior to further dilution with an infusion buffer The skilled practitioner is cautious in regulating the final concentration of solvent in the infusion solution to avoid solvent-related toxicity For example, a final ethanol concentration in an infusion solution up to 5-10% (v/v) is tolerated by most mammalian subjects with negligible toxicity While the inventive method does not depend on any particular mechanism by which increased microvascular permeability to the medicant is achieved, it is thought that administration of the potassium channel activator increases potassium flux through potassium channels in endothelial cell membranes of the capillaries and arterioles delivering blood to abnormal brain regions and/or malignant tumors This results in a loosening of tight junctions in the microvascular epithelium and/or increased pinocytotic activity, enhancing the uptake of medicants from the blood vessels In practicing the inventive methods, it is not necessary to measure potassium channel activity (i e , potassium cation flux therethrough) But the skilled artisan is aware that potassium flux can be measured by any suitable method, for example, by measuring cellular uptake of 42K~ or 201τi+ or channel conductance using patch-clamp or microelectrode devices (e g , T Brismar et al, Thallιum-201 uptake relates to membrane potential and potassium permeability in human glioma cells, Brain Res 500(1-2) 30-36 [1989], T Brismar et al. , Mechanism of high K+ and TT uptake in cultured human glioma cells, Cell Mol Neurobiol 15(3) 351-60 [1995], S Cai et al, Single-channel characterization of the pharmacological properties of the K(Ca2+) channel of intermediate conductance in bovine aortic endothelial cells, ! Membr Biol 163(2) 147-58 [1998])
The medicant is administered simultaneously or substantially simultaneously with the potassium channel activator, and the medicant is delivered by the blood stream selectively to the abnormal brain region and/or to the malignant cells compared to normal brain tissue or non-malignant cells "Simultaneously" means that the medicant is administered contemporaneously or concurrently with the potassium channel activator "Substantially simultaneously" means that the medicant is administered within about one hour after the potassium channel activator is last administered, preferably within about 30 minutes after, and most preferably, is administered simultaneously with the potassium channel activator Alternatively, "substantially simultaneously" means that the medicant is administered within about 30 minutes before, and preferably within about 15 minutes before the potassium channel activator is first administered
The methods of delivering a medicant to an abnormal brain region and/or to a malignant tumor in a mammalian subject are effective in selectively delivering any medicant across the microvascular of an abnormal brain region and/or malignant tumor The medicant is a drug, i e , a chemotherapeutic agent Example of chemotherapeutic agents including therapeutic cytotoxic agents (e g , cisplatin, carboplatin, methotrexate, 5-fluorouracil, amphotericin), naked DNA expression vectors, therapeutic proteins, therapeutic oligonucleotides or nucleotide analogs, interferons, cytokines, or cytokine agonists or antagonists, adrenergic agents, anticonvulsants, anti-trauma agents, or any neuropharmaceutical agent used to treat or prevent an injury or disorder of the brain Chemotherapeutic agents also include ischemia-protective drugs such as N-methyl-D-aspartate (NMD A) receptor antagonists, antimicrobial agents, such as antibiotics, immunotoxins, immunosuppressants, boron compounds, monoclonal antibodies and specific antigen-binding antibody fragments (e g , Fab, Fab', F(ab')2, or F(v) fragments), and cytokines, such as interferons, interleukins (e g , interleukin [IL]-2), tumor necrosis factor
(TNF)-α, or transforming growth factors (e g , TGF-β)
The medicant also includes anticancer chemotherapeutic agents Typically, anticancer chemotherapeutic agents are cytotoxic agents, such as 5-fluorouracil, cisplatin, carboplatin, methotrexate, daunorubicin, doxorubicin, vincristine, vinblastine, or a cytotoxic alkylating agent, such as, but not limited to, busulfan (1,4-butanediol dimethanesulphonate, Myleran, Glaxo
Wellcome), chlorambucil, cyclophosphamide, melphalan, or ethyl ethanesulfonic acid
The anticancer chemotherapeutic agents are particularly useful in practicing the method of selectively delivering a medicant to a malignant tumor, in the brain or in any other tissue of the body, and in the method of treating a malignant tumor in a human subject Medicants also include any therapeutic viral particle, for example an adenovirus-derived or herpes simplex virus (HSV)-derived viral vector for delivering genetic material to a cellular target in vivo Medicants also include diagnostic agents, such as imaging or contrast agents, for example, radioactively labeled substances (e g , [99Tc]-glucoheptonate), gallium-labeled imaging agents (e.g , gallium-EDTA), ferrous magnetic, fluorescent, luminescent, or iodinated contrast agents Where suitable, any of the afore-mentioned medicants having anticancer activity can also be used in practicing the method of selectively delivering a medicant to a malignant tumor or the method of treating a malignant tumor in a human subject
Thus, the medicant can be a molecular substance having a molecular weight between about 50 Daltons and about 250 kD Or it can be a particle, such a s a viral particle, having a diameter between about 50 to 250 nanometers This is by no means intended to be an exhaustive list of the kinds of medicants that can be employed in practicing the inventive methods. The medicant can be, but is preferably not, an agent that is highly lipid soluble and thus inherently able to penetrate cell membranes, for example nitrosourea. The amount of medicant that is employed is within a conventional dose range for each medicant, however by practicing the inventive method, the increased transvascular permeability afforded can provide a greater selective therapeutic effect per dose or permit a lower effective dose to be used, if desired, for example to lessen systemic toxic effects from anti-cancer medication in a particular subject. The medicant is administered by any appropriate method that can deliver it to the blood stream. Typically, this is by intravenous, intramuscular, or intra-arterial (including intracarotid) injection or infusion. However, for some applications other acceptable delivery routes can be used as long as the dose of medicant enters the blood stream substantially simultaneously with the potassium channel activator. Examples include ingestion (e.g., of a powder, suspension, solution, emulsion, tablet, capsule or caplet); subcutaneous injection; stereotactic injection; or transdermal or transmucosal delivery by adhesive patch, suppository or gel for delivery through the skin, mucosa or epithelium of the mouth including the sublingual epithelium, the rectum, or the vaginal epithelium.
Alternatively, the medicant is administered together with the potassium channel activator in a pharmaceutical composition of the present invention. The inventive pharmaceutical composition comprises a combination of a potassium channel activator, other than bradykinin or a bradykinin analog, as described above, formulated in a pharmaceutically acceptable solution together with a medicant, as described above, for delivery by intravascular infusion or bolus injection into a mammal, such as a human. The solution is thus suitably balanced, osmotically (e.g., about 0.15 M saline) and with respect to pH, typically between pH 7.2 and 7.5; preferably the solution further comprises a buffer, such as a phosphate buffer (e.g., in a phosphate buffered saline solution). The solution is formulated to deliver a dose rate of about 0.075 to 1500 micrograms of potassium channel activator per kilogram body mass in a pharmaceutically acceptable fluid volume over a maximum of about thirty minutes. For human subjects, the solution is preferably formulated to deliver a dose rate of about 0.075 to 150 micrograms of potassium channel activator per kilogram body mass in a pharmaceutically acceptable fluid volume over a period of up to about thirty minutes
The invention also relates to a kit for enhancing the delivery of a medicant to an abnormal brain region and/or to a malignant tumor The kit is an assemblage of materials or components, including a potassium channel activator, other than bradykinin or a bradykinin analog, as described above In addition, the kit contains instructions for using the potassium channel activator to enhance the permeability of abnormal microvascular, including neomicrovasculature, to a medicant in general, or alternatively, to a particular medicant Optionally, the kit also contains other components, such as a particular medicant in any pharmaceutically acceptable formulation, or paraphernalia for injection or infusion, for example syringes, infusion lines, clamps, and/or infusion bags/bottles, which can contain a pharmaceutically acceptable infusible formulation of the potassium channel activator with or without a particular medicant also contained therein The materials or components assembled in the kit can be provided to the practitioner stored in any convenient and suitable ways that preserve their operability and utility For example the components can be in a dissolved, dehydrated, or lyophilized form, they can be provided at room, refrigerated or frozen temperatures
The foregoing descriptions of the methods and kits of the present invention are illustrative and by no means exhaustive The invention will now be described in greater detail by reference to the following non-limiting examples
EXAMPLES
Example 1 Methods
Malignant Cell Line and Tumor Implantation A rat glioma cell line, RG2, was used for implantation of experimental brain tumors in female Wistar rats The techniques for RG2 cell propagation and maintenance in tissue culture have been described (Sugita, M and Black,
K L , Cyclic GMP-specific phosphodiesterase inhibition and intracarotid bradykinin infusion enhances permeability into brain tumors, Cancer Res 58(5) 914-20 [1998], Inamura et al
[1994], Nakano, S et al, Increased brain tumor microvessel permeability after intracarotid bradykinin infusion is mediated by nitric oxide, Cancer Res 56( 17) 4027-31 [1996]) Briefly, RG2 cells derived from a rat glioma are kept frozen until use, then are thawed and maintained in a monolayer culture in F12 medium with 10% calf serum In some experiments C6 glioma cells were used The Wistar rats (approximately 140-160 g body weight) were anesthetized with intra-peritoneal ketamine (50 mg/kg), and glial cells (1 x 105) were implanted into the right hemisphere, but not the contralateral hemisphere, by intracerebral injection suspended in 5 μL F12 medium (1-2% methylcellulose) by a Hamilton syringe The implantation coordinates were 3 -mm lateral to the bregma and 4 5 mm deep to the dural surface
Intracarotid Infusion of Potassium Channel Activators Seven days after implantation of RG2 cells, the rats were anesthetized as described above and prepared for permeability studies Animals were infused with either NS-1619 (a selective large conductance Ca2+-activated K+ channel activator, RBI, Natick, MA) or minoxidil sulfate (a KATP channel activator) into the right carotid artery at a dose rate of 5 3 μg kg"1 min"1 (in 53 3 μL/min) for 15 minutes, in an infusion vehicle of PBS, pH 7 4, 5% (v/v) ethanol Ethanol (25% [v/v]) was used to dissolve the potassium channel activators before dilution in PBS For blood volume studies, 5 and 14 minutes after the start of the intracarotid infusion of potassium channel activator compounds, [14C] Dextran (100 μCi/kg, Dupont-New England Nuclear Co , Boston, MA) was injected as an intravenous bolus and maintained for 1 minute and 10 minutes to obtain two different time points For regional permeability studies, 5 minutes after the start of the intracarotid infusion of vasoactive compounds, 100 μCi/kg of [14C]α-aminoisobutyric acid (Dupont-New England Nuclear Co., Boston, MA) was injected as an intravenous bolus into the right femoral vein A peristaltic withdrawal pump was used to withdraw femoral arterial blood at a constant rate of 0.083 rnL/min immediately after the injection of the tracer to determine serum radioactivity Fifteen minutes after the intracarotid infusion, rat decapitated and the brain rapidly removed and frozen for quantitative autoradiography
In permeability studies with nitric oxide (NO) donors, intracarotid infusion of NO donors was done as described above at a dose rate of 2 66 μg min"1 kg"1 for 15 minute Nitric oxide donors included sodium 2-(N,N-diethylamino)-diazenolate-2-oxide (DEA/NO), a nitric oxide-donor with a half-life of 2 1 min or PAPA/NO ([Z]-l-[N-[3-Amino propyo]-N-[N-propyo amino] diazen-l-ium-l,2-diolate, (Alexis Corp ), a nitric oxide-donor with a half-life of 15 minutes DEA/NO or PAPA/NO were dissolved in PBS and administered to RG2 glioma-bearing rats to determine permeability (K,) of [14C]-AIB without affecting the physiological parameters [14C]-AIB was administered intravenously as a bolus and K, determined Physiological parameters were monitored during the experiments
Unidirectional Transport Constant (K,) The unidirectional transfer constant K, for [14C]α- aminoisobutyric acid (AIB) was measured in normal tissue and tumor tissue as an indicator of permeability across the blood-tumor and blood-brain barriers Quantitative autoradiography was used to obtain K, values (μL g"1 min"1) The initial rate for blood-to-brain transfer was calculated using a previously described equation (Ohno, K, et al, Lower limits of cerebrovascular permeability to nonelectrolytes in the conscious rat, Am J Physiol 235(3) H299-307, [1978], Inamura, T , et al. , Bradykinin selectively opens blood-tumor barrier in experimental brain tumors, J. Cereb. Flow Metab 14(5) 862-70 [1994]) Quantitative data were analyzed using, two group t-test and two-group Fishers-exact test of equal proportions or equal means (equal numbers) at 90% power requires a minimum of 6 animals in each group to achieve statistical significance Multiple treatment groups were compared with control group by ANOVA and P values determined by post-hoc Bonferroni test
Dose-dependence studies NS-1619 was dissolved in 25% ethanol and diluted with PBS to obtain various concentrations for infusion NS-1619 was administered by intracarotid infusion (dose rates 0, 13, 26 5, 53, 80, 100 and 110 μg kg"1 min"1, all at 53 3 μL/min) to RG2 glioma-bearing rats to determine a dose that produces increased permeability (K,) of [14C]-AIB, which was administered intravenously K, was determined as described above Physiological parameters were monitored during the experiments
Inhibition studies Since NS-1619 increased permeability, the specificity of its effect was examined using the specific KCa channel inhibitor, iberiotoxin (RBI, Natick, MA, ([IBTX] was diluted in saline to a final stock concentration of 100 μg/mL) EBTX (2 3 μg kg"1 min"1) was co-infused with NS-1619 (26 5 μg kg"1 min"1) to block KCa channel-induced permeability in abnormal capillaries in the RG2 glioma rat model Seventeen rats were used for these studies (3 vehicle-only control [i e , PBS + 5% (v/v) ethanol], 3 IBTX, 8 NS-1619, 3 NS-1619 + IBTX) In other experiments, IBTX (02 μg min"1 kg"1 for 15 min) was co-infused with the nitric oxide donor DEA/NO (2 66 μg min"1 kg"1 for 15 min) to block KCa channel-induced permeability in abnormal capillaries in the RG2 glioma rat model Thirteen rats were used for these studies (4 vehicle-only control, 3 DEA/NO, 3 PAPA/NO, and 3 DEA/NO+IBTX)
In other in vivo permeability experiments using Wistar rats harboring intracranial RG2 tumors, as described above, the unidirectional transport constant (K,) for [14C]-AIB to the tumor center and tissue surrounding tumor was determined after intracarotid infusion of bradykinin (10 μg min"1 kg"1 for 15 min) or minoxidil sulfate (26 6 μg min"1 kg"1 for 15 min) or co-infusion of bradykinin with minoxidil sulfate for 15 min Physiological parameters were monitored during the experiments The selective KATP inhibitor, glibenclamide (13 3 μg min"1 kg"1 for 15 min) was used to determine whether inhibition of KATP channel blocks bradykinin-inducible or minoxidil sulfate-inducible increases in blood-tumor barrier permeability
In other experiments, RG2 glioma cells were implanted into the right hemisphere of rat brains as described above Seven days after implantation, regional permeability study was performed by intracarotid infusion of octobromo-cyclic GMP (8Br-cGMP, 16 7 μg kg"1 min"1), an activator of cyclic GMP-dependent protein kinase (PKG), without or with PKG inhibitors
(KT5823, 6 77 μg kg"1 min"1, or Rp-pCPT-cGMP, 60 μg kg"1 min"1) Saline or 2% DMSO was infused into rats in a control group At the termination of the study, rats were killed under anesthesia, and brains were removed for measurement of K, for [14C]-AJB (μL g"1 min"1) Western blot analysis was also used to compare PKG expression levels in normal brain and tumor tissue
Quantitative autoradiography and calculations of K, for [14C]-AIB were performed as described above, according to Ohno et al. (1978)
Immunohistochemical analysis for K and KΛTP channels Brain sections (12 μm thick) obtained from the permeability studies were incubated with 1 100 dilution of affinity-purified KCa channel antibody (Alomone Labs, Jerusalem, Israel) for 1 hour, and biotinylated horse anti-mouse immunoglobin (Vector Laboratories, Burlingame, CA) for 30 minutes After washing 3 times with PBS, the peroxidase sites were visualized using an avidin biotinylated enzyme complex (ABC) kit (KCa) For immunolocalization of KATP, tissue sections as described above were incubated overnight at 4°C with rabbit anti-mouse Kir 6 2 antiserum (1 200 dilution, anti-serum generously provided by Dr Susumu Seino), which antiserum is cross-reactive with human Kir 6 2 and was prepared as previously described (Suzuki, M et al, Immuno-localizatwn of sulphonylurea receptor 1 in rat pancreas, Diabetologia 42(10) 1204-1 1 [1999]) Kir 6 2 is a potassium inward rectifier (Kir) protein component of mammalian KATP Visualization of bound rabbit anti-mouse Kir 6 2 antibody was done after washing 3 times with
PBS and binding with secondary antibody (rhodamine-labeled anti-rabbit IgG [1.200 dilution], Santa Cruz Biotechnology, Ine , Santa Cruz, CA) as directed by the manufacturer, and examining with a confocal fluorescence microscope
Transmission Electron Microscopy f TEM) Rat brain tissue was prepared for TEM analysis by the following procedures After experiments involving intracarotid infusion of potassium channel activators, inhibitors, horse radish peroxidase (HRP), and/or vehicle control buffers, rats were infusion-fixed from the heart first with PBS (50-100 mL) followed with 1 0% glutaraldehyde (250 mL) The brain tissue was cut cronally to expose the tumor region, and the region of interest was selected, sliced into small pieces of about 1 mm thickness, and was immersion-fixed in 1 0% glutaraldehyde, at 4°C for 2 hours The sample was rinsed in 5% sucrose / 0 1 M PBS at 4°C, overnight, with continuous shaking and changes of the solution The samples then were immersion-fixed with 1% OsO4 at 4°C for 2 hours with continuous shaking The sample was then dehydrated with an increasing concentration (50-100%) of ethanol at 4°C for 15 minutes with constant shaking and frequent changes of solution Samples were infiltrated with propylene oxide, and then with epon at room temperature with shaking Embedding was done at 60°C over 48 hours Semi-thin sections (about 30-50 μm thick) were taken with a microtome equipped with a diamond-edge blade Finally, ultrathin sections (about 1-10 nm thick) were prepared with a microtome and stained with uranyl acetate for 45-60 minutes, lead citrate for 5 minutes and processed for TEM analysis T e n capillaries in each treatment group were selected and photographed at low magnification (total 90 capillaries) The basic morphology of the capillaries was examined by way of measuring and calculating the following parameters which represent the basic profile of the capillary abluminal circumference, luminal circumference, total area of the capillary (excluding nuclei and vacuoles), and mean thickness of the capillary
In a separate study, the density of the vesicles was determined for three vessels selected from each rat (each treatment group contained 5-6 rats) by taking from each blood vessel four electron micrographs as a test zone at high magnification The test zones were selected randomly, such as at 3, 6, 9, and 12 o'clock on the EM screen The areas of the test zones were measured, and the number of vesicles was counted by a person who was not aware of the background of the micrographs Vesicular density was expressed as the number of vesicles per square micrometer of cytoplasm The proportion of the total vesicular area to the cytoplasmic area was also determined using the same micrographs total vesicular area was measured and the proportion to the cytoplasm which included those vesicles was calculated and expressed as percentage The mean diameter of the vesicles was also calculated
Morphological changes resulting from the treatments were examined by selecting six tight junctions from each rat (five rats were in each treatment group for a total 270 tight junctions) The tight junctions were photographed at high magnification The analysis of tight junction was by comparing "cleft index" and "cleft area index" for each tight junction The cleft index was expressed by the following formula (length of unfused segments) - (length of junctions) (Stewart, 1987) The cleft area index was expressed by the formula (area of unfused segments) - (length of junction)
Temporary Middle Cerebral Artery fMCA) occlusion MCA occlusion was carried out as described by Liu, Y et al, The time course of glucose metabolism in rat cerebral ischemia with middle cerebral artery occlusion-reperfuswn model and the effect ofMK-80, Neurological Research 18 (6) 505-508 (1996) with slight modifications Briefly, the right MCA was occluded temporarily with a silicone rubber cylinder inserted from the bifurcation of the intracarotid artery after ligation of ipsilateral common and external carotid arteries The cylinder is made of 4-0 nylon 17 mm long surgical thread, coated with silicone mixed with a hardener to grade the diameter of the distal 5 mm to 0 25-0 30 mm The thread was inserted into the intracarotid artery through the external carotid artery near the bifurcation and ligated at the point of insertion The silicone rubber cylinder reached the proximal portion of the anterior cerebral artery The origin of the right MCA and posterior-communicating artery were occluded by the thread Following the surgery, animals were immobilized by means of loose fitting Pasteur casts and allowed to recover from anesthesia The thread was pulled out from the intracarotid artery after 1 hour or 2 hours of ischemia to allow reperfusion of the ischemic tissue with blood Potassium channel activator(s) and/or inhibitor(s) were injected by intracarotid infusion after 45 minutes of reperfusion for an additional 15 minutes
In some experiments, Evans blue dye was injected intravenously after 50 minutes of reperfusion (five minutes after commencing administration of potassium channel activator and/or inhibitor) Ten minutes after injection of Evans blue, the rat was perfused with 200 mL PBS through the heart to wash off excess Evans blue dye from the brain microvessels
Example 2 Results
Potassium Channel Activators Selectively Increase Transport Across the Blood-tumor Barrier
When Wistar rats bearing implanted glioma cells were infused with either NS-1619 or minoxidil sulfate, at 7 5 μg kg"1 min"1 for 15 minutes, the unidirectional transport constant K, for [14C]α-aminoisobutyric acid (AIB) was significantly increased by NS-1619 and minoxidil sulfate with respect to transport across the neovasculature forming the blood-tumor barrier, but not with respect to transport across normal brain microvasculature (Figure 12A and Figure 12B) These results demonstrated that activation of potassium calcium channels selectively increases the permeability of molecules across the capillaries of solid malignant tumors compared to capillaries supplying normal brain tissue
Increasing the dose of NS-1619 resulted in an increase in the unidirectional transfer constant K, for [14C]α-aminoisobutyric acid in RG2 glioma capillaries in a dose-dependent manner (Figure 13) At higher doses (100 and 110 μg/kg/min) a significant drop in the arterial blood pressure of the rats was observed The numbers of rats used in each group is shown in parentheses in Figure 13 The specificity of this effect was demonstrated, because the ability of NS-1619 to increase the unidirectional transfer constant K, for [14C]α-aminoisobutyric acid was inhibited by the K^-channel-specific inhibitor iberiotoxin (IBTX, Figure 14) The K, was determined in RG2 tumor-bearing rats using [14C]-ATB with NS-1619 (26 5 μg min"1 kg"1 ) with or without IBTX (2 3 μg kg"1 min"1, n=3), for 15 minutes Increase of K, in response to NS-1619 infusion ( n=8,
** P<0 001 compared with PBS with or without 5% ethanol) was attenuated by IBTX co-treatment IBTX alone at the dose investigated did not affect the brain-tumor barrier permeability of abnormal capillaries However, IBTX significantly (n=3, ** P<0 001 compared with NS-1619-treated group) decreased NS-1619-induced increase of permeability (K,), indicating a potassium channel-specific effect Controls receiving PBS plus 5% ethanol were indistinguishable from controls receiving PBS minus ethanol
A comparable increase in blood-tumor barrier permeability was obtained when the soluble guanylyl cyclase activator YC-1 was administered (Figure 1) IBTX and a selective inhibitor of soluble guanylyl cyclase, lH-[l,2,4]oxadiazolo[4,3-a]quinozalin-l-one (ODQ), attenuated the enhancement of K, by YC-1, showing it was KCa-mediated
Infusion of minoxidil sulfate resulted in a significant increase in K, (28 3 ± 6 0 μL/g/min, P<0 001), compared to the vehicle control group (Figure 7) This increase in blood-tumor permeability was attenuated by the KATP channel inhibitor glibenclamide, which significantly attenuated (K, = 12 7 ± 2 0 μL/g/min) the effect of minoxidil sulfate Thus, the K, increases with minoxidil sulfate were due to activation of KATP channels, which selectively enhanced blood-tumor barrier permeability The increase in permeability resulting from treatment with minoxidil sulfate was comparable to that caused by the KCa activator bradykinin (K, = 34 ± 8 0 μL/g/min), and a significant additive effect on permeability (K, = 42 ± 6 μL/g/min, P<0 05) was achieved with combination treatment with bradykinin and minoxidil sulfate as compared to individual treatment groups (Figure 7)
The co-infusion with glibenclamide failed to block bradykinin-inducible K, increase (Figure 8) is consistent with other studies showing bradykinin does not modulate KATP channels However, the K^ channel antagonist, IBTX, significantly attenuated bradykinins effect (K, = 177 ± 6 μL /g/min, P<0 01) indicating that the K, increase was due to activation of K^ and not KATP channels by bradykinin (Figure 8) Minoxidil sulfate-induced effect was significantly attenuated by glibenclamide (K, = 12.7
± 2 μL/g/min; P<0.001; Figure 9) However, this effect was not blocked by IBTX indicating that KATP channels mediate minoxidil sulfate-induced permeability. These results imply independent pathways for blood-tumor barrier permeability, one regulated by K^ and the other by KATP channels in the microcapillary of the brain tumor.
Nitric oxide donors also increased the permeability of the blood-tumor barrier Figure 2 shows that RG2 glioma tissue was stained in vivo by Evans blue stain (MW 960 82) in DEA/NO-treated Wistar rats (Figure 2B) compared to PBS-treated controls (Figure 2 A) These results were comparable to the results obtained by in vivo staining by Evans blue using NS-1619 or YC-1 (data not shown).
Figure 3 shows that IC, increased markedly in response to DEA/NO and PAPA NO (K, = 32 ± 9.0 μL g"1 min"1 and 36.4 ± 4.0 μL g"1 min"1, respectively) compared to controls (saline). However, co-treatment with IBTX attenuated the effect of infusions of DEA/NO (K, = 15.3 ± 2.3 μL g"1 min-1; P< 0.001) or PAPA/NO (K, = 15.3 ± 1.7 μL g"1 min"1; P< 0 001) and , showing that the effect was K^-mediated. IBTX alone at the dose investigated did not affect the brain-tumor barrier permeability of abnormal capillaries.
Western blots revealed PKG expression in both normal brain and tumor tissue PKG activator 8Br-cGMP increased IC, values compared with vehicle-only controls (K, = 22 1 ± 6.6 μL g"1 min"1 versus 14.2 ± 5.1 μL g"1 min"1, p<0.05). These results imply that endogenous production of cGMP, which activates PKG, selectively increases blood-tumor barrier permeability, presumably by PKG-mediated activation of KCa
Immunohistochemical Analysis Shows Potassium Channels Are More Abundant in Neovasculature and Malignant Cells Compared to Normal Tissue. Kca channel protein was immunolocalized using a specific antibody as described above.
Immunohistochemical analysis showed that K^ channels were selectively increased in tumor tissue and tumor capillaries in RG2 bearing rat brain sections, compared to sections of normal contralateral tissue (Figure 15). Similarly, immunolocalization of KATP using a KATP-specific antibody showed that KATP channels were selectively increased in tumor tissue and tumor capillaries in RG2 tumor-bearing and C6-tumor-bearing rat brain sections (Figure 16). These immunohistochemical results are consistent with the permeability data in which activation of KCa channel by NS-1619, or KATP by minoxidil, selectively opened the blood-tumor barrier
Together, the permeability and immunohistochemical data demonstrate that compounds that activate potassium channels can be used to selectively increase delivery of anti-tumor compounds to malignant tumor tissue
Transmission Electron Microscopic studies Transmission electron microscopic (TEM) studies revealed accelerated formation and movement of pinocytotic vesicles along the luminal-abluminal axis of capillary endothelium, as well as tumor cells, following intracarotid infusion of KCa activators NS-1619 and bradykinin, compared to a PBS control (Figure 4)
Quantitative analysis showed that both bradykinin and NS-1619 significantly increased the number of vesicles per unit surface area of nucleus-free cytoplasm, compared to the PBS control The pinocytotic vesicles had an average diameter of 75-80 nm Invagination of luminal membrane of tumor capillary endothelial cells gives rise to arrays of pinocytotic vesicles, which migrate along a luminal-abluminal axis in endothelial cytoplasm (E) These vesicles dock and fuse with the basement membrane, releasing their contents on abluminal side of endothelial cell membrane Cells from the treatment group appear to have intact basal membrane (BM) and endothelial tight junctions Transport pinocytotic vesicles (P V) are present near luminal (L) as well as abluminal (Ab) areas (Figure 4) Cells from gliomas after infusion of either bradykinin or NS-1619 have intact basal membrane and endothelial tight junctions (TJ) (E g , Figure 5)
Additional TEM studies revealed the presence of electron dense horse radish peroxidase (HRP, protein marker with a molecular weight of about 40,000) within the arrays of pinocytotic vesicles in endothelial cells of brain tumor capillaries and showed HRP being transported from the luminal to the abluminal area of the endothelium to the tumor cells after the IC^ activator bradykimn was co-infused with HRP into rats with RG2 tumors (Figures 5 and 6) This evidence of HRP-laden transport vesicles implies that transendothelial vesicular transport is a significant cellular mechanism for drug delivery to the tumor cells
Additional TEM studies with tumor-bearing rats infused with the potassium channel activator bradykinin, and intravenously injected with the tracer horseradish peroxidase (HRP), revealed the presence of electron dense HRP within the arrays of pinocytotic vesicles in endothelial cells of brain tumor capillaries (Figure 5, P = plasma membrane) Following bradykinin infusion there was an increase in the number of HRP-laden vesicles (observed in the tumor cells compared to PBS-treated tumor cells Figure 6, arrows), implying that vesicular transport is a primary cellular mechanism for drug delivery to the tumor cells, once the drug molecules cross the capillary endothelium into the brain tumor mass
No significant difference was seen in any of the examined morphological parameters between the vehicle control and the bradykinin (BK) infusion group, but there was a significant difference in these parameters in comparing the different kinds of tissue examined Abluminal and luminal circumference, area of the capillary, and mean thickness of the capillary of RG2 tumor and C6 tumor were significantly larger than those of normal brain basal ganglia However, a significant increase in density and proportion of vesicle area in the basal ganglia was observed inbradykinin-treated rats in RG2 and C6 tumor area as compared to the untreated rats (Table 1)
Table 1 Density and proportion of the area of vesicles to endothelial cytoplasm
Tissue3 Infusion33 Density of the vesicles Total area of the vesicles (%)c (number/m2)b
BG PBS 2 02 ± 0 35 (n = 5) 0 65 ± 0 26 (n = 6)
BK 1 75 ± 0 90 (n = 5) 0 54 ± 0 35 (n = 6)
RG2 tumor PBS 3 29 ± 0 66 (n = 5) 1 30 ± 0 36 (n = 6)
BK 9 88 ± 2 93 d (n = 6) 3 71 ± 0 96d (n = 6)
C6 tumor PBS 5 46 ± 3 29 (n = 5) 1 34 ± 0 57 (n = 6) B BKK 11 90 ± 2 40d (n = 5) 4 31 ± 1 34d (n = 6)
" BG = basal ganglia
"* PBS = phosphate buffer saline vehicle control, BK = PBS + bradykinin b Values are means SD, n = number of rats Q Values are means SD, n = number of capillaries d Significant difference from saline control group at p < 0 01 Potassium channel activators increase permeability in abnormal brain capillaries of ischemic brain region.
In preliminary MCA occlusion/reperfusion studies it was shown that the K^., activator bradykinin increases permeability in infarct tissue after transient-ischemia. NOS activity was significantly elevated in transient ischemia-reperfusion rat models (data not shown), corresponding to the ability of bradykinin to increase permeability in ischemic capillaries (Figure 10B). As shown in Figure 10B, the permeability of microvasculature in ischemic brain regions exhibited enhanced responsiveness to potassium channel activator after two hours of MCA occlusion, compared to normal brain microvasculature (i.e., contralateral tissue). In contrast, Figure 10A shows that the permeability of brain microvasculature was not responsive to a potassium channel activator after only one hour of MCA occlusion. This indicates that properties of the microvasculature are changed by prolonged ischemia as the tissue becomes increasingly abnormal.
Using the ischemia-reperfusion rat model, it was directly observed that enhanced permeability to Evans blue dye was regulated, by potassium channels in ischemic brain regions.
In Figure 11, coronal brain sections show Evans blue (EB) staining within the tumor tissue. The uptake of Evans blue dye is a known qualitative measure of blood-brain barrier/blood-tumor barrier permeability. Rats subjected to 2 hour of MCA occlusion followed by 1 hour of reperfusion did not show an increase in permeability in ischemic areas after PBS vehicle infusion (Figure 11). In contrast, rats subjected to similar ischemic conditions followed by intra arterial infusion of activators of K^ channels, for example bradykinin or NS-1619 (Figure 11) or nitric oxide donors (data not shown), enhanced permeability to the dye. The increased permeability to the dye produced by the potassium channel activators was diminished by co-infusion with the specific Kca inhibitor IBTX, indicating the specific involvement of K^ (Figure 11).

Claims

IN THE CLAIMS
1. A method of delivering a medicant to an abnormal brain region in a mammalian subject, comprising: administering to a mammalian subject having an abnormal brain region a potassium channel activator selected from the group consisting of
(A) activators of soluble guanylyl cyclase; and
(B) activators of cyclic GMP-dependent protein kinase, under conditions and in an amount sufficient to increase the permeability to the medicant of a capillary or arteriole delivering blood to cells of the abnormal brain region; and administering to the subject simultaneously or substantially simultaneously with the potassium channel activator the medicant, so that the medicant is delivered selectiveK' to the cells of the abnormal brain region compared to normal brain regions.
2. The method of Claim 1, wherein the abnormal brain region is a region of brain tissue physiologically affected by injury, trauma, infection, stroke, or ischemia.
3. The method of Claim 1 , wherein the abnormal brain region is a region of benign or malignant tumor tissue.
4. The method of Claim 1, wherein the activator of guanylyl cyclase is nitric oxide or a nitric oxide donor.
5. The method of Claim 4, wherein the nitric oxide donor is selected from the group consisting of organic nitrate compounds, iron nitrosyl compounds, S-nitrosothiol compounds, sydnonimine compounds, and NONOate compounds.
6. The method of Claim 5, wherein the organic nitrate compound is glyceryl trinitrate, nitroglycerin, pentaerythrityl tetranitrate, isosorbide dinitrate, or isosorbide 5-mononitrate.
7. The method of Claim 5, wherein the iron nitrosyl compound is sodium nitroprusside.
8. The method of Claim 5, wherein the sydnonimine compound is molsidomine, linsidomine, or pirsidomine.
9. The method of Claim 5, wherein the S-nitrosothiol compound is S-nitroso-N-acetyl-D,L-penicillamine, S-nitrosoglutathione, S-nitrosoalbumin, or S-nitrosocysteine.
10. The method of Claim 5, wherein the NONOate compound is diethylamine-NONOate, diethylene triamine-NONOate, dipropylenetriamine-NONOate, spermine-NONOate, propylamino-propylamine-NONOate, MAHMA-NONOate, piperazi-NONOate, proli-NONOate, sulfo-NONOate, Angelis salt, or sulfite NONOate.
11. The method of Claim 1, wherein the activator of cyclic GMP-dependent protein kinase is selected from the group consisting of octobromo-cyclic GMP and dibutyryl cyclic GMP.
12. The method of Claim 1, wherein said mammal is a human, non-human primate, canine, feline, bovine, porcine, ovine, mouse, rat, gerbil, hamster, or rabbit.
13. The method of Claim 1, wherein the medicant is a therapeutic cytotoxic agent, DNA expression vector, viral vector, protein, oligonucleotide, nucleotide analog, antimicrobial agent, interferon, cytokine, cytokine agonist, cytokine antagonist, immunotoxin, immunosuppressant, boron compound, monoclonal antibody, adrenergic agent, anticonvulsant, ischemia-protective agent, anti-trauma agent, anticancer chemotherapeutic agent, or diagnostic agent.
14 The method of Claim 13, wherein the diagnostic agent is an imaging or contrast agent
15 The method of Claim 13, wherein the diagnostic agent is a radioactively labeled substance, a gallium-labeled substance, or a contrast agent selected from the group consisting of ferrous magnetic, fluorescent, luminescent, and iodinated contrast agents
16 The method of Claim 1, wherein the medicant is a N-methyl-D-aspartate (NMDA) receptor antagonist, antibiotic, interleukin-2, or transforming growth factor-β, cisplatin, carboplatin, tumor necrosis factor-α, methotrexate, 5-fluorouracil, amphotericin, daunorubicin, doxorubicin, vincristine, vinblastine. busulfan, chlorambucil, cyclophosphamide, melphalan, or ethyl ethanesulfonic acid
17 The method of Claim 13, wherein the viral vector is an adenovirus-derived vector or herpes simplex virus-derived vector
18 The method of Claim 1 , wherein administering the potassium channel activator is by intravenous or intra-arterial infusion or injection
19 The method of Claim 1, wherein administering the potassium channel activator is by intracarotid infusion or injection
20 The method of Claim 1, wherein the potassium channel activator is administered to the mammalian subject by a bolus injection
21 The method of Claim 1, wherein the potassium channel activator is administered to the mammalian subject in an amount from about 0 075 to 1500 micrograms per kilogram body mass
22 The method of Claim 21, wherein the potassium channel activator is administered to the subject in an amount from about 0 075 to 150 micrograms per kilogram body mass
23 The method of Claim 1, wherein the potassium channel activator is administered to the mammalian subject at a dose rate of about 0 075 to about 100 μg kg"1 min"1 for up to about 30 minutes
24 The method of Claim 23, wherein the potassium channel activator is administered to the mammalian subject at a dose rate of about 0 075 to about 15 μg kg"1 min"1
25 A method of selectively delivering a medicant to an abnormal brain region in a mammalian subject, comprising administering to a mammalian subject having an abnormal brain region a potassium channel activator selected from the group consisting essentially of nitric oxide, nitric oxide donors and activators of cyclic GMP-dependent protein kinase, under conditions and in an amount sufficient to increase potassium flux through a calcium-activated or ATP-sensitive potassium channel in an endothelial cell membrane of a capillary or arteriole delivering blood to cells of the abnormal brain region, whereby the capillary or arteriole is made more permeable to the medicant, and administering to the subject simultaneously or substantially simultaneously with the potassium channel activator the medicant, so that the medicant is delivered selectively to the cells of the abnormal brain region compared to normal brain regions
26 The method of Claim 25, wherein the abnormal brain region is a region of brain tissue physiologically affected by injury, trauma, infection, stroke, or ischemia
27 The method of Claim 25, wherein the abnormal brain region is a region of benign or malignant tumor tissue
28 The method of Claim 25, wherein the nitric oxide donor is selected from the group consisting of organic nitrate compounds, iron nitrosyl compounds, S-nitrosothiol compounds, sydnonimine compounds, and NONOate compounds
29 The method of Claim 28, wherein the organic nitrate compound is glyceryl trinitrate, nitroglycerin, pentaerythrityl tetranitrate, isosorbide dinitrate, or isosorbide 5-mononitrate
30 The method of Claim 28, wherein the iron nitrosyl compound is sodium nitroprusside
31 The method of Claim 28, wherein the sydnonimine compound is molsidomine, linsidomine, or pirsidomine.
32 The method of Claim 28, wherein the S-nitrosothiol compound is
S-nitroso-N-acetyl-D,L-penicillamine, S-nitrosoglutathione, S-nitrosoalbumin, or S-nitrosocysteine
33 The method of Claim 28, wherein the NONOate compound is diethylamine-NONOate, diethylene triamine-NONOate, dipropylenetriamine-NONOate, spermine-NONOate, propylamino-propylamine-NONOate, MAHMA-NONOate, piperazi-NONOate, proli-NONOate, sulfo-NONOate, Angelis salt, or sulfite NONOate
34. The method of Claim 25, wherein the activator of cyclic GMP-dependent protein kinase is selected from the group consisting of octobromo-cyclic GMP and dibutyryl cyclic GMP.
35 The method of Claim 25, wherein said mammal is a human, non-human primate, canine, feline, bovine, porcine, ovine, mouse, rat, gerbil, hamster, or rabbit
36 The method of Claim 25, wherein the medicant is a therapeutic cytotoxic agent, DNA expression vector, viral vector, protein, oligonucleotide, nucleotide analog, antimicrobial agent, interferon, cytokine, cytokine agonist, cytokine antagonist, immunotoxin, immunosuppressant, boron compound, monoclonal antibody, adrenergic agent, anticonvulsant, ischemia-protective agent, anti-trauma agent, anticancer chemotherapeutic agent, or diagnostic agent
37 The method of Claim 36, wherein the diagnostic agent is an imaging or contrast agent
38 The method of Claim 36, wherein the diagnostic agent is a radioactively labeled substance, a gallium-labeled substance, or a contrast agent selected from the group consisting of ferrous magnetic, fluorescent, luminescent, and iodinated contrast agents
39 The method of Claim 25, wherein the medicant is a N-methyl-D-aspartate (NMD A) receptor antagonist, antibiotic, interleukin-2, or transforming growth factor-β, cisplatin, carboplatin, tumor necrosis factor-α, methotrexate, 5-fluorouracil, amphotericin, daunorubicin, doxorubicin, vincristine, vinblastine, busulfan, chlorambucil, cyclophosphamide, melphalan, or ethyl ethanesulfonic acid
40 The method of Claim 36, wherein the viral vector is an adenovirus-derived vector or herpes simplex virus-derived vector
41 The method of Claim 25, wherein administering the potassium channel activator is by intravenous or intra-arterial infusion or injection
42 The method of Claim 25, wherein administering the potassium channel activator is by intracarotid infusion or injection
43 The method of Claim 25, wherein the potassium channel activator is administered to the mammalian subject by a bolus injection
44 The method of Claim 25, wherein the potassium channel activator is administered to the mammalian subject in an amount from about 0 075 to 1500 micrograms per kilogram body mass
45 The method of Claim 44, wherein the potassium channel activator is administered to the subject in an amount from about 0 075 to 150 micrograms per kilogram body mass
46 The method of Claim 25, wherein the potassium channel activator is administered to the mammalian subject at a dose rate of about 0 075 to about 100 μg kg"1 min"1 for up to about 30 minutes
47 The method of Claim 46, wherein the potassium channel activator is administered to the mammalian subject at a dose rate of about 0 075 to about 15 μg kg"1 min"1
48 A method of delivering a medicant to a malignant tumor in a mammalian subject, comprising administering to a mammalian subject having a malignant tumor a potassium channel activator selected from the group consisting of
(A) activators of soluble guanylyl cyclase, and
(B) activators of cyclic GMP-dependent protein kinase, under conditions and in an amount sufficient to increase the permeability to the medicant of a capillary or arteriole delivering blood to cells of the malignant tumor, and administering to the subject simultaneously or substantially simultaneously with the potassium channel activator the medicant, so that the medicant is delivered selectively to the malignant cells compared to non-malignant cells
49 The method of Claim 48, wherein the activator of soluble guanylyl cyclase is nitric oxide or a nitric oxide donor
50 The method of Claim 49, wherein the nitric oxide donor is selected from the group consisting of organic nitrate compounds, iron nitrosyl compounds, S-nitrosothiol compounds, sydnonimine compounds, and NONOate compounds
51 The method of Claim 50, wherein the organic nitrate compound is glyceryl trinitrate, nitroglycerin, pentaerythrityl tetranitrate, isosorbide dinitrate, or isosorbide 5-mononitrate
52 The method of Claim 50, wherein the iron nitrosyl compound is sodium nitroprusside
53 The method of Claim 50, wherein the sydnonimine compound is molsidomine, linsidomine, or pirsidomine
54 The method of Claim 50, wherein the S-nitrosothiol compound is S-nitroso-N-acetyl-D,L-penicillamine, S-nitrosoglutathione, S-nitrosoalbumin, or S-nitrosocysteine
55 The method of Claim 50, wherein the NONOate compound is diethylamine-NONOate, diethylene triamine-NONOate, dipropylenetriamine-NONOate, spermine-NONOate, propylamino-propylamine-NONOate, MAHMA-NONOate, piperazi-NONOate, proli-NONOate, sulfo-NONOate, Angelis salt, or sulfite NONOate
56 The method of Claim 48, wherein the activator of cyclic GMP-dependent protein kinase is selected from the group consisting of octobromo-cyclic GMP and dibutyryl cyclic GMP
57 The method of Claim 48, wherein the malignant tumor is a glioma, glioblastoma, oligodendroglioma, astrocytoma, ependymoma, primitive neuroectodermal tumor, atypical meningioma, malignant meningioma, neuroblastoma, sarcoma, melanoma, lymphoma, or carcinoma
58 The method of Claim 48, wherein the malignant tumor is contained in the skull, brain, spine, thorax, lung, peritoneum, prostate, ovary, uterus, breast, stomach, liver, bowel, colon, rectum, bone, lymphatic system, or skin, of said subject
59 The method of Claim 48, wherein said mammal is a human, non-human primate, canine, feline, bovine, porcine, ovine, mouse, rat, gerbil, hamster, or rabbit
60 The method of Claim 48, wherein the medicant is a therapeutic cytotoxic agent, DNA expression vector, viral vector, protein, oligonucleotide, nucleotide analog, antimicrobial agent, interferon, cytokine, cytokine agonist, cytokine antagonist, immunotoxin, immunosuppressant, boron compound, monoclonal antibody, adrenergic agent, anticonvulsant, ischemia-protective agent, anti-trauma agent, anticancer chemotherapeutic agent, or diagnostic agent
61 The method of Claim 60, wherein the diagnostic agent is an imaging or contrast agent
62 The method of Claim 60, wherein the diagnostic agent is a radioactively labeled substance, a gallium-labeled substance, or a contrast agent selected from the group consisting of ferrous magnetic, fluorescent, luminescent, and iodinated contrast agents
63 The method of Claim 48, wherein the medicant is a Nmethyl-D-aspartate
(NMDA) receptor antagonist, antibiotic, interleukin-2, or transforming growth factor-β, cisplatin, carboplatin, tumor necrosis factor- , methotrexate, 5-fluorouracil, amphotericin, daunorubicin, doxorubicin, vincristine, vinblastine, busulfan, chlorambucil, cyclophosphamide, melphalan, or ethyl ethanesulfonic acid
64 The method of Claim 60, wherein the viral vector is an adenovirus-derived vector or herpes simplex virus-derived vector
65 The method of Claim 48, wherein administering the potassium channel activator is by intravenous or intra-arterial infusion or injection
66 The method of Claim 48, wherein the tumor is an intracranial tumor and the potassium channel activator is administered by intracarotid infusion or injection
67 The method of Claim 48, wherein the potassium channel activator is administered to the mammalian subject by a bolus injection
68 The method of Claim 48, wherein the potassium channel activator is administered to the mammalian subject in an amount from about 0 075 to 1500 micrograms per kilogram body mass
69 The method of Claim 68, wherein the potassium channel activator is administered to the mammalian subject in an amount from about 0 075 to 150 micrograms per kilogram body mass
70 The method of Claim 48, wherein the potassium channel activator is administered to the mammalian subject at a dose rate of about 0 075 to about 100 μg kg"1 min"1 for up to about 30 minutes
71 The method of Claim 70, wherein the potassium channel activator is administered to the mammalian subject at a dose rate of about 0 075 to about 15 μg kg"1 min"1
72. A method of delivering a medicant to a malignant tumor in a mammalian subject, comprising: administering to the mammalian subject having a malignant tumor a potassium channel activator selected from the group consisting essentially of nitric oxide donors and activators of cyclic GMP-dependent protein kinase, under conditions and in an amount sufficient to increase potassium flux through a calcium-activated or ATP-sensitive potassium channel in an endothelial cell membrane of a capillary or arteriole delivering blood to malignant cells of the tumor, whereby the capillary or arteriole is made more permeable to the medicant; and administering to the subject simultaneously or substantially simultaneously with the potassium channel activator the medicant, so that the medicant is delivered selectively to the malignant cells compared to non-malignant cells.
73. The method of Claim 72, wherein the nitric oxide donor is selected from the group consisting of organic nitrate compounds, iron nitrosyl compounds, S-nitrosothiol compounds, sydnonimine compounds, and NONOate compounds.
74. The method of Claim 73, wherein the organic nitrate compound is glyceryl trinitrate, nitroglycerin, pentaerythrityl tetranitrate, isosorbide dinitrate, or isosorbide 5-mononitrate.
75. The method of Claim 73, wherein the iron nitrosyl compound is sodium nitroprusside.
76. The method of Claim 73, wherein the sydnonimine compound is molsidomine, linsidomine, or pirsidomine.
77. The method of Claim 73, wherein the S-nitrosothiol compound is S-nitroso-N-acetyl-D,L-penicillamine, S-nitrosoglutathione, S-nitrosoalbumin, or S-nitrosocysteine.
78. The method of Claim 73, wherein the NONOate compound is diethylamine-NONOate, diethylene triamine-NONOate, dipropylenetriamine-NONOate, spermine-NONOate, propylamino-propylamine-NONOate, MAHMA-NONOate, piperazi-NONOate, proli-NONOate, sulfo-NONOate, Angelis salt, or sulfite NONOate.
79. The method of Claim 72, wherein the activator of cyclic GMP-dependent protein kinase is selected from the group consisting of octobromo-cyclic GMP and dibutyryl cyclic GMP.
80. The method of Claim 72, wherein the malignant tumor is a glioma, glioblastoma, oligodendroglioma, astrocytoma, ependymoma, primitive neuroectodermal tumor, atypical meningioma, malignant meningioma, neuroblastoma, sarcoma, melanoma, lymphoma, or carcinoma.
81. The method of Claim 72, wherein the malignant tumor is contained in the skull, brain, spine, thorax, lung, peritoneum, prostate, ovary, uterus, breast, stomach, liver, bowel, colon, rectum, bone, lymphatic system, or skin, of said subject.
82. The method of Claim 72, wherein said mammal is a human, non-human primate, canine, feline, bovine, porcine, ovine, mouse, rat, gerbil, hamster, or rabbit.
83. The method of Claim 72, wherein the medicant is a therapeutic cytotoxic agent, DNA expression vector, viral vector, protein, oligonucleotide, nucleotide analog, antimicrobial agent, interferon, cytokine, cytokine agonist, cytokine antagonist, immunotoxin, immunosuppressant, boron compound, monoclonal antibody, adrenergic agent, anticonvulsant, ischemia-protective agent, anti-trauma agent, anticancer chemotherapeutic agent, or diagnostic agent.
84. The method of Claim 83, wherein the diagnostic agent is an imaging or contrast agent.
85. The method of Claim 83, wherein the diagnostic agent is a radioactively labeled substance, a gallium-labeled substance, or a contrast agent selected from the group consisting of ferrous magnetic, fluorescent, luminescent, and iodinated contrast agents.
86. The method of Claim 72, wherein the medicant is a Nmethyl-D-aspartate (NMDA) receptor antagonist, antibiotic, interleukin-2; or transforming growth factor-β, cisplatin, carboplatin, tumor necrosis factor-α, methotrexate, 5-fluorouracil, amphotericin, daunorubicin, doxorubicin, vincristine, vinblastine, busulfan, chlorambucil, cyclophosphamide, melphalan, or ethyl ethanesulfonic acid.
87. The method of Claim 83, wherein the viral vector is an adenovirus-derived vector or herpes simplex virus-derived vector.
88. The method of Claim 72, wherein administering the potassium channel activator is by intravenous or intra-arterial infusion or injection.
89. The method of Claim 72, wherein the tumor is an intracranial tumor and the potassium channel activator is administered by intracarotid infusion or injection.
90. The method of Claim 72, wherein the potassium channel activator is administered to the mammalian subject by a bolus injection.
91. The method of Claim 72, wherein the potassium channel activator is administered to the mammalian subject in an amount from about 0.075 to 1500 micrograms per kilogram body mass.
92. The method of Claim 91, wherein the potassium channel activator is administered to the mammalian subject in an amount from about 0.075 to 150 micrograms per kilogram body mass.
93 The method of Claim 72, wherein the potassium channel activator is administered to the mammalian subject at a dose rate of about 0 075 to about 100 μg kg-1 min"1 for up to about 30 minutes
94 The method of Claim 93, wherein the potassium channel activator is administered to the mammalian subject at a dose rate of about 0 075 to about 15 μg kg"1 min"1
95 A method of treating a malignant tumor in a human subject, comprising administering to a human subject having a malignant tumor a potassium channel activator, selected from the group consisting essentially of nitric oxide, nitric oxide donors, and activators of cyclic GMP-dependent protein kinase, under conditions and in an amount sufficient to increase the permeability to the medicant of a capillary or arteriole delivering blood to cells of the malignant tumor, and administering to the subject simultaneously or substantially simultaneously with the potassium channel activator the medicant, so that the medicant is delivered selectively to the malignant cells compared to non-malignant cells
96 The method of Claim 95, wherein the nitric oxide donor is selected from the group consisting of organic nitrate compounds, iron nitrosyl compounds, S-nitrosothiol compounds, sydnonimine compounds, and NONOate compounds
97 The method of Claim 96, wherein the organic nitrate compound is glyceryl trinitrate, nitroglycerin, pentaerythrityl tetranitrate, isosorbide dinitrate, or isosorbide 5-mononitrate
98 The method of Claim 96, wherein the iron nitrosyl compound is sodium nitroprusside
99 The method of Claim 96, wherein the sydnonimine compound is molsidomine, linsidomine, or pirsidomine
100 The method of Claim 96, wherein the S-nitrosothiol compound is S-nitroso-N-acetyl-D,L-penicillamine, S-nitrosoglutathione, S-nitrosoalbumin, or
S-nitrosocysteine
101 The method of Claim 96, wherein the NONOate compound is diethylamine-NONOate, diethylene triamine-NONOate, dipropylenetriamine-NONOate, spermine-NONOate, propylamino-propylamine-NONOate, MAHMA-NONOate, piperazi-NONOate, proli-NONOate, sulfo-NONOate, Angelis salt, or sulfite NONOate
102 The method of Claim 95, wherein the activator of cyclic GMP-dependent protein kinase is selected from the group consisting of octobromo-cyclic GMP and dibutyryl cyclic
GMP
103 The method of Claim 95, wherein the malignant tumor is a glioma, glioblastoma, oligodendroglioma, astrocytoma, ependymoma, primitive neuroectodermal tumor, atypical meningioma, malignant meningioma, neuroblastoma, sarcoma, melanoma, lymphoma, or carcinoma
104 The method of Claim 95, wherein the malignant tumor is contained in the skull, brain, spine, thorax, lung, peritoneum, prostate, ovary, uterus, breast, stomach, liver, bowel, colon, rectum, bone, lymphatic system, or skin, of said subject
105 The method of Claim 95, wherein the medicant is a therapeutic cytotoxic agent, DNA expression vector, viral vector, protein, oligonucleotide, nucleotide analog, antimicrobial agent, interferon, cytokine, cytokine agonist, cytokine antagonist, immunotoxin, immunosuppressant, boron compound, monoclonal antibody, adrenergic agent, anticonvulsant, ischemia-protective agent, anti-trauma agent, anticancer chemotherapeutic agent, or diagnostic agent
106 The method of Claim 105, wherein the diagnostic agent is an imaging or contrast agent
107 The method of Claim 105, wherein the diagnostic agent is a radioactively labeled substance, a gallium-labeled substance, or a contrast agent selected from the group consisting of ferrous magnetic, fluorescent, luminescent, and iodinated contrast agents
108 The method of Claim 95, wherein the medicant is a Nmethyl-D-aspartate
(NMDA) receptor antagonist, antibiotic, interleukin-2, or transforming growth factor-β, cisplatin, carboplatin, tumor necrosis factor-α, methotrexate, 5-fluorouracil, amphotericin, daunorubicin, doxorubicin, vincristine, vinblastine, busulfan, chlorambucil, cyclophosphamide, melphalan, or ethyl ethanesulfonic acid
109 The method of Claim 105, wherein the viral vector is an adenovirus-derived vector or herpes simplex virus-derived vector
110 The method of Claim 95, wherein administering the potassium channel activator is by intravenous or intra-arterial infusion or injection
111 The method of Claim 95, wherein the tumor is an intracranial tumor and the potassium channel activator is administered by intracarotid infusion
112 The method of Claim 95, wherein the potassium channel activator is administered to the mammalian subject by a bolus injection
113 The method of Claim 95, wherein the potassium channel activator is administered to the subject in an amount from about 0 075 to 150 micrograms per kilogram body mass
114 The method of Claim 95, wherein the potassium channel activator is administered to the subject at a dose rate of about 0 075 to about 15 μg kg"1 min"1
115 A method of treating a malignant tumor in a human subject, comprising administering to a human subject, having a malignant tumor, a potassium channel activator selected from the group consisting essentially of nitric oxide, nitric oxide donors, and activators of cyclic GMP-dependent protein kinase, under conditions and in an amount sufficient to increase potassium flux through a calcium-activated or ATP-sensitive potassium channel in an endothelial cell membrane of a capillary or arteriole delivering blood to malignant cells of the malignant tumor, whereby the capillary or arteriole is made more permeable to the medicant, and administering to the subject simultaneously or substantially simultaneously with the potassium channel activator the medicant, so that the medicant is delivered selectively to the malignant cells compared to non-malignant cells
116 The method of Claim 115, wherein the nitric oxide donor is selected from the group consisting of organic nitrate compounds, iron nitrosyl compounds, S-nitrosothiol compounds, sydnonimine compounds, and NONOate compounds
117 The method of Claim 116, wherein the organic nitrate compound is glyceryl trinitrate, nitroglycerin, pentaerythrityl tetranitrate, isosorbide dinitrate, or isosorbide
5-mononitrate
118 The method of Claim 116, wherein the iron nitrosyl compound is sodium nitroprusside
119 The method of Claim 116, wherein the sydnonimine compound is molsidomine, linsidomine, or pirsidomine
120 The method of Claim 116, wherein the S-nitrosothiol compound is S-nitroso-N-acetyl-D,L-penicillamine, S-nitrosoglutathione, S-nitrosoalbumin, or S-nitrosocysteine
121 The method of Claim 116, wherein the NONOate compound is diethylamine-NONOate, diethylene triamine-NONOate, dipropylenetriamine-NONOate, spermine-NONOate, propylamino-propylamine-NONOate, MAHMA-NONOate, piperazi-NONOate, proli-NONOate, sulfo-NONOate, Angelis salt, or sulfite NONOate.
122 The method of Claim 115, wherein the activator of cyclic GMP-dependent protein kinase is selected from the group consisting of octobromo-cyclic GMP and dibutyryl cyclic GMP.
123 The method of Claim 115, wherein the malignant tumor is a glioma, glioblastoma, oligodendroglioma, astrocytoma, ependymoma, primitive neuroectodermal tumor, atypical meningioma, malignant meningioma, neuroblastoma, sarcoma, melanoma, lymphoma, or carcinoma.
124 The method of Claim 115, wherein the malignant tumor is contained in the skull, brain, spine, thorax, lung, peritoneum, prostate, ovary, uterus, breast, stomach, liver, bowel, colon, rectum, bone, lymphatic system, or skin, of said subject
125 The method of Claim 115, wherein the medicant is a therapeutic cytotoxic agent, DNA expression vector, viral vector, protein, oligonucleotide, nucleotide analog, antimicrobial agent, interferon, cytokine, cytokine agonist, cytokine antagonist, immunotoxin, immunosuppressant, boron compound, monoclonal antibody, adrenergic agent, anticonvulsant, ischemia-protective agent, anti-trauma agent, anticancer chemotherapeutic agent, or diagnostic agent.
126 The method of Claim 125, wherein the diagnostic agent is an imaging or contrast agent
127 The method of Claim 125, wherein the diagnostic agent is a radioactively labeled substance, a gallium-labeled substance, or a contrast agent selected from the group consisting of ferrous magnetic, fluorescent, luminescent, and iodinated contrast agents
128 The method of Claim 115, wherein the medicant is a Nmethyl-D-aspartate (NMDA) receptor antagonist, antibiotic, interleukin-2, or transforming growth factor-β, cisplatin, carboplatin, tumor necrosis factor-α, methotrexate, 5-fluorouracil, amphotericin, daunorubicin, doxorubicin, vincristine, vinblastine, busulfan, chlorambucil, cyclophosphamide, melphalan, or ethyl ethanesulfonic acid
129 The method of Claim 125, wherein the viral vector is an adenovirus-derived vector or herpes simplex virus-derived vector
130 The method of Claim 115, wherein administering the potassium channel activator is by intravenous or intra-arterial injection
131 The method of Claim 115, wherein the tumor is an intracranial tumor and the potassium channel activator is administered by intracarotid infusion
132 The method of Claim 115, wherein the potassium channel activator is administered to the mammalian subject by a bolus injection
133 The method of Claim 115, wherein the potassium channel activator is administered to the subject in an amount from about 0 075 to 150 micrograms per kilogram body mass
134 The method of Claim 115, wherein the potassium channel activator is administered to the mammalian subject at a dose rate of about 0 075 to about 15 μg kg"1 min"1
135 A pharmaceutical composition comprising a combination of a potassium channel activator selected from the group consisting of activators of soluble guanylyl cyclase and activators of cyclic GMP-dependent protein kinase, formulated in a pharmaceutically acceptable solution together with a medicant for delivery by intravascular infusion or injection into a mammal
136 The pharmaceutical composition of Claim 135, wherein the solution is formulated to deliver a dose rate of about 0 075 to 1500 micrograms of the potassium channel activator per kilogram body mass in a pharmaceutically acceptable fluid volume over a maximum of about thirty minutes.
137 The pharmaceutical composition of Claim 135, wherein the solution is formulated to deliver a dose rate of about 0 075 to 150 micrograms of the potassium channel activator per kilogram body mass in a pharmaceutically acceptable fluid volume over a period of up to thirty minutes
138 The pharmaceutical composition of Claim 135, wherein the activator of soluble guanylyl cyclase is nitric oxide or a nitric oxide donor
139 The pharmaceutical composition of Claim 135, wherein the nitric oxide donor is selected from the group consisting of organic nitrate compounds, iron nitrosyl compounds, S-nitrosothiol compounds, sydnonimine compounds, and NONOate compounds
140 The pharmaceutical composition of Claim 139, wherein the organic nitrate compound is glyceryl trinitrate, nitroglycerin, pentaerythrityl tetranitrate, isosorbide dinitrate, or isosorbide 5-mononitrate
141 The pharmaceutical composition of Claim 139, wherein the iron nitrosyl compound is sodium nitroprusside
142 The pharmaceutical composition of Claim 139, wherein the sydnonimine compound is molsidomine, linsidomine, or pirsidomine
143 The pharmaceutical composition of Claim 139, wherein the S-nitrosothiol compound is S-nitroso-N-acetyl-D,L-penicillamine, S-nitrosoglutathione, S-nitrosoalbumin, or S-nitrosocysteine
144 The pharmaceutical composition of Claim 139, wherein the NONOate compound is diethylamine-NONOate, diethylene triamine-NONOate, dipropylenetriamine- NONOate, spermine- NONOate, propylamino-propylamine-NONOate, MAHMA-NONOate, piperazi-NONOate, proli- NONOate, sulfo-NONOate, Angelis salt, or sulfite NONOate
145 The pharmaceutical composition of Claim 135, wherein the activator of cyclic GMP-dependent protein kinase is selected from the group consisting of octobromo-cyclic GMP and dibutyryl cyclic GMP
146 The pharmaceutical composition of Claim 135, wherein the medicant is a therapeutic cytotoxic agent, DNA expression vector, viral vector, protein, oligonucleotide, nucleotide analog, antimicrobial agent, interferon, cytokine, cytokine agonist, cytokine antagonist, immunotoxin, immunosuppressant, boron compound, monoclonal antibody, adrenergic agent, anticonvulsant, ischemia-protective agent, anti-trauma agent, anticancer chemotherapeutic agent, or diagnostic agent
147 The pharmaceutical composition of Claim 146, wherein the diagnostic agent is an imaging or contrast agent
148 The pharmaceutical composition of Claim 146, wherein the diagnostic agent is a radioactively labeled substance, a gallium-labeled substance, or a contrast agent selected from the group consisting of ferrous magnetic, fluorescent, luminescent, and iodinated contrast agents
149 The pharmaceutical composition of Claim 135, wherein the medicant is a Nmethyl-D-aspartate (NMDA) receptor antagonist, antibiotic, interleukin-2, or transforming growth factor-β, cisplatin, carboplatin, tumor necrosis factor-α, methotrexate, 5-fluorouracil, amphotericin, daunorubicin, doxorubicin, vincristine, vinblastine, busulfan, chlorambucil, cyclophosphamide, melphalan, or ethyl ethanesulfonic acid
150 The pharmaceutical composition of Claim 146, wherein the viral vector is an adenovirus-derived vector or herpes simplex virus-derived vector
151 The pharmaceutical composition of Claim 135, further comprising a buffer solution pharmaceutically acceptable for intravascular infusion into a mammal
152 The pharmaceutical composition of Claim 152, wherein the buffer solution is phosphate buffered saline
153. A kit for enhancing the delivery of a medicant to an abnormal brain region and/or to a malignant tumor, comprising a potassium channel activator selected from the group consisting of activators of soluble guanylyl cyclase and activators of cyclic GMP-dependent protein kinase, and instructions for using the potassium channel activator for enhancing the delivery of a medicant to an abnormal brain region or to a malignant tumor
154 The kit of Claim 153, wherein the activator of soluble guanylyl cyclase is nitric oxide or a nitric oxide donor 155 The kit of Claim 154, wherein the nitric oxide donor is selected from the group consisting of organic nitrate compounds, iron nitrosyl compounds, S-nitrosothiol compounds, sydnonimine compounds, and NONOate compounds
156 The kit of Claim 155, wherein the organic nitrate compound is glyceryl trinitrate, nitroglycerin, pentaerythrityl tetranitrate, isosorbide dinitrate, or isosorbide 5-mononitrate
157 The kit of Claim 155, wherein the iron nitrosyl compound is sodium nitroprusside
158 The kit of Claim 155, wherein the sydnonimine compound is molsidomine, linsidomine, or pirsidomine
159 The kit of Claim 155, wherein the S-nitrosothiol compound is S-nitroso-N-acetyl-D,L-penicillamine, S-nitrosoglutathione, S-nitrosoalbumin, or S-nitrosocysteine
160 The kit of Claim 155, wherein the NONOate compound is diethylamine-NONOate, diethylene triamine-NONOate, dipropylenetriamine-NONOate, spermine-NONOate, propylamino-propylamine-NONOate, MAHMA-NONOate, piperazi-NONOate, proli-NONOate, sulfo-NONOate, Angelis salt, or sulfite NONOate
161 The kit of Claim 153, wherein the activator of cyclic GMP-dependent protein kinase is selected from the group consisting of octobromo-cyclic GMP and dibutyryl cyclic GMP
EP01905141A 2000-01-26 2001-01-26 Method for using potassium channel activation for delivering a medicant to an abnormal brain region and/or a malignant tumor Withdrawn EP1251840A2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US491500 2000-01-26
US09/491,500 US7018979B1 (en) 2000-01-26 2000-01-26 Method for using potassium channel agonists for delivering a medicant to an abnormal brain region and/or a malignant tumor
US61585400A 2000-07-14 2000-07-14
US615854 2000-07-14
PCT/US2001/002742 WO2001054680A2 (en) 2000-01-26 2001-01-26 Method for using potassium channel activation for delivering a medicant to an abnormal brain region and/or a malignant tumor

Publications (1)

Publication Number Publication Date
EP1251840A2 true EP1251840A2 (en) 2002-10-30

Family

ID=27050462

Family Applications (1)

Application Number Title Priority Date Filing Date
EP01905141A Withdrawn EP1251840A2 (en) 2000-01-26 2001-01-26 Method for using potassium channel activation for delivering a medicant to an abnormal brain region and/or a malignant tumor

Country Status (4)

Country Link
EP (1) EP1251840A2 (en)
JP (2) JP2004508279A (en)
AU (2) AU2001233052A1 (en)
WO (2) WO2001054680A2 (en)

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MXPA02010551A (en) 2000-04-26 2003-09-25 Univ Kingston Formulations and methods of using nitric oxide mimetics against a malignant cell phenotype.
US7678391B2 (en) 2000-04-26 2010-03-16 Queen's University At Kingston Formulations and methods of using nitric oxide mimetics against a malignant cell phenotype
GB0021487D0 (en) * 2000-09-01 2000-10-18 Pfizer Ltd Pharmaceutical
US7049334B2 (en) * 2001-09-14 2006-05-23 Carlsbad Technology, Inc. Enhancement of learning and memory and treatment of amnesia
US7211561B2 (en) * 2001-10-12 2007-05-01 Cedars-Sinai Medical Center Method for inducing selective cell death of malignant cells by activation of calcium-activated potassium channels (KCa)
WO2003074082A1 (en) * 2002-03-06 2003-09-12 Cellegy Pharmaceuticals, Inc. Formulations and methods of using nitric oxide mimetics in cancer treatment
AU2003222020B2 (en) 2002-03-20 2008-08-28 University Of Maryland Baltimore A non-selective cation channel in neural cells and methods for treating brain swelling
US8980952B2 (en) 2002-03-20 2015-03-17 University Of Maryland, Baltimore Methods for treating brain swelling with a compound that blocks a non-selective cation channel
US7166293B2 (en) 2002-03-29 2007-01-23 Carlsbad Technology, Inc. Angiogenesis inhibitors
WO2004002962A1 (en) * 2002-06-26 2004-01-08 Poseidon Pharmaceuticals A/S Novel benzimidazol-2-one derivatives and their use
DE10240735A1 (en) * 2002-08-29 2004-03-18 Eberhard-Karls-Universität Tübingen Universitätsklinikum Regulating human endostatin expression, e.g. for treating CNS disorders or tumors, using nitrogen monoxide signal cascade modulators
ES2222831B2 (en) * 2003-07-30 2006-02-16 Laboratorios Del Dr. Esteve, S.A. COMBINATION OF ACTIVE PRINCIPLE THAT INCLUDES A 2,5-DIHYDROXIBENCENOSULPHONE COMPOUND AND A K + CHANNEL MODULATOR.
WO2005025511A2 (en) * 2003-09-10 2005-03-24 Cedars-Sinai Medical Center Potassium channel mediated delivery of agents through the blood-brain barrier
ES2338022T3 (en) 2004-06-10 2010-05-03 Nippon Kayaku Kabushiki Kaisha POTENTIAL OF THE ANTINEOPLASIC EFFECT.
JP5307397B2 (en) 2004-09-18 2013-10-02 ユニバーシティ オブ メリーランド,ボルチモア Therapeutic agent targeting NCCa-ATP channel and method of use thereof
CA2580606A1 (en) 2004-09-18 2006-03-30 University Of Maryland, Baltimore Therapeutic agents targeting the ncca-atp channel and methods of use thereof
WO2006088798A2 (en) * 2005-02-14 2006-08-24 Albert Einstein College Of Medicine Of Yeshiva University Modulation of hypothalamic atp-sensitive potassium channels
WO2006091587A2 (en) * 2005-02-22 2006-08-31 Cedars-Sinai Medical Center Use of minoxidil sulfate as an anti-tumor drug
WO2007002254A2 (en) * 2005-06-23 2007-01-04 Merck & Co., Inc. Benzocycloheptapyridines as inhibitors of the receptor tyrosine kinase met
CA2674949A1 (en) 2007-01-12 2008-07-24 J. Marc Simard Targeting ncca-atp channel for organ protection following ischemic episode
US8557867B2 (en) 2007-06-22 2013-10-15 The United States Of America As Represented By The Department Of Veterans Affairs Inhibitors of NCCa-ATP channels for therapy
JP2011521908A (en) * 2008-05-09 2011-07-28 デューク ユニバーシティ Treatment of diseases based on the finding that thioredoxin regulates nitric oxide release in cells
GB0921525D0 (en) * 2009-12-08 2010-01-27 Isis Innovation Product and method
KR102282716B1 (en) * 2020-02-07 2021-07-29 포항공과대학교 산학협력단 Composition for increasing permeability of blood-brain barrier comprising nitric oxide donor and use thereof
KR102490837B1 (en) * 2020-03-19 2023-01-19 포항공과대학교 산학협력단 Method for increasing permeability of blood-brain barrier

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA1336963C (en) * 1988-07-18 1995-09-12 Gary James Grover Method for inhibiting myocardial cell necrosis and preserving heart function during myocardial ischemia and/or reperfusion
US5112596A (en) * 1990-04-23 1992-05-12 Alkermes, Inc. Method for increasing blood-brain barrier permeability by administering a bradykinin agonist of blood-brain barrier permeability
EP0555681A1 (en) * 1992-02-13 1993-08-18 E.R. SQUIBB &amp; SONS, INC. Use of a combination of potassium channel activators and sulfhydryl containing compounds for the manufacture of a medicament for the treatment of ischemia and myocarolial infarct
US5262419A (en) * 1992-06-11 1993-11-16 E. R. Squibb & Sons, Inc. Method for the prophylaxis and/or treatment of ulcerative gastrointestinal conditions using a potassium channel activator
IT1282686B1 (en) * 1996-02-26 1998-03-31 Nicox Sa COMPOUNDS ABLE TO REDUCE DRUG TOXICITY
EP1135157A4 (en) * 1998-10-19 2004-12-08 Univ New York Method for regulating the permeability of the blood brain barrier

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0154680A2 *

Also Published As

Publication number Publication date
WO2001054680A3 (en) 2002-06-27
WO2001054771A2 (en) 2001-08-02
WO2001054680A2 (en) 2001-08-02
JP2003523965A (en) 2003-08-12
WO2001054771A3 (en) 2002-06-20
AU2001233052A1 (en) 2001-08-07
JP2004508279A (en) 2004-03-18
AU2001234602A1 (en) 2001-08-07

Similar Documents

Publication Publication Date Title
WO2001054680A2 (en) Method for using potassium channel activation for delivering a medicant to an abnormal brain region and/or a malignant tumor
US7846900B2 (en) Cancer treatment using C-type natriuretic peptide
EP2433653B1 (en) Use of aprotinin polypeptides as carriers in pharmaceutical conjugates
JP5944018B2 (en) Induction of tumor hypoxia for cancer treatment
US8198241B2 (en) Use of an agent that restores tissue perfusion and oxygenation
AU2013329121B2 (en) Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence
JP2000510460A (en) Treatment of epithelial malignancies using squalamine in combination with other anticancer drugs
WO2001041779A2 (en) Combined use of glp-2 receptor agonist and chemotherapeutic agent in treatment
JP2020500190A (en) HDM2-p53 interaction inhibitor dose and regimen
Armstrong et al. Bcl-2 inhibits apoptosis induced by mitochondrial uncoupling but does not prevent mitochondrial transmembrane depolarization
US20060153770A1 (en) Method of treating tumors
JP2010502638A (en) Sensitization of tumor cells to radiation therapy by administration of endothelin agonists
US20080050337A1 (en) Method for using calcium-sensitive potassium channel agonist for delivering a medicant to an abnormal brain region and/or a malignant tumor
US7018979B1 (en) Method for using potassium channel agonists for delivering a medicant to an abnormal brain region and/or a malignant tumor
US20050153940A1 (en) Method for using potassium channel activation for delivering a medicant to an abnormal brain region and/or a malignant tumor
WO1995033479A1 (en) Cytotoxic enhancement of tnf with copper
EP1251838A2 (en) Method for using potassium channel agonists for delivering a medicant to an abnormal brain region and/or a malignant tumor
US20050095196A1 (en) Method for using potassium channel agonists for delivering a medicant to an abnormal brain region and/or a malignant tumor
US20090048179A1 (en) Use of 5-phosphodiesterase inhibitors to enhance the permeability of the blood-brain barrier of abnormal brain tissue and the blood-tumor barrier
US20230381349A1 (en) Methods and compositions for imaging and treating cancer
박초롱 Therapeutic effect and safety of human serum albumin-CDDP complex in U87MG subcutaneous xenograft tumor models
JPH03502805A (en) Nephroprotective continuous infusion solution
JP2021526550A (en) Methods and compositions for inhibiting harmful injuries
Dewhirst Digital access to libraries
Foley et al. Hsp90 Inhibitor STA-9090 Induces HIF1A Degradation in the Hypoxic Regions of Solid Tumors

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20020823

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

AX Request for extension of the european patent

Free format text: AL;LT;LV;MK;RO;SI

17Q First examination report despatched

Effective date: 20061013

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20080801