WO2010057317A1 - Composition liposomale pour administration améliorée par convection vers le système nerveux central - Google Patents

Composition liposomale pour administration améliorée par convection vers le système nerveux central Download PDF

Info

Publication number
WO2010057317A1
WO2010057317A1 PCT/CA2009/001708 CA2009001708W WO2010057317A1 WO 2010057317 A1 WO2010057317 A1 WO 2010057317A1 CA 2009001708 W CA2009001708 W CA 2009001708W WO 2010057317 A1 WO2010057317 A1 WO 2010057317A1
Authority
WO
WIPO (PCT)
Prior art keywords
animals
topotecan
tumor
delivery vehicle
tpt
Prior art date
Application number
PCT/CA2009/001708
Other languages
English (en)
Inventor
Thomas Redelmeier
Matthias Luz
Original Assignee
Medgenesis Therapeutix, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Medgenesis Therapeutix, Inc. filed Critical Medgenesis Therapeutix, Inc.
Priority to AU2009317837A priority Critical patent/AU2009317837B2/en
Priority to CN200980151445.7A priority patent/CN102256596B/zh
Priority to BRPI0922100A priority patent/BRPI0922100A2/pt
Priority to JP2011536717A priority patent/JP5795537B2/ja
Priority to US13/130,525 priority patent/US9295735B2/en
Priority to EP09827097.8A priority patent/EP2370058B1/fr
Priority to CA2743959A priority patent/CA2743959C/fr
Publication of WO2010057317A1 publication Critical patent/WO2010057317A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0063Preparation for luminescence or biological staining characterised by a special physical or galenical form, e.g. emulsions, microspheres
    • A61K49/0069Preparation for luminescence or biological staining characterised by a special physical or galenical form, e.g. emulsions, microspheres the agent being in a particular physical galenical form
    • A61K49/0076Preparation for luminescence or biological staining characterised by a special physical or galenical form, e.g. emulsions, microspheres the agent being in a particular physical galenical form dispersion, suspension, e.g. particles in a liquid, colloid, emulsion
    • A61K49/0084Preparation for luminescence or biological staining characterised by a special physical or galenical form, e.g. emulsions, microspheres the agent being in a particular physical galenical form dispersion, suspension, e.g. particles in a liquid, colloid, emulsion liposome, i.e. bilayered vesicular structure
    • 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
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/0019Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules
    • A61K49/0021Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules the fluorescent group being a small organic molecule
    • A61K49/0023Di-or triarylmethane dye
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/0019Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules
    • A61K49/0021Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules the fluorescent group being a small organic molecule
    • A61K49/0041Xanthene dyes, used in vivo, e.g. administered to a mice, e.g. rhodamines, rose Bengal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • 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
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M25/00Catheters; Hollow probes
    • A61M25/0021Catheters; Hollow probes characterised by the form of the tubing
    • A61M2025/0042Microcatheters, cannula or the like having outside diameters around 1 mm or less
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M25/00Catheters; Hollow probes
    • A61M25/0067Catheters; Hollow probes characterised by the distal end, e.g. tips
    • A61M25/0068Static characteristics of the catheter tip, e.g. shape, atraumatic tip, curved tip or tip structure
    • A61M25/007Side holes, e.g. their profiles or arrangements; Provisions to keep side holes unblocked

Definitions

  • the present invention relates to liposomal formulations that are deliverable by convection-enhanced delivery and useful for the treatment of central nervous system disorders.
  • CED convection-enhanced delivery
  • CED provides reproducible distribution within a given target tissue and can produce homogeneous drug concentrations throughout the volume of distribution (V d ) (Croteau et al., 2005; Lonser et al., 2002).
  • Chemotherapeutic agents delivered locally by CED have produced favorable therapeutic outcomes (Bruce et al., 2000; Degen et al., 2003; Kaiser et al. 2000). However, most cytotoxic agents delivered directly to the nervous system have the capacity to damage healthy cells. Accordingly, good candidates for CED administration into brain tumors must have the highest possible therapeutic index against tumor cells in comparison with healthy neuronal cells.
  • DM VAN/253729-17775/7468072 1 lnt J Phartn 354-56-62 Epub Nov 9 2007
  • PEGylated liposomes may cause non-lgE-mediated hypersensitivity reactions, which include symptoms of cardiopulmonary distress (e.g , dyspnea, tachypnea, tachycardia, chest pain, hypertension, and hypotension) (Ishida and Kiwada (2008), supra, Moghimi et al (2006) FASEB J 20 2591-3 Epub Oct 25, 2006)
  • a CNS disorder e g , a disorder associated with the death and/or dysfunction of a particular neuronal population in the CNS
  • the methods involve administering a therapeutically effective amount of a pharmaceutical composition to a patient having a CNS disorder, wherein the pharmaceutical composition is locally delivered to the particular neuronal population by convection-enhanced delivery, and wherein the pharmaceutical composition comprises at least one therapeutic agent encapsulated in non-PEGylated liposomes comprising a mixture of a neutral saturated phospholipid and at least one anionic saturated lipid, and wherein the convection-enhanced delivery of the pharmaceutical composition treats a patient having a CNS disorder
  • Therapeutic agents finding advantageous use in the subject invention include, e g , antineoplastic agents, radioiodinated compounds, toxins (including protein toxins), cytotoxic agents including cytostatic or cytolytic drugs, genetic and viral vectors, vaccines, synthetic vectors, growth factors, neurotrophic factors, antivirals, antibiotics, neurotransmitters, cytokines, enzymes and agents for targeted lesioning of specific sites
  • CNS disorders that may be treated by the compositions and methods provided herein include, e g , cancer, infection, head trauma, spinal cord injury, multiple sclerosis, dementia with Lewy bodies, ALS, lysosomal storage disorders, psychiatric disorders, neurodegenerative disorders, stroke, epilepsy, and other acute and chronic disorders of the CNS
  • DM_VAN/253729 17775/7468072 1 In one embodiment, provided herein are methods for inhibiting the growth of a CNS tumor, reducing a CNS tumor, killing one or more CNS tumor cells, and/or treating a patient having a CNS tumor
  • the methods involve administering a therapeutically effective amount of a pharmaceutical composition to a patient having a CNS tumor, wherein the pharmaceutical composition is locally delivered to the CNS tumor by convection-enhanced delivery, and wherein the pharmaceutical composition comprises at least one cytotoxic agent encapsulated in non-PEGylated liposomes comprising a mixture of a neutral saturated phospholipid and at least one anionic saturated lipid, and wherein the convection-enhanced delivery of the pharmaceutical composition inhibits the growth of a CNS tumor, reduces a CNS tumor, kills one or more of the CNS tumor cells and/or treats a patient having a CNS tumor
  • the methods involve administering a therapeutically effective amount of a pharmaceutical composition to a patient having epilepsy, wherein the pharmaceutical composition is locally delivered to an aggregate of CNS neurons exhibiting abnormal or excessive hypersynchronous discharges by convection-enhanced delivery, and wherein the pharmaceutical composition comprises at least one therapeutic agent encapsulated in non-PEGylated liposomes comprising a mixture of a neutral saturated phospholipid and at least one anionic saturated lipid, and wherein the convection-enhanced delivery of the pharmaceutical composition inhibits or reduces the number or duration of seizures in a patient having epilepsy
  • the therapeutic agent is a toxin, e g , a peptide toxin
  • the peptide toxin is a ⁇ z-conotoxin, e g , ⁇ -conotoxin MVIIA or ⁇ -conoto
  • the pharmaceutical composition further comprises at least one diagnostic agent (sometimes referred to herein as a "tracing agent” or “tracer”) encapsulated in similar non-PEGylated anionic liposomes, which allows for visualization of the distribution of the therapeutic agent during and after CED
  • the non-PEGylated liposomes encapsulating the diagnostic agent are composed of the same lipids as the non-PEGylated liposomes encapsulating the therapeutic agent Accordingly, in one embodiment, methods described herein further comprise the step of detecting the diagnostic agent
  • the non-PEGylated liposomes may contain a therapeutic drug
  • the therapeutic drug is an insoluble therapeutic drug
  • the therapeutic drug is a topoisomerase I inhibitor (e g , a camptothecin and derivatives thereof), which includes but is not limited to topoisomerase l/ll inhibitors
  • the therapeutic drug is a camptothecin derivative selected from the group consisting of 9-aminocamptothec ⁇ n, 7-ethylcamptothec ⁇ n, 10-hydroxycamptothec ⁇ n, 9-nitrocamptothecin, 10,11- methlyenedioxycamptothecin, 9-am ⁇ no-10,11-methylened ⁇ oxycamptothec ⁇ n 9-chloro-10,11-
  • DM VAN/253729-17775/7468072 1 methylenedioxycamptothecin, irinotecan, topotecan, 7-(4-methylpiperazinometriylene)-10,11- ethylenedioxy-20(S)-camptothecin, 7-(4-methylpiperazinomethylene)-10,11-methylenedioxy-20(S)- camptothecin and 7-(2-(N-isopropylamino)ethyl)-(20S)-camptothecin.
  • the camptothecin derivative is selected from the group consisting of irinotecan, topotecan, (7-(4- methylpiperazinomethylene)-10,11-ethylenedioxy-20(S)-camptothecin, 7-(4- methylpiperazinomethylene)-10,11-methylenedioxy-20(S)-camptothecin or 7-(2-(N- isopropylamino)ethyl)-(20S)-camptothecin.
  • the camptothecin is topotecan.
  • the topoisomerase inhibitor is a topoisomerase l/ll inhibitor, such as 6-[[2-(dimethylamino)-ethyl]amino]-3-hydroxy-7H-indeno[2,1-c]quinolin-7-one dihydrochloride, azotoxin or 3-methoxy-11 H-pyrido[3',4'-4,5]pyrrolo[3,2-c]quinoline-1 ,4-dione.
  • a topoisomerase l/ll inhibitor such as 6-[[2-(dimethylamino)-ethyl]amino]-3-hydroxy-7H-indeno[2,1-c]quinolin-7-one dihydrochloride, azotoxin or 3-methoxy-11 H-pyrido[3',4'-4,5]pyrrolo[3,2-c]quinoline-1 ,4-dione.
  • the therapeutic drug is a toxin, e.g., a protein toxin, e.g., ⁇ -conotoxin, (e.g., ⁇ -conotoxin MVIIA or ⁇ -conotoxin, GVIA), a botulinum toxin (e.g., a botulinum toxin serotype A such as BOTOX® or DYSPORT®, a botulinum toxin serotype B such as MYOBLOC®) ⁇ -conotoxin, ⁇ -conantokin peptide, etc.
  • a protein toxin e.g., ⁇ -conotoxin, (e.g., ⁇ -conotoxin MVIIA or ⁇ -conotoxin, GVIA)
  • a botulinum toxin e.g., a botulinum toxin serotype A such as BOTOX® or DYSPORT®, a botulinum tox
  • the initial drug concentration is at least about 100 ug/mL, preferably at least about 200 ug/mL, and more preferably at least about 300 ug/mL. In another embodiment, the initial drug concentration is about 2 mg/ml to about 5 mg/ml. In one embodiment, the therapeutic drug and/or diagnostic agent to lipid ratio is from about 0.1 to about 0.5. In another embodiment, the therapeutic drug and/or diagnostic agent to lipid ratio is about 0.1. In another embodiment, the therapeutic drug and/or diagnostic agent to lipid ratio is about 0.3. In another embodiment, the therapeutic drug and/or diagnostic agent to lipid ratio is about 0.5.
  • the non-PEGylated liposome contains a diagnostic agent.
  • the diagnostic agent is an MRI magnet.
  • the diagnostic agent is gadolinium chelate.
  • the diagnostic agent is selected from the group consisting of gadodiamide and rhodamine.
  • the diagnostic agent is gadodiamide.
  • the methods described herein comprise convection-enhanced delivery of a liposomal formulation comprising at least one therapeutic agent and/or at least one diagnostic agent encapsulated in non-PEGylated liposomes composed of a mixture of at least one neutral saturated phospholipid and at least one anionic saturated phospholipid.
  • the neutral saturated phospholipid is selected from the group consisting of derivatives of phosphatidylcholine and mixtures thereof, for example dipalmitoylphosphatidylcholine (DPPC), distearoylphosphatidylcholine (DSPC), dimyristoylphosphatidylcholine (DMPC), and mixtures thereof.
  • DPPC dipalmitoylphosphatidylcholine
  • DSPC distearoylphosphatidylcholine
  • DMPC dimyristoylphosphatidylcholine
  • the anionic saturated phospholipid is selected from a group consisting of derivatives of phosphatidylglycerol (e.g., distearoylphosphatidylglycerol (DSPG)), dipalmitoyl phosphatidyl glycerol (DPPG), phosphatidylserine, phosphatidylinositol, phosphatidic acid and mixtures thereof.
  • DSPG distearoylphosphatidylglycerol
  • DPPG dipalmitoyl phosphatidyl glycerol
  • phosphatidylserine phosphatidylinositol
  • phosphatidic acid phosphatidic acid
  • the liposomal formulations described herein may also contain other lipid components such as sterols and derivatives (for example cholesterol (CHOL)) or sphingolipids (for example sphingomyelins and glycosphingolipids, in particular ganghosides)
  • sterols and derivatives for example cholesterol (CHOL)
  • sphingolipids for example sphingomyelins and glycosphingolipids, in particular ganghosides
  • the liposomal formulations will consist essentially of or consist of at least one neutral saturated phospholipid, at least one anionic saturated phospholipid and a stabilizer such as, e g , cholesterol
  • the non-PEGylated liposome is composed of a combination of distearoylphosphatidylcholine (DSPC) and distearoylphosphatidylglycerol (DSPG)
  • the non-PEGylated liposome comprises about 10 to about 95 mole percent DSPC
  • the non-PEGylated liposome comprises about 5 to about 90 mole percent DSPG
  • the non-PEGylated liposome further comprises cholesterol (CHOL), e g , about 5 to about 45 mole percent cholesterol
  • the liposome comprises or consists essentially of about 60 to about 90 mole percent DSPC, about 5 to about 10 mole percent cholesterol, and about 5 to about 30 mole percent DSPG
  • the non-PEGylated liposome comprises or consists essentially of DSPC, DSPG, and CHOL at a 7 2 1 molar ratio
  • the non-PEGylated liposome comprises or consists essentially of DSPC, DSPG,
  • convection-enhanced delivery (CED) of non-PEGylated liposomal formulations as described herein provides increased tissue distribution, decreased toxicity and increased in vivo half-life of the therapeutic drug as compared to the respective tissue distribution, toxicity, and in vivo half-life of the freely administered therapeutic drug
  • the invention provides a cannula comprising a liposomal formulation described herein, e g , a liposomal formulation comprising at least one therapeutic agent encapsulated in non-PEGylated liposomes composed of a mixture of at least one neutral saturated phospholipid and at least one anionic saturated phospholipid, and wherein the formulation may be delivered by convection-enhanced delivery (CED)
  • the cannula further comprises a liposomal formulation comprising a diagnostic agent encapsulated in non-PEGylated liposomes composed of a mixture of at least one neutral saturated phospholipid and at least one anionic saturated phospholipid, and wherein the formulation may be delivered by CED
  • the cannula comprises a liposomal formulation comprising a first liposome containing a therapeutic drug and a second liposome containing a diagnostic agent, wherein neither the first nor second liposome are PEGylated, wherein the first
  • the invention provides methods for producing the liposomal formulations described herein In one aspect, the invention provides methods for producing a medicament useful
  • the method comprises entrapping the therapeutic drug or diagnostic agent within the liposomes by remote loading, for example, via an ammonium sulfate gradient
  • Figs. 1A - 1F compare the effect of lipid composition, drug concentration and drug lipid ratio on the release characteristics of topotecan from pegylated and non-pegylated liposomal formulations
  • Fig. 2 shows the pharmacokinetics of Ls-TPT Formulations and free topotecan in Normal brain tissue
  • Fig. 3 shows the effect of sucrose on convectability of rhodamine liposomes
  • Fig. 4 shows the distribution volume (Vd) of rhodamine loaded liposomes after a 20 ⁇ infusion into the striatum
  • Fig. 5 shows survival of animals by treatment group
  • Fig. 6 shows survival of animals by combined treatment group vs group 2 (0 5 mg/mL dual dosing)
  • Fig. 7 shows overall survival by U87 cell load at tumor implantation
  • Fig. 8 shows survival of animals by combined treatment groups vs group 2 (0 5 mg/mL dual dosing) in animals with low U87MG Cell Load (6 8X10 3 )
  • Fig. 9 shows survival of animal by combined treatment groups vs group 2 (0 5 mg/mL dosing) in animals with high U87MG cell load (9 7X10 5 )
  • Fig. 10 shows volume of distribution of Ls-TPT-mar ⁇ na blue DHPE coinfused with Ls-Gd- rhodam ⁇ ne-PE in naive rodent brain tissue
  • n 3 and 20 ⁇ L was infused in each hemisphere
  • FIG. 11 shows volume of distribution of Ls-TPT-mar ⁇ na blue DHPE coinfused with Ls-Gd- rhodam ⁇ ne-PE in U87MG xenograft rodent brain tissue
  • n 4 and 20 ⁇ L was infused in each hemisphere
  • Fig. 12 shows survival of animals by treatment groups (euthanized animals considered as uncensored)
  • Fig. 13 shows survival of animals by treatment groups (euthanized animals considered as censored
  • liposome refers to a lipid bilayer membrane containing an entrapped aqueous volume Liposomes may be unilamellar vesicles having a single membrane bilayer or multilamellar vesicles having multiple membrane bilayers separated from each other by an aqueous layer
  • the liposomal bilayer is composed of two lipid monolayers having a hydrophobic "tail” region and a hydrophilic "head” region
  • the structure of the membrane bilayer is such that the hydrophobic (non-polar) "tails” of the lipid monolayers orient toward the center of the bilayer while the hydrophilic (polar) "heads” orient toward either the entrapped aqueous volume or the extraliposomal aqueous environment
  • a liposome of the invention includes a targeting moiety, e g , an antibody or other hgand
  • Lipomal formulations are understood to be those in which part or all of the therapeutic drug and/or diagnostic agent is encapsulated inside the liposomes
  • Consisting essentially of as used herein in reference to liposomal formulations refers to liposomes having the recited lipid components only, and no additional lipid components
  • Phospholipid is understood to mean an amphiphile derivative of glycerol in which one of its hydroxyl groups is esterified with phosphoric acid and the other two hydroxyls are esterified with long- chain fatty acids, which may be equal or different from each other
  • a saturated phospholipid will be that whose fatty acids only have simple (not multiple) covalent carbon-carbon bonds
  • a neutral phospholipid will generally be one in which another phosphoric acid hydroxyl is esterified by an alcohol substituted by a polar group (usually hydroxyl or amine) and whose net charge is zero at physiological pH
  • An anionic phospholipid will generally be one in which another phosphoric acid hydroxyl is esterified by an alcohol substituted by a polar group and whose net charge is negative at physiological PH
  • charged saturated phospholipid also includes other amphiphile compounds whose net charge is different from zero
  • amphiphile compounds include, but are not limited to, long chain hydrocarbonate derivatives, substituted by a polar group (for example amine) and derivatives of fatty acids
  • active agent or “therapeutic agent” refers to any molecule that may be delivered to CNS target tissue in the form of a high molecular weight neurotherapeutic, and when so delivered, effects a desirable response in the target CNS tissue
  • Therapeutic agents include but are not limited to antineoplastic agents, radioiodinated compounds, toxins (including protein toxins), cytotoxic agents including cytostatic or cytolytic drugs, genetic and viral vectors, vaccines, synthetic vectors, growth factors, neurotrophic factors, antivirals, antibiotics, neurotransmitters, cytokines, enzymes and agents for targeted lesioning of specific sites
  • Therapeutic agents include, but are not limited to, nucleic acids, including nucleic acid analogs, proteins, including antibodies, and small molecule chemical compositions Active agents include agents that exhibit toxicity and unwanted effects when administered systemically
  • a "CNS disorder” refers to a disorder of the central nervous system of a subject
  • the disorder may be associated with the death and/or dysfunction of a particular neuronal population in the CNS
  • the disorder may be associated with the aberrant growth of cells within the CNS
  • the aberrantly growing cells of the CNS may be native to the CNS or derived from other tissues Included among CNS disorders are cancer, infection, head trauma, spinal cord injury, multiple sclerosis, dementia with Lewy bodies, ALS, lysosomal storage disorders, psychiatric disorders, neurodegenerative disorders, stroke, epilepsy, and other acute and chronic disorders of the CNS
  • Gliomas are the most common primary tumors of the central nervous system (CNS) Glioblastoma multiforme (GBM) is the most frequent and the most malignant type of glioma There is a much higher incidence of GBM in adults than in children According to the Central Brain Tumor Registry of the United States statistical report, GBM accounts for about 20% of all brain tumors in the USA (CBTRUS, 1998-2002)
  • Other tumors of the CNS include, but are not limited to, other gliomas, including astrocytoma, including fibrillary (diffuse) astrocytoma, pilocytic astrocytoma, pleomorphic xanthoastrocytoma, and brain stem glioma, oligodendroglioma, and ependymoma and related paraventricular mass lesions, neuronal tumors, poorly differentiated neoplasms, including medulloblastoma, other parenchymal tumors, including primary
  • Epilepsy is the most common serious CNS disorder associated with the dysfunciton of a particular neuronal population in the CNS (Shorvon, S , Epidemiology, classification, natural history, and genetics of epilepsy, Lancet 1990 JuI 14, 336(8707) 93-6, McNamara J , The neurobiological basis of epilepsy, Trends Neurosci 1992 October, 15(10) 357-9) Severe, penetrating head trauma is associated with up to a 50% risk of leading to epilepsy Other causes of epilepsy include stroke, infection and genetic susceptibility
  • a seizure is a neurological dysfunction which results from abnormal, excessive, hypersynchronous discharges from an aggregate of central nervous system neurons A seizure can be manifested behaviorally (if motor systems are involved) or
  • Epilepsy describes a condition in which a person has recurrent seizures due to a chronic, underlying process. Although there are various epilepsy syndromes in which the clinical and pathologic characteristics differ the common underlying etiology is neuronal hyperexcitability. Thus, epilepsy encompasses disorders of central nervous system (CNS) hyperexcitability, characterized by chronic, recurrent, paroxysmal changes in neurological function that can be categorized according to electroencephalograph ⁇ and clinical presentation (Dichter M., Basic mechanisms of epilepsy: targets for therapeutic intervention, Epilepsia 1997; 38 Suppl 9:S2-6).
  • CNS central nervous system
  • Epileptic seizures are broadly categorized into two groups: focal (partial) and generalized seizures.
  • Focal seizures arise from abnormal activity of a limited group of neurons in cortical or subcortical regions of the brain.
  • the underlying structural abnormality or lesion can develop as a result of birth injury, head trauma, tumor, abscess, infarction, vascular malformation or genetic disease (Dichter 1997, Ibid).
  • the location of the focal activity can be identified by the clinical seizure presentation or may be cryptic. Equivalently, the active focus may not involve the lesion itself but may arise in adjacent or distant (but connected) neuronal populations, supporting the hypothesis of plastic synaptic reorganization underlying focal hyperexcitability. (See e.g. Prince D.
  • Focal seizures are termed “simple” if there is no apparent change in consciousness, otherwise they are termed “complex”. Complex focal seizures involve the temporal lobe and limbic system, and are the most common manifestation of epilepsy in adults. Focal seizures that spread to become bilateral electrographically, with concomitant loss of consciousness and with or without motor manifestations, are said to be secondarily generalized. Primary generalized seizures initiate with bilateral electrographic activity, loss of consciousness, and with or without motor convulsions. Focal epilepsy can involve almost any part of the brain and usually results from a localized lesion of functional abnormality. Current therapy for focal epilepsy includes use of an EEG to localize abnormal spiking waves originating in areas of organic brain disease that predispose to focal epileptic attacks, followed by surgical excision of the focus to prevent future attacks.
  • Liposomal formulations described herein may be formed in a variety of ways, including by active or passive loading methodologies.
  • one or more therapeutic drug(s) and/or diagnostic agent(s) may be encapsulated using a transmembrane pH gradient loading technique.
  • General methods for loading liposomes with therapeutic drugs through the use of a transmembrane potential across the bilayers of the liposomes are well known to those in the art (e.g., U.S. Patent Nos. 5,171 ,578; 5,077,056); and 5,192,549).
  • the lipids may be first dissolved in an organic solvent, such as ethanol, t-butanol, mixtures thereof, etc., and gently heated (e.g., 60 °C - 70 0 C).
  • an organic solvent such as ethanol, t-butanol, mixtures thereof, etc.
  • DM VAN/253729-17775/7468072 1 forming the non-PEGylated liposomes may be selected from a variety of vesicle-forming lipids, typically including phospholipids and sterols (e g , U S Patent Nos 5,059,421 and 5,100,662)
  • phospholipids derived from egg yolk, soybean or other vegetable or animal tissue such as phosphatidylcholines, phosphatidylethanolamines, phosphatide acid, phosphatidylse ⁇ nes, phosphatidylinositols, phosphatidylglycerols, sphingomyelins, etc , mixtures thereof such as egg yolk phospholipid, soybean phospholipid, etc , hydrogenation products thereof, and synthetic phospholipids such as dipalmitoylphosphatidlcholines, distearoylphosphatidylcholines, distearoylphosphatidylglycerols or the like
  • the non-PEGylated anionic liposomes of the subject invention are a mixture of two or more non-PEGylated lipids, e g , a neutral phospholipid and an anionic phospholipid
  • the neutral phospholipid is chosen from the group composed of derivatives of phosphatidylcholine and their combinations, for example dipalmitoylphosphatidylcholine (DPPC), distearoylphosphatidylcholine (DSPC), dimy ⁇ stoylphosphatidylcholine (DMPC) and their combinations
  • the anionic phospholipid is selected from a group composed of derivatives of phosphatidylglycerol, dipalmitoyl phosphatidyl glycerol (DPPG), phosphatidylse ⁇ ne, phosphatidyhnositol, phosphatide acid and their combinations, for example, distearoyl phosphatidyl glycerol
  • a pre-heated aqueous solution may be added while vigorously mixing
  • a solution containing 150-300 mM buffer may be added
  • Buffers that may be used include, but are not limited to, ammonium sulphate, citrate, maleate and glutamate
  • the resulting multilamellar vesicles may be heated and extruded through an extrusion device to convert the MLVs to unilamellar liposome vesicles
  • the organic solvent used initially to dissolve the lipids may be removed from the liposome preparation by dialysis, diafiltration, etc
  • One or more therapeutic drugs and/or diagnostic agents may be entrapped in the liposomes using transmembrane pH gradient loading By raising the pH of the solution external to the liposomes, a pH differential will exist across the liposome bilayer Thus, a transmembrane potential is created across the liposome bilayer and the one or more therapeutic drug and/or diagnostic agent is loaded into the liposomes by means of the transmembrane potential
  • the therapeutic drug and/or diagnostic agent to lipid ratio is about 0 01 to about 0 5 (wt/wt) In one embodiment, therapeutic drug and/or diagnostic agent to lipid ratio is about 0 1 In another embodiment, the therapeutic drug and/or diagnostic agent to lipid ratio is about 0 3 In one embodiment, vesicles are prepared with a transmembrane ion gradient, and incubated with a therapeutic drug and/or diagnostic agent that is a weak acid or base under conditions that result in encapsulation of the therapeutic agent or diagnostic agent In another embodiment vesicles are prepared in the presence of the therapeutic drug and/or diagnostic agent and the unecapsulated
  • a preferred embodiment for loading is based upon U S Patent No 5,192,549 and involves removing ammonium from the external media The result creates a transmembrane ammonium concentration gradient that induces a pH gradient The drug is added to the vesicles, and "remote" loaded following incubation at elevated temperatures
  • the agent is present in the buffer that is used to make the liposomes and becomes passively encapsulated at the time of vesicle formation
  • an agent that is essentially impermeable e g , a diagnostic agent such as gadodiamide
  • the agent is present in the buffer that is used to make the liposomes and becomes passively encapsulated at the time of vesicle formation
  • This preferred method also applies to other zwitterionic drugs such as methotrexate
  • weak bases (and acids) can be remote loaded into liposomes
  • the liposomal formulations described herein may be used for convection-enhanced delivery to central nervous system regions, and CED can achieve high tissue distribution volumes within the CNS Accordingly, the liposomal formulations may be used for the treatment of CNS disorders
  • CNS disorders include, but are not limited to CNS tumors such as, e g , glioblastoma, and disorders associated with dysfunction of neuronal cells such as , e g , epilepsy
  • therapeutic drugs used in the treatment of CNS disorders may be entrapped within the liposomal formulations described herein for use in methods described herein
  • therapeutic drugs include antitumor agents, toxins, biogenic agents (e g , dopamine, serotonin), neurotrophic factors (e g GDNF, CDNF, MANF), etc
  • topoisomerase I inhibitors are comprised within the liposomal formulations described herein
  • the topoisomerase inhibitor is camptothecan or a derivative thereof
  • the therapeutic drug is a camptothecin derivative selected from the group consisting of 9- aminocamptothecin, 7-ethylcamptothec ⁇ n, 10-hydroxycamptothec ⁇ n, 9-n ⁇ trocamptothec ⁇ n, 10,11-methlyenedioxycamptothecin, 9-am ⁇ no-10,11-methylened ⁇ oxycamptothec ⁇ n 9-chloro-10,11- methylenedioxycamptothecin, irinotecan, topotecan, 7-(4-methylp ⁇ peraz ⁇ nomethylene)-10,11- ethylened ⁇ oxy-20(S)-camptothec ⁇ n, 7-(4-methylp ⁇ peraz ⁇ nomethylene
  • toxins e g , protein toxins, including ⁇ -conotox ⁇ ns (e g , ⁇ -conotoxin GIIIA, ⁇ -conotoxin GIIIB, ⁇ -conotoxin GIIIC, ⁇ -conotoxin PIIIA, ⁇ -conotoxin SmIIIA, ⁇ - conotoxin KIIIA, etc ), ⁇ -conotoxins (e g , ⁇ conotoxin GVIA (also referred to herein as " ⁇ -conotoxin G” and “ ⁇ -CTX-G”)), ⁇ -conotoxm MVIIA (also referred to herein as " ⁇ -conotoxin M” and " ⁇ -CTX-M”), botulinum toxins (e g , botulinum toxin A (also referred to herein as BTX-A), botul
  • ⁇ -conotox ⁇ ns e g , ⁇ -conotoxin GIIIA,
  • conotoxins derived from the venom of Conus snails can be delivered using the subject formulations
  • the active components of the venom are small peptide toxins, usually 10 to 30 amino acid residues in length and typically highly constrained due to their high density of disulphide bonds
  • the venom components act on voltage-gated ion channels, hgand-gated ion channels, and G protein-coupled receptors
  • the pharmaceutical selectivity of conotoxins is at least in part determined by specific disulfide bond frameworks combined with hyperva ⁇ able amino acids within disulfide loops Due to the high potency and vibrant selectivity of the conotoxin peptides, several have been evaluated for the treatment of human disorders and one of these ⁇ -conotoxm MVIIA (ziconotide), an N-type calcium channel blocker, is currently used to treat pain in human patients by means of an implantable, programmable pump with a catheter threaded into the intrathecal space
  • the antiepileptic drug formulation comprises ⁇ -conotoxins such as ⁇ conotoxin GVIA, ⁇ -conotoxin MVIIA and ⁇ -conotoxin CVID See, e g , Gasior et al J Pharmacol Exp Ther 323 458-68 (2007)
  • the antiepileptic drug formulation comprises ⁇ -conotoxms such as ⁇ -conotoxin GIIIA, ⁇ -conotoxin GIIIB, ⁇ -conotoxin GIIIC, ⁇ -conotoxin PIIIA, ⁇ -conotoxin SmIIIA, ⁇ -conotoxin KIIIA See, e g , Zhang et al , J Biol Chem 282 30699-30706 (2007)
  • Other embodiments utilize derivatives or pharmaceutically acceptable salts of the conotoxins, as described herein
  • the different serotypes of botulinum toxin vary in the animal species that they affect and in the severity and duration of the paralysis they evoke For example, it has been determined that botulinum toxin type A is 500 times more potent, as measured by the rate of paralysis produced in the rat than is botulinum toxin type B Additionally, botulinum toxin type B has been determined to be non-toxic in primates at a dose of 480 U/kg which is about 12 times the primate LD50 for botulinum toxin type A Accordingly, non-type A botulinum toxin serotypes may have a lower potency and/or a
  • botulinum toxins serotypes Although all the botulinum toxins serotypes apparently inhibit release of the neurotransmitter at the neuromuscular junction, they do so by affecting different neurosecretory proteins and/or cleaving these proteins at different sites
  • botulinum types A and E both cleave the 25 kiloDalton (kD) synaptosomal associated protein (SNAP-25), but they target different amino acid sequences within this protein
  • Botulinum toxin types B, D, F and G act on vesicle-associated protein (VAMP, also called synaptobrevin), with each serotype cleaving the protein at a different site
  • VAMP vesicle-associated protein
  • botulinum toxin type C1 has been shown to cleave both syntaxin and SNAP-25
  • botulinum toxin inhibits potassium induced release of various neurotransmitters from primary cell cultures and brain synaptosome preparations
  • Glutamate is the neurotransmitter responsible for the bulk of synaptic excitation in the brain, and it is believed to be integral to the generation and spread of seizure discharges It has been reported that botulinum toxin inhibits the evoked release of glutamate in primary cultures of spinal cord neurons and that in brain synaptosome preparations botulinum toxin inhibits the release of glutamate and other neurotransmitters
  • the antiepileptic drug is botulinum toxin A or botulinum toxin B
  • the toxin is a fragment or an analog of botulinum toxin A or botulinum toxin B that possesses biological activity of the parent toxins
  • the toxins are modified to bind specifically to appropriate targets on brain neurons
  • recombinant techniques are used to produce the clostridial neurotoxins or their fragments or analogs
  • Suitable agents include a paramagnetic ion for use with MRI, referred to herein as "MRI magnets "Suitable metal ions include those having atomic numbers of 22-29 (inclusive), 42, 44 and 58-70 (inclusive) and have oxidation states of +2 or +3 Examples of such metal ions are chromium (III), manganese (II), iron (II), iron (III), cobalt (II), nickel (II), copper (II), praseodymium (III), neodymium (III), samarium (III), gadolinium (III), terbium (III), dysprosium (III), holmium (III), erbium (III) and ytterbium (III)
  • the diagnostic agent may comprise a radiopaque material
  • Suitable radiopaque materials are well known and include iodine compounds, barium compounds, gallium compounds, thallium compounds, and the like Specific examples of radiopaque materials include barium, diatrizoate, ethiodized oil, gallium citrate, iocarmic acid, iocetamic acid, iodamide, iodipamide, iodoxamic acid, logulamide, lohexol, lopamidol, iopanoic acid, ioprocemic acid, iosefamic acid, ioseric acid, iosulamide meglumine,
  • DM VAN/253729-17775/7468072 1 iosumetic acid, lotasul, iotetric acid, iothalamic acid, lotroxic acid, loxaglic acid, loxotriroic acid, ipodate, meglumine, met ⁇ zamide, met ⁇ zoate, propyliodone, and thallous chloride
  • the liposomal formulations are suitable for convection-enhanced delivery
  • Convection-enhanced delivery is a direct intracranial drug delivery technique that utilizes a bulk-flow mechanism to deliver and distribute macromolecules to clinically significant volumes of solid tissues
  • CED offers a greater volume of distribution than simple diffusion and is designed to direct a therapeutic drug to a specific target site
  • CED convection-enhanced delivery
  • CED is a method that circumvents the blood-brain barrier and allows large molecular weight substances, such as drug-loaded liposomes, to be administered uniformly and in a controlled fashion within a defined region of brain (See for example, USSN 11/740,548, incorporated herein in its entirety by reference)
  • CED may be used to administer a fluid pharmacological agent (e g , a liposomal formulation) to a solid tissue (e g , a brain tumor) through direct convective interstitial infusion and
  • CED may be effectively used for the delivery of therapeutic drugs and also optionally diagnostic agents encapsulated in non-PEGylated liposome formulations, where the formulations comprise or consist essentially of a mixture of at least one neutral saturated phospholipid and at least one anionic saturated lipid
  • CED of a composition comprising at least one therapeutic drug (e g , topotecan) and/or diagnostic agent encapsulated in a non-PEGylated liposome formulation as described herein increases the volume of distribution and dramatically improves the serum half-life of the therapeutic drug
  • a suitable apparatus that may be used for administration of a liposomal formulation may comprise a pump device that contains a reservoir filled with the liposomal formulation
  • the pump may be external to the body or implanted within the body
  • the pump may be connected to a catheter, which may be implanted into discrete t ⁇ ssue(s) within the CNS
  • the pump may be activated to release the liposomal formulation at a pressure and flow rate that causes the solute to convect within the specific tissue
  • the duration and other parameters of the infusion may be adjusted to distribute the liposomal formulation throughout the discrete t ⁇ ssue(s) to areas adjacent to the discrete t ⁇ ssue(s), e g , not into the cerebrospinal fluid Depending upon the size and shape of the discrete t ⁇ ssue(s), it may
  • DM VAN/253729-17775/7468072 1 be necessary to use multiple implanted infusion catheters or to use an infusion catheter with multiple solution exit ports.
  • a liposomal formulation may be distributed by slow infusion into the interstitial space under positive pressure through a fine cannula.
  • Bulk flow driven by hydrostatic pressure derived from a pump may be used to distribute the liposomal formulation within the extracellular spaces of the CNS.
  • the blood-brain barrier is bypassed and discrete tissues in the central nervous system may be targeted, including discrete tissue defined, e.g., as cancerous or identified as for resection by a conventional presurgical evaluation, and in different foci if more than one focus are in need of treatment.
  • CED may be used to distribute liposomal formulations reliably, safely, and homogeneously over a range of volumes. See for example USSN 11 ,/740,508. Further, CED does not cause structural or functional damage to the infused tissue and provides greater control over the distribution of the liposomal formulation. Additionally, liposomal formulations may be distributed homogeneously throughout a distribution volume that is proportional to the infusion volume regardless of the molecular weight of the liposomes comprised in the liposomal formulations.
  • an ultrafine delivery catheter (constructed of polyurethane and fused silica in a novel "step" design) may be permanently implanted with a transcutaneous port.
  • the novel catheter design may be rapidly biointegrated and may be internally sealed and filtered to prevent bacterial ingress and capped for further safety.
  • a liposomal formulation may be infused as needed through the port of this catheter system.
  • CED may be applied with a small diameter catheter permanently implanted in the brain region using an infusion pump.
  • Liposomal formulations to be administered may be prepared as an aqueous isotonic solution, or other appropriate formulation. During the administration (e.g., infusion), the liposomal solution may flow within the extracellular space and cause minimal to no damage to the brain tissue.
  • an ultrafine (0.2 mm OD at tip), minimally traumatic catheter system specially designed for transcutaneous CED delivery may be used.
  • the catheter system has a step design, which may eliminate solution reflux along the sides of the catheter. Such solution leakage is a major problem with straight-sided catheters.
  • the catheter system may be constructed of polyurethane and fused silica or Peek Optima so that it is highly biocompatible and does not interfere with MRI signals. Treatment of CNS disorders may require readministration of a liposomal formulation at varying intervals, e.g., weekly intervals, monthly intervals, etc.
  • transcutaneous port may remain capped during the interval period.
  • Multiple catheter designs are feasible so that it may be possible to perfuse a larger area of discrete tissue(s) than is feasible with a single catheter. It has been found that the volume of distribution of liposomes after CED infusion is linearly related to the solution volume infused.
  • DM_VAN/253729-17775/7468072.1 An especially preferred cannula is disclosed in Krauze et al , J Neurosurg November 2005 ,103(5) 923-9, incorporated herein by reference in its entirety, as well as in LJ S Patent Application Publication No US 2007/0088295 A1 , incorporated herein by reference in its entirety, and United States Patent Application Publication No US 2006/0135945 A1 , incorporated herein by reference in its entirety
  • CED comprises an infusion rate of between about 0 1 ⁇ L/m ⁇ n and about 10 ⁇ L/min
  • CED comprises an infusion rate of greater than about 0 1 ⁇ L/m ⁇ n to about 0 3 ⁇ L/m ⁇ n, e g , about 0 2 ⁇ L/min , more preferably greater than about 0 7 ⁇ L/m ⁇ n, more preferably greater than about 1 ⁇ L/min, more preferably greater than about 1 2 ⁇ L/min, more preferably greater than about 1 5 ⁇ L/min, more preferably greater than about 1 7 ⁇ L/min, more preferably greater than about 2 ⁇ L/min, more preferably greater than about 2 2 ⁇ L/min, more preferably greater than about 2 5 ⁇ L/min, more preferably greater than about 2 7 ⁇ L/min, more preferably greater than about 3 ⁇ L/min, and preferably less than about 12 ⁇ L/min, more preferably less than about 10 ⁇ L/min
  • CED comprises incremental increases in flow rate, referred to as "stepping" or up-titration, during delivery
  • stepping comprises infusion rates of between about 0 1 ⁇ L/m ⁇ n and about 10 ⁇ L/min
  • stepping comprises infusion rates of greater than about 0 5 ⁇ L/m ⁇ n, more preferably greater than about 0 7 ⁇ L/m ⁇ n, more preferably greater than about 1 mL/m ⁇ n, more preferably greater than about 1 2 ⁇ L/min, more preferably greater than about 1 5 ⁇ L/min, more preferably greater than about 1 7 ⁇ L/min, more preferably greater than about 2 ⁇ L/min, more preferably greater than about 2 2 ⁇ L/min, more preferably greater than about 2 5 ⁇ L/min, more preferably greater than about 2 7 ⁇ L/min, more preferably greater than about 3 ⁇ L/min, and preferably less than about 12 ⁇ L/min, more preferably less than about 10 ⁇ L/min
  • Treatment methods herein also preferably comprise neuroimaging via a diagnostic agent, preferably MRI, for target localization and guided cannula placement
  • a diagnostic agent preferably MRI
  • a stereotactic holder is used in conjunction with neuroimaging of a diagnostic agent to provide for guided cannula placement at or proximal to a target neuronal population
  • a tracing agent is preferably detectable by magnetic resonance imaging (MRI) or X-ray computed tomography
  • MRI magnetic resonance imaging
  • the distribution of tracing agent is monitored and used as an indirect measure of the distribution of high molecular weight neurotherapeutic This monitoring is done to detect unwanted delivery of infusate to non-target tissue and to verify that the high molecular weight neurotherapeutic is reaching target tissue and achieving an effective concentration therein
  • the diagnostic agent is separate from the therapeutic agent
  • the diagnostic agent is distributed at a rate that correlates with that of the therapeutic agent and thus is an indirect indicator of therapeutic distribution
  • DM VAN/253729 17775/7468072 1 therapeutic agent are separately administered but encapsulated by the same non-PEGylated anionic liposomal formulation, which confers highly similar distribution characteristics
  • the diagnostic agent and the therapeutic agent are co-administered
  • Treatment methods herein also preferably comprise neuroimaging for monitoring infusate distribution
  • a treatment method comprises the use of MRI for monitoring distribution of an infused pharmaceutical composition of the invention, wherein the pharmaceutical composition comprises an MRI magnet
  • Example 1 1 1 DSPC/CHOL (60/40 mole ratio)
  • Example 1 1 2 DSPC/CHOL/PEGzoooDSPE (59 5/40/0 5 mole ratio)
  • DM VAN/253729-17775/7468072 1 [0105] Rehydrate the lipids at 6O 0 C in 1 5 ml HBS (5 mM HEPES-145 mM NaCI pH 7 0) to form MLVs
  • Example 1 1 3 DSPC/CHOL/PEG 20 o 0 DSPE (55/40/5 mole ratio)
  • Example 1 1 4 DSPC/CHOL/NG-DOPE (55/40/5 mole ratio)
  • Example 1.1.6 DSPC/PEG 200 oDSPE (95/5 mole ratio)
  • Example 1.1.7 DSPC/DSPG (70/30 mole ratio)
  • Gadodiamide (GD) for Ls-GD preparation was obtained from Beijing SHLHT Science & Trade (Beijing, China) Ls-GD was prepared similarly to Ls-TPT, except that the gadodiamide was passively encapsulated in the liposomes
  • the internal buffer solution consisted of 520 mM gadodiamide, pH 3 5 instead of 250 mM ammonium sulfate, pH 5 5 Assuming an encapsulation efficiency of 4-6%, a gadodiamide to lipid ratio of 0 3 (w/w) and a particle size of 75 to 120 nm were targeted
  • the final formulation lipid and gadodiamide concentrations were 51 1 mg/mL and 17 0 mg/mL, respectively
  • Ls-TPT test articles were stored frozen (-20 to -3O 0 C) Dosing solutions were prepared fresh on the day of dosing and kept at room temperature Appropriate dilutions with 5 mM histidine, 145 mM NaCI pH 6 0, 300 mM sucrose of stock solution (Ls-TPT and free topotecan) were performed to yield the desired concentrations Fresh vials of the stock test article solution were used on each dosing day
  • the amount of lipid required for the batch was calculated and the lipid powders were weighed into weighing boats A solvent solution consisting of t-butanol, ethanol and water (45 45 10 vol/vol) was prepared and heated to 70 0 C While stirring, the lipid powders were added to the solvent solution The solvent was maintained at 7O 0 C and stirred until all the lipids were dissolved ( ⁇ 1 hour) The concentration of lipids in solution at that point was 320 mg/mL A 250 mM solution of ammonium sulphate was prepared (volume was nine times that of the lipid solvent solution) and heated to 7O 0 C After the ammonium sulphate had reached temperature, the lipid solution was poured into the ammonium sulphate solution while stirring to generate multilamellar vesicles (MLVs) The MLVs were maintained at 7O 0 C and extruded through 4-stacked polycarbonate filters with 80 nm pores Two passes were required to generate large unilamellar vesicles
  • This step also served to exchange the external buffer from sodium chloride solution to sucrose which acted as a cryo-protectant and allowed the formulation to be frozen without changing its physical characteristics
  • the estimated lipid content at this stage was 8 3 mg/mL (for the 0 3 1 drug lipid ratio)
  • the formulation was heated to 5O 0 C and passed through a 0 2 ⁇ m syringe filter The product was then vialed The product was finally frozen, completing the manufacturing process
  • the animals were divided in 4 groups based on Ls-TPT formulations or free topotecan as outlined in Table 1
  • DSPC/DSPG d ⁇ stearoylphosphat ⁇ dylchol ⁇ ne/d ⁇ stearoylphosphat ⁇ dylglycerol
  • D L ratio drug lipid ratio (w/w)
  • Ls-TPT liposomal topotecan
  • Ls-GD liposomal gadodiamide
  • Rats were assigned to groups based on body weight in a manner to achieve comparable group mean body weights and standard deviations The groups were then to be randomly assigned to treatment and time point
  • Example 2 1 4 Surgical Procedures
  • Rats were anesthetized with either isoflurane (5% for induction, 2 5 to 3 0% for maintenance during surgery) inhalation or a combination of ketamine (60 mg/kg) and xylazine (8mg/kg) via an intraperitoneal injection
  • the skin over the cranium was shaved and the animal mounted in a stereotaxic frame with the head positioned by the use of ear bars and the incisor bar Aseptic techniques were used for all surgical procedures
  • the skin was disinfected with 70% alcohol followed by betadine solution
  • a longitudinal incision was made in the skin on top of the skull and blunt dissection was used to remove connective tissue overlying the skull Craniectomy was performed using a small electric dental drill with 1-mm diameter burr holes, 0 5 mm anterior and 3 mm left and
  • DM VAN/253729 17775/7468072 1 right from the bregma A fused silica cannula (OD 168 /vm, ID 102 ⁇ m) (PolyMicro Technologies, Phoenix, AZ) connected to an automated pump (BASi, lnc , West Lafayette, IN) was used for CED and was lowered to the dorso-ventral appropriate coordinates (-4 5 to -5 mm with the tooth bar at -3 3 mm) Dorso-ventral coordinates were calculated from the pial surface
  • the cannula was inserted into a 27-gauge needle connected with a 10- ⁇ L Hamilton syringe and secured with superglue on the tubing
  • the test article was injected bilaterally at one site into each striatum
  • a progressive infusion rate increment was used in this study to achieve a 20 ⁇ L dose per hemisphere with 0 2 ⁇ L/min (15 mm) followed by 0 5 ⁇ L/min (10 mm) and
  • Rats were maintained in a draft free environment, and kept warm via heating lamp or water bottle or other appropriate warming methods and monitored during anesthesia recovery Buprenorphine was administered subcutaneously on an as needed basis Rats were allowed to recover in the procedure room prior to return to their home cages
  • Example 2 1 5 Tissue collection and processing
  • HPLC high performance liquid chromatography
  • Example 2 2 2 Formulation screening pharmacokinetics
  • DSPC/DSPG distearoylphosphatidylchohne/distearoylphosphatidylglycerol
  • D L ratio drug lipid ratio (w/w)
  • Ls-TPT liposomal topotecan
  • Ls-GD liposomal gadodiamide
  • Topotecan brain tissue concentrations were measurable at 1 and 6 hours only in the free topotecan group (formulation 4) In contrast, measurable brain tissue concentrations were found through 48 hours (formulation 1 ) or even 96 hours (formulations 2 and 3) in the Ls-TPT groups None of the formulations had detectable levels at 7 days At all time points, formulation 2 had the highest tissue concentrations, except at 96 hours where formulations 2 and 3 had very low and similar concentrations The topotecan levels detected are assumed to reflect encapsulated topotecan for liposomal formulations 1 , 2 and 3, particularly beyond 6 hours, given the short half life of free topotecan Table 4 summarizes the brain tissue concentrations of topotecan by formulation and time point
  • DSPC/DSPG distearoylphosphatidylcholine/distearoylphosphatidylglycerol
  • D:L ratio drug:lipid ratio (w/w)
  • Ls-TPT liposomal topotecan
  • Ls-GD liposomal gadodiamide
  • the AUC(O-last) was markedly larger for the DSPC/DSPG/Chol 0.3 D:L ratio formulation (153.8 ⁇ g « day/g) compared to DSPC/Chol 0.1 and DSPC/DSPG/Chol 0.1 (38.27 and 68.21 ⁇ g « day/g, respectively), and free topotecan (5.5 ⁇ g « day/g). All the nanoliposomal formulations yielded half-lives in the range of one day while the half-life of free topotecan was much shorter. Based on these results, the Ls-TPT formulation 2 (DSPC/DSPG/Chol 0.3 D.L ratio) was selected for further study.
  • Example 2 evaluated the pharmacokinetic profiles in rat normal brain tissue of a combined drug delivery approach comparing 3 novel Ls-TPT formulations and free topotecan delivered via intracerebral CED.
  • formulation 2 DSPC/DSPG/Chol with drug to lipid ratio of 0.3 and a topotecan concentration of 0.5 mg/mL, was determined to result in the most optimal intracerebral pharmacokinetic profile with an AUC(O-last) of 153.8 ⁇ g-day/g and a half-life of approximately one day.
  • Ls- TPT formulation 2 (DSPC/DSPG/Chol 0.3 D:L ratio) far exceeded that of free topotecan indicating longer drug release kinetics from the liposome, a desirable characteristic for CED delivery.
  • the better pharmacokinetic profile observed for Ls-TPT formulation 2 is likely related to better drug release characteristics with slower release from liposomes of the active drug.
  • DM VAN/253729-17775/7468072 1 [0200]
  • Rhodamine liposomes (DSPC/DSPG/ Choi, 70:20:10 mole ratio) with 0.5 mole% rhodamine PE were delivered bilaterally to the rat striatum by CED infusion.
  • Dilutions of rhodamine liposomes were prepared using histidine/saline buffer and added sucrose to achieve final sucrose concentrations of 3 mM 15 mM and 5 mM according to Table 7.
  • the test article was injected bilaterally at one site into each striatum.
  • the infusion rates used in this study to achieve a 20 ⁇ L dose per hemisphere were 0.2 ⁇ L /min (15 min) + 0.5 ⁇ L /min (10 min) + 0.8 ⁇ L /min (15 min). Rats were sacrificed immediately following CED delivery. The brains were removed and divided into left and right hemispheres.
  • Right hemispheres were frozen at -60 0 C in dry ice/isopentane and stored at -80°C for 24h prior to histological analysis.
  • the left hemispheres of each animal were frozen at -80°C for subsequent analysis by Northern Lipids, Inc. In some of the rats, the striatum was removed from the left hemisphere for analysis.
  • Rhodamine fluorescence was detected in all rats receiving CED infusions. In all rats, the label distributed within the striatum. Some rats showed strong labeling in the corpus callosum and the internal capsule fiber tracks (data not shown). Table 8 indicates the volume of distribution (Vd) for individual rats at the sucrose concentrations of 3mM, 15 mM and 75 mM. Due to technical difficulty with the CED tubing, three rats were bilaterally infused with 40 uL of rhodamine liposomes into each striatum (shaded area) rather than 20 uL into each striatum. These rats were not included in the analysis. One animal in the 75 mM group died during surgery (at 5 min) and was not included. Infusion was continued on this animal, however, the liposomes were extruded from the site following the animal's death and did not distribute into the parenchyma.
  • Rhodamine fluorescence was detected in all rats receiving CED regardless of sucrose concentration with both 40 uL and the 20 uL infusion volumes (Table 9) The mean volumes of distribution ranged from 12 6 mm3 to 24 9 mm 3 in all groups
  • DM VAN/253729 17775/7468072 1 high degree of variability in the procedure that is likely to be related to technical aspects of the infusion procedure Moreover, the distribution of liposomes to adjacent structures and fiber tracts close to the striatum in rat may account for the within group differences noted in both studies , since distribution of liposomes outside of the striatal region was not included in the Vd calculations
  • GLP grade material of both Ls-TPT and Ls-GD were prepared as indicated in Examples 1 1 and 1 2
  • Rats were assigned to groups based on body weight in a manner to achieve comparable group mean body weights and standard deviations. The groups were then randomly assigned to treatment regimen. Single treatment was planned 8 days post tumor implantation and dual treatment at 8 and 12 days post tumor implantation.
  • Example 4.1.3 Surgical Procedures and Treatment
  • Example 4.1.3.1 Intracranial Tumor Xenograft Implantation
  • U87MG tumor cells human glioblastoma cells; Perry Scientific Inc, San Diego, CA, lot W5051507U87MC
  • Rats were anesthetized with isoflurane (2.5%) and the skin over the cranium was shaved.
  • the rat was mounted in a stereotaxic frame with the head positioned by the use of ear bars and the incisor bar. Aseptic techniques were used for all surgical procedures.
  • the skin was disinfected with Betadine solution. A longitudinal incision was performed in the skin on top of the skull and blunt dissection was used to remove connective tissue overlying the skull.
  • a small dental drill was used to drill a burr hole burr hole 0.5 mm anterior and 3.0 mm lateral from the bregma.
  • U87MG cells were stereotactically injected into the striatum using the appropriate dorso-ventral coordinates from pial surface (-4.5 to -5 mm with the tooth bar at -3.3 mm).
  • a total volume of 10 ⁇ L containing approximately 5.0 X 10 5 cells total was injected in the right striatum over a period of 10 minutes.
  • the tumor implantation was done on 2 different days because the number of animals planned did not allow performing all interventions on one single day. Therefore, 2 separate tumor suspensions were prepared.
  • Rats were maintained in a draft free environment, and kept warm via heating lamp or water bottle or other appropriate warming methods and monitored during anesthesia recovery Buprenorphine was administered subcutaneously on an as needed basis Rats were allowed to recover in the procedure room prior to return to their home cages
  • Example 4 1 4 Euthanasia Criteria Before Day 60
  • mice were grouped by treatment arm
  • animals in the highest topotecan total dose group (group 2) were compared to all other treatment arms combined including the control group
  • the latter grouping was also performed within the approximate U87MG cell load groups as described below Since the number of U87MG cells implanted potentially varied as animals were treated on 2 different days with preparation of 2 separate tumor cell suspensions without pre-implantation cell count, treatment groups were analyzed by tumor cell suspensions and therefore indirectly by approximate U87MG cell load at tumor implantation based on
  • DM VAN/253729 17775/7468072 1 the post-implantation cell count (see Example 4.2.1 ).
  • the Log-rank test was used to compare survival among the different groups.
  • Each animal was identified by a numbered ear tag. Additionally, each animal's cage was identified by a cage card listing the animal identification number, study number, group, and sex of the animal.
  • the animals were housed individually in microisolator cages so they did not disturb each other's wounds.
  • the room(s) in which the animals were kept were documented in the study records. No other species was housed in the same room(s).
  • the rooms were well ventilated (greater than 10 air changes per hour) with 100% fresh air (no air recirculation).
  • a 12-hour light/12-hour dark photoperiod was maintained, except when room lights had to be turned on during the dark cycle to accommodate blood sampling or other study procedures.
  • Room temperature was maintained between 18 and 26°C.
  • Post-implantation cell counts revealed that the actual numbers of U87MG tumor cells implanted were significantly higher than stipulated by the protocol. Also, the tumor cell density differed markedly between the two suspensions prepared. Specifically, the post-implantation counts for the two suspensions were 6.8 X 10 5 and 9.7 X 10 5 cells per 10 ⁇ L, as compared to the protocol-specified number of 5.0 X 10 5 . The observed differences are presumably attributable to cell growth between suspension preparation and cell count. Conceivably, the respective pre-implantation counts may therefore have been lower and less different, but it seems unlikely that they were much closer to the protocol-specified number. In order to account for these differences in the analysis of the results, treatment groups were analyzed by approximate U87MG cell load at tumor implantation based on the post-implantation cell count as described in Example 4.1.8.
  • DM VAN/253729-17775/7468072 1 [0247] Four animals, two assigned to group 1 , one to group 3 and one to group 4 died before tumor implantation probably related to anesthesia performed for the procedure Within the tumor implanted groups (29 animals), four animals were found dead in their cage during the course of the study One animal was assigned to group 1 , one to group 2 and two to group 4 The 2 animals assigned to group 4 had the high tumor cell load implanted while the others had the low tumor cell load The other 25 animals were euthanized because they appeared in poor condition, the most common signs being weight loss ⁇ 5% in the great majority of the animals, lethargy, hunched back posture, motor deficits, tremor and laborious breathing
  • Example 4 evaluated the efficacy of a combined drug delivery approach using a novel Ls-TPT formulation delivered to an intracranial glioma xenograft model in athymic rats by intracerebral CED
  • This example used 2 dose levels one previously reported safe by another group, 0 5 mg/mL (Saito 2006), and a lower one at 0 1 mg/mL
  • 2 dosing regimens were assessed single dosing for 0 5 mg/mL and dual dosing 4 days apart for both dose levels studied as only single dosing has been studied thus far Longer overall and median survivals were observed for the highest Ls-TPT total topotecan dose (0 5 mg/mL dual dosing) compared to the other groups, individually (not statistically significant) or combined (statistically significant)
  • a dose dependent effect was also observed when comparing total dose accounting for dose levels and number of dosing
  • Example 5 Cytotoxicity of topotecan and liposomal topotecan on U87MG cells
  • Example 5.1.1 Test Articles:
  • Free topotecan formulations were obtained from GlaxoSmithKline (Research Triangle Park, NC) and Hisun Pharmaceuticals (Taizhou City, Zhejiang, China).
  • Topotecan for GLP-grade Ls-TPT formulation preparation was obtained from Hisun Pharmaceuticals (Taizhou City, Zhejiang, China).
  • liposomes were composed of distearoylphosphatidylcholine (DSPC), distearoylphosphatidylglycerol (DSPG) and cholesterol at a 7:2:1 molar ratio with 75 to 90 nm target size.
  • Topotecan was remotely loaded (actively encapsulated) into liposomes in response to a transmembrane pH gradient using internal and external buffers consisting of ammonium sulfate 250 mM pH 5.5 and histidine 10 mM/NaCI 145 mM pH 6.0 respectively.
  • a topotecan concentration 2.0 mg/mL and a 0.3 (w/w) drug.lipid ratio were targeted assuming a 90-95% drug encapsulation efficiency.
  • Gadodiamide for Ls-GD preparation was obtained from was obtained from Estech Pharma, Ansan-Si, Gyeonggi-Do, Korea.
  • GLP-grade Ls-GD was prepared similarly topoCED, except that the GD was passively encapsulated in the nanoliposomes. Following removal of un-encapsulated GD and solvents by diafiltration, the final GD encapsulation was >90%.
  • the target GD content was 5.0 mg/mL ⁇ 10 % and a particle size range of 75 to 120 nm.
  • Test articles of Ls-TPT were stored frozen (-20 to -30 0 C) and Ls-GD were stored refrigerated (2 to 8 0 C), respectively, and protected from light. Test article solutions were prepared fresh on the day of dosing and kept at room temperature. Appropriate dilutions of the test article stock solution with 5 mM histidine, 145 mM NaCI pH 6.0, 300 mM sucrose or 0.9% saline were performed to yield test solutions at appropriate concentrations at the desired test volumes.
  • U87MG human glioblastoma cell line was used for all experiments (UCSF culture facility, San Francisco, CA). The cells were established in T175 Falcon flasks (BD Bioscience, San Jose, CA). The cells were maintained in complete minimal essential medium (CMEM), consisting of Eagle's minimal essential medium (MEM) supplemented with 10% fetal bovine serum, non-essential amino acids and antibiotics (streptomycin 100 ⁇ g/mL, penicillin 100 DImL). All media components were from UCSF cell culture facility. Cultures were incubated at 37°C in a humidified chamber with 5% CO ⁇ . Once a 95% confluence was achieved, cells were trypsinized briefly with 0.05% trypsin-0.02%
  • DM VAN/253729-17775/7468072.1 ethylenediaminetetra-acetic acid (UCSF culture facility, San Francisco, CA), and cells were centrifuged at 500 X g for 10 minutes. After the supernatant was aspirated, the cells were resuspended directly in 5 ml of a complete cell growth medium (with antibiotics and 10% fetal bovine serum). The cell count was done with trypan blue in a hematocyter (Hausser Scientific, Horsham, PA).
  • the appropriate amount of complete cell growth medium was added to achieve a final concentration of 10,000 cells in 100 ⁇ L for transfer in each well of 96-well plates designed for luminescence-based cell viability assay (CellTiter-GloTM, Promega, Madison, Wl). Cells were allowed to attach for 24 hours before any exposure to test article. The culture medium was removed from the 96-well plates just before adding 100 ⁇ L of test article using a 12-multichannel pipettor. After exposure to test article, cytotoxic assays were conducted at 24, 48 and 72 hours. All time points of each test article and control were run in triplicates.
  • Table 15 outlines the different test article and concentrations evaluated along with controls.
  • test article dilutions were performed with the culture medium used for U87MG culture.
  • the assay is based on quantification of the ATP present, as an indicator of metabolically active cells using a thermostable form of luciferease.
  • the luciferase uses luciferin, oxygen and ATP as substrates in a reaction producing oxyluciferin and releasing energy in the form of light.
  • the amount of light produced is proportional to the amount of ATP present, reflecting the number of viable cells.
  • 20 ⁇ L of CellTiter-Glo luminescent cell viability assay reagent Promega, Madison, Wl
  • Example 6 Convection Profile and Tissue Distribution in Normal Brain and Xenografted U87MG Tumors of Different Formulations of Liposomal Topotecan and Liposomal Gadodiamide Administered by Intracerebral Convection-Enhanced Delivery to the Adult Athymic Rat
  • Example 6.1.1 Test Articles
  • Ls-TPT and Ls-Gd were prepared as described in Examples 2.1.1 and 2.1.2. Gadodiamide for Ls-GD preparation was obtained from Estech Pharma, Ansan-Si, Gyeonggi- Do, Korea. Ls-GD was prepared similarly topoCED, except that the GD was passively encapsulated in the nanoliposomes. Following removal of un-encapsulated GD and solvents by diafiltration, the final GD encapsulation was >90%. The target GD content was 5.0 mg/mL ⁇ 10 % and a particle size range of 75 to 120 nm.
  • DM VAN/253729-17775/7468072 1 added to the lipid powder at the same time as the solvent solution based on a DSPC:DSPG:cholesterol:fluorophore molar ratio of 69.7:20:10:0.3.
  • Test articles of Ls-TPT were stored frozen (-20 to -30°C) and Ls-GD were stored refrigerated (2 to 8 "C), respectively, and protected from light. Dosing solutions were prepared fresh on the day of dosing and kept at room temperature. Appropriate dilutions with 0.9% saline of the test article stock solution were performed to yield the desired concentrations. Fresh vials of the stock test article solutions were used on each dosing day. No control article was used in this study.
  • Group 1 Na ⁇ ve brain tissue - DSPC/DSPG/Chol D:L 0.1 + Ls-TPT 0.38 mg/mL + Ls-GD 1.15 mg/mL
  • Group 2 Na ⁇ ve brain tissue - DSPC/DSPG/Chol D:L 0.3 + Ls-TPT 1.02 mg/mL + Ls-GD 1.15 mg/mL
  • Group 3 Tumor tissue - DSPC/DSPG/Chol D:L 0.1 + Ls-TPT 0.38 mg/mL + Ls-GD 1.15 mg/mL
  • DSPC/DSPG distearoylphosphatidylcholine/ distearoylphosphatidylglycerol
  • Ls-TPT liposomal topotecan
  • Ls-GD liposomal gadodiamide
  • rats were assigned to the formulation and tissue groups based on body weights in a manner so as to achieve comparable group mean body weights and standard deviations. Animals in both formulation groups were to receive CED infusions on Day 1 if they were assigned to the na ⁇ ve brain tissue group, and on Day 10, if they were assigned to the tumor tissue group.
  • Example 6.1.3 Surgical Procedures and Treatment
  • Example 6 1 3 1 Intracranial Tumor Xenograft Implantation
  • the test articles were administered via CED infusion on study Day 1 in rats assigned to the naive brain tissue groups, and on Day 10 in rats assigned to the tumor tissue groups
  • Doses were administered via CED bilaterally to the dorsolateral striatum of the rats in the naive tissue groups, and intratumorally in the rats in the tumor tissue groups using the same coordinates that were used for the tumor implantation
  • Rats were dosed in a systematic order that distributed the time of dosing similarly across all groups
  • Anesthesia was performed with either isoflurane (2 5%) or a combination of ketamine (90 mg/kg) and xylazine (12mg/kg) via an intraperitoneal injection
  • a stereotactic frame with blunt ear bars was used to perform CED
  • bilateral burr holes were created as outlined in section 5 3 1
  • rats assigned to the tumor tissue groups the scalp incision was reopened to visualize the previously prepared burr hole Only the blood clots were removed
  • DM VAN/253729 17775/7468072 1 withdrawn. Following completion of the procedure, the rats were maintained in a draft free environment, and kept warm via heating lamp or water bottle or other appropriate warming methods and monitored during anesthesia recovery. Buprenorphine was administered subcutaneously on an as needed basis.
  • the brain was removed, incubated in 4% paraformaldehyde for up to 24 h, and then equilibrated in 30% sucrose. Following sucrose equilibration, the tissue was frozen at -60°C in a mixture of dry ice and isopentane and stored at -70°C for subsequent processing.
  • Example 6.1.3.5 Histopathological analyses and volume of distribution assessment
  • the convection profiles and tissue distribution of both Ls-TPT and Ls-GD were determined by means of fluorescence microscopy, the image captured using a SPOT camera, SPOT software and a Macintosh G4 computer, and the volume of distribution (Vd) of both marina blue-DHPE and rhodamine-PE fluorophores in the sections was calculated using Macintosh-based image analysis system [ImageJ, National Institute of Health (NIH), Bethesda, MD].
  • Region of interests (ROI) were drawn using NIH image software and distribution data was transferred to an excel spreadsheet.
  • Distribution volumes (mm 3 ) were calculated by multiplying the mean ROI area (mm 2 ) and the distribution distance (mm). Remaining sections were stored at 4°C and could be used for additional immunohistochemical analyses.
  • Rats receiving intracerebral injections of U87MG tumor cells typically have a life span of 17- 25 days, and they remain asymptomatic until shortly before death Although unlikely given the early sacrifice at Day 10 in this study, if any one or combination of the symptoms (nasal/pe ⁇ orbital bleeding, paresis, hunching, inactivity or not feeding or grooming or weight loss >15% of baseline body weight) were observed, the animal could be euthanized If possible, blood or other specimens were collected and analyzed as appropriate (e g , for clinical pathology parameters) to help reveal the cause of malaise/morbidity
  • each animal was identified by a numbered ear tag Additionally, each animal's cage was identified by a cage card listing the animal identification number, study number, group, source, arrival date, species/strain, date of birth and sex of the animal
  • the animals were housed individually in isolator cages
  • the bedding material was shaved hardwood chips (Sanichips, Harlan, CA) and was changed weekly
  • Room temperature was centrally maintained at 18-26°C (64-79°F), with relative humidity at 30-70%
  • Temperature and humidity were continuously monitored and daily minimums and maximums recorded
  • a 12-hour l ⁇ ght/12-hour dark cycle illumination period was maintained, except when room lights had to be turned on (during the dark cycle) to accommodate study procedures
  • the rats were to have ad libitum access to irradiated Teklad Global 18% Protein Rodent Diet (Harlan, San Diego, CA, USA) and municipal tap water throughout the study period No contaminants were known to be present in the diet or water at levels that would have a deleterious effect on the results of the study Records of annual water quality testing are maintained in the PSI archives
  • DM VAN/253729- 17775/7468072 1 [0310] Upon arrival at the designated housing, all rats accepted for receipt following an initial health inspection were allowed to acclimatize to the housing environment (pnmary enclosure and room) for a minimum of 3 days prior to initiating any animal-related study procedures During the acclimatization period, the general health of the rats was monitored daily Only rats that were visually appraised to be in good clinical condition ( ⁇ e , within body weight specifications) were enrolled in the study Any rats that appeared abnormal and exhibited signs of poor health ( ⁇ e , ruffled coat, significantly low body weight) were excluded from the study
  • Example 6 2 3 Convection Profiles and Tissue Distribution
  • Group 1 Naive brain tissue - DSPC/DSPG/Chol D.L 0.1 + Ls-TPT 0.38 mg/mL + Ls-GD 1.15 mg/mL
  • Group 2 Naive brain tissue - DSPC/DSPG/Chol D:L 0.3 + Ls-TPT 1.02 mg/mL + Ls-GD 1.15 mg/mL
  • Vd values of Ls-TPT-marina blue DHPE for both formulations were in a tight range in the naive brain tissue groups (means of 39.0 ⁇ 3.0 and 38.5 ⁇ 5.6 mm 3 for D:L 0.1 :1 and 0.3:1 , respectively), with corresponding Vd:Vi ratios of 1.9 2.0. In contrast, the Vd values were markedly
  • DM VAN/253729-17775/7468072.1 smaller and generally more variable in the tumor tissue groups, with means of 25 8 mm 3 + 10 6 and
  • This Example 6 evaluated the convection profile and volumes of distribution of two formulations of a therapeutic nanoliposomal compound, Ls-TPT, and an imaging tracer surrogate for Ls-TPT, Ls-Gd, using different fluorophores to co-label these liposomes in order to demonstrate any differential tissue distribution
  • GLP grade material of both Ls-TPT and Ls-GD were prepared as described in Example 6 1 1
  • Rats were assigned to groups based on body weight in a manner to achieve comparable group mean body weights and standard deviations
  • Rats were dosed in a systematic order that distributed the time of dosing similarly across both groups Rats were anesthetized with either isoflurane (5% for induction, 2 5 to 3 0% for maintenance during surgery) inhalation or a combination of ketamine (90 mg/kg) and xylazine (12 mg/kg) via an intraperitoneal injection
  • isoflurane 5% for induction, 2 5 to 3 0% for maintenance during surgery
  • ketamine 90 mg/kg
  • xylazine (12 mg/kg) via an intraperitoneal injection
  • the skin over the cranium was shaved and the animal mounted in a stereotaxic frame with the head positioned by the use of ear bars and the incisor bar Aseptic techniques were used for all surgical procedures
  • the skin was disinfected with 70% alcohol followed by betadine solution
  • a longitudinal incision was made in the skin on top of the skull and blunt dissection was used to remove connective tissue overlying the skull Craniectomy was performed using a small
  • Example 7 1 4 Tissue Collection and Processing
  • the brains were removed, equilibrated in 30% sucrose and subsequently frozen at 6O 0 C in a mixture of dry ice and isopentane Brains were stored at 70 0 C for subsequent processing
  • DM VAN/253729 17775/7468072 1 fluorescence detection and gadodiamide plasma levels by inductively coupled plasma mass spectroscopy (ICP-MS)
  • Each animal was identified by a numbered ear tag and by cage cards specifying the animal identification number, study number, species/strain, sex, date of birth, source, and arrival date
  • the animals were housed individually in isolator cages
  • the bedding material was shaved hardwood chips (Sanichips, Harlan, CA) and was changed weekly
  • Room temperature was centrally maintained at 18-26°C (64-79T), with relative humidity at 30-70%
  • Temperature and humidity were continuously monitored and daily minimums and maximums recorded
  • a 12-hour l ⁇ ght/12-hour dark cycle illumination period was maintained, except when room lights had to be turned on (during the dark cycle) to accommodate study procedures
  • the rats were to have ad libitum access to irradiated Teklad Global 18% Protein Rodent Diet (Harlan, San Diego, CA, USA) and municipal tap water throughout the study period No contaminants were known to be present in the diet or water at levels that would have a deleterious effect on the results of the study Records of annual water quality testing are maintained in the PSI archives
  • Example 7.2.2 Topotecan Plasma Level Measurements
  • Plasma extract measurements 7 days after the last treatment revealed that both topotecan and gadodiamide levels were either absent or below the lower limit of quantification.
  • a minute peak at the retention time of topotecan lactone form which was present in all samples could not clearly be attributed to topotecan or plasma.
  • noticeable plasma levels would be somewhat unexpected given the loco-regional delivery method bypassing the blood brain barrier and the time between last treatment and sample collection.
  • Brain tissue concentrations were not measured in this study because the brains were sectioned for histopathological analysis. It is therefore impossible to conclude whether the above peak in plasma was correlated with persisting brain parenchymal levels.
  • no intracerebral topotecan was detected at 7 days after a single treatment with Ls-TPT in both hemispheres at a topotecan concentration of 0.5 ⁇ g/mL (Example 2).
  • Ls-TPT at concentrations of 1.0 and 1.6 mg/mL co-infused with Ls-GD appears safe with no evidence of changes attributable to the test article in rat naive brain tissue.
  • Topotecan and gadodiamide plasma levels were below the lower level of quantitation for the assay consistent with the delivery method and drug properties.
  • Example 8 Convection-Enhanced Delivery of Liposomal Topotecan and Liposomal Gadodiamide To Intracranial Xenografted U87MG Tumors in the Adult Athymic Rat
  • GLP grade material of both Ls-TPT and Ls-GD were prepared as described in Example 6.1.1.
  • Example 8.1.3 Surgical Procedures and Treatment
  • Example 8.1.3.1 Intracranial Tumor Xenograft Implantation
  • Human glioblastoma cells (U87MG) were obtained from frozen cell stock (Perry Scientific Inc, San Diego, CA) two weeks prior to the scheduled inoculation. Cells were harvested on the day of tumor inoculation surgery and adjusted to a concentration of 50,000 to 100,000 cells/ ⁇ L. On the day of inoculation (Day 0), each rat was implanted with a total of 500,000 U87MG tumor cells unilaterally into the right striatum using a 5-10 ⁇ L of suspension. A stereotaxic technique and anesthesia with isoflurane (2.5%) were used. The rat was mounted in a stereotaxic frame with the head positioned by the use of ear bars and the incisor bar. Aseptic techniques were used for all surgical procedures.
  • the skin was disinfected with 70% alcohol followed by betadine solution.
  • a longitudinal incision was made in the skin on top of the skull and blunt dissection was used to remove connective tissue overlying the skull.
  • a burr hole was drilled 0.5 mm anterior and 3.0 mm lateral from the bregma.
  • U87MG cells were stereotaxically injected into the striatum using the appropriate dorso-ventral coordinates from pial surface (-4.5 to -5 mm with the tooth bar at -3.3 mm).
  • the volume of injection was adjusted between 5 and 10 ⁇ L to ensure that a total of 500,000 ⁇ 25,000 cells be delivered over a period of 10 minutes.
  • DM VAN/253729-17775/7468072.1 [0375] Following inoculation, the skin was stapled. The rats were monitored during anesthesia recovery. Buprenorphine was administered subcutaneously (SC) before the end of the procedure then buprenorphine was administered SC on an as needed basis. Rats were monitored twice daily following tumor cell implantation. The survival time following implantation was expected to be approximately 0-60 days, wherein the animal was euthanized and the brain harvested.
  • a fused silica cannula (OD 168 ⁇ m, ID 102 ⁇ m) (PolyMicro Technologies, Phoenix, AZ) connected to an automated pump (BASi, Inc., West Lafayette, IN) was used for CED and was lowered to the dorso-ventral appropriate coordinates (-4.5 to -5 mm with the tooth bar at -3.3 mm). Dorso-ventral coordinates were calculated from the pial surface.
  • the cannula was inserted into a 27-gauge needle and secured with superglue on the tubing. A progressive infusion rate increment was used.
  • the infusion rates used in this study to achieve a 20 ⁇ l_ volume per treatment were 0.2 ⁇ l_ /min for 15 min, 0.5 ⁇ l_ /min for 10 min and 0.8 ⁇ l_ /min for 15 min. Following infusion the cannula was left in place for 5 minutes to avoid infusate outflow, and then slowly withdrawn.
  • the rats were maintained in a draft free environment, and kept warm via heating lamp or water bottle or other appropriate warming methods and monitored during anesthesia recovery. Buprenorphine was administered subcutaneously on an as needed basis. Rats were allowed to recover in the procedure room prior to return to their home cages.
  • Example 8.1.3.3 Euthanasia Criteria Before Day 60
  • DM VAN/253729-17775/7468072 1 external surfaces and orifices, cranial cavity and external surface of the brain, neck with associated organs and tissues, thoracic, abdominal and pelvic cavities with their associated organs and tissues.
  • the brain was removed, incubated in 4% paraformaldehyde for up to 24 h, and then equilibrated in 30% sucrose. Following sucrose equilibration, the tissue was frozen and stored at -70°C until cryosectioning. The heart, lungs, liver, kidneys, spleen (or portions of), when present, were also to be collected and preserved. These tissues were fixed in neutral-buffered 10% formalin. Formalin fixed organs were then to be grossed and processed to paraffin blocks for subsequent histopathological analyses if required.
  • At least three randomly selected brains from both survivors and non-survivors at 60 days in each of the 3 treatment groups were sectioned with a 20 ⁇ m thickness and every fourth section was collected onto glass slides, fixed in 4% paraformaldehyde and processed for hematoxylin/eosin to assess the size and histology of the tumor mass. Remaining sections were stored at 4°C and could be used for additional immunohistochemical analyses.
  • Clinical observations and measurements were performed at least once daily throughout the acclimation and study period. Recording of cage side observations were to commence at least 3 days prior to the first dose and were to continue until termination. Each animal was observed for changes in general appearance and behavior. The clinical observations and measurements are outlined in Table 22.
  • DM VAN/253729-17775/7468072 1 immediately, the animal was refrigerated (not frozen) to minimize tissue autolysis. The necropsy should be performed no later than 12 hours after death.
  • Each animal was identified by a numbered ear tag. Additionally, each animal's cage was identified by a cage card listing the animal identification number, study number, group, source, arrival date, species/strain, date of birth and sex of the animal.
  • the animals were housed individually in isolator cages.
  • the bedding material was shaved hardwood chips (Sanichips, Harlan, CA) and was changed weekly.
  • Room temperature was centrally maintained at 18-26°C (64-79°F), with relative humidity at 30-70%. Temperature and humidity were continuously monitored and daily minimums and maximums recorded. A 12-hour light/12-hour dark cycle illumination period was maintained, except when room lights had to be turned on (during the dark cycle) to accommodate study procedures.
  • the rats were to have ad libitum access to irradiated Teklad Global 18% Protein Rodent Diet (Harlan, San Diego, CA, USA) and municipal tap water throughout the study period. No contaminants were known to be present in the diet or water at levels that would have a deleterious effect on the results of the study. Records of annual water quality testing are maintained in the PSI archives.
  • DM VAN/253729-17775/7468072 1 Longer overall and median survivals were observed for both active treated groups. As compared to controls, the higher Ls-TPT concentration (1.0 mg/mL) resulted in a highly statistically significant increase in overall survival (p ⁇ 0.0001 ), with a 65% and 76% increase in median and mean survival, respectively. The lower Ls-TPT concentration (0.5 mg/mL) also produced a highly statistically significant increase in overall survival when compared to controls (p ⁇ 0.0001 ), but the effect size was slightly more moderate than with the higher Ls-TPT concentration and thus, suggestive of a dose/concentration dependent effect.
  • Ls-TPT administered by CED in a rat glioma model using U87MG results in a clear and consistent survival advantage as compared to untreated controls.
  • Example 9 Convection-Enhanced Delivery of Liposomal ⁇ -Conotoxin To Kindled Rats
  • Synthetic ⁇ -CTX-G (27 amino acids; MW, 3037), ⁇ -CTX-M (25 amino acids; MW, 2639), and carbamazepine are obtained from Sigma-Aldrich (St. Louis, Mo.). Each is loaded into liposomes composed of distearoylphosphatidylcholine (DSPC), distearoylphosphatidylglycerol (DSPG), and cholesterol (see, e.g., Example 2.1.2).
  • DSPC distearoylphosphatidylcholine
  • DSPG distearoylphosphatidylglycerol
  • cholesterol see, e.g., Example 2.1.2.
  • a cannula-bipolar stimulating electrode assembly is chronically implanted into each rat such that the electrode tip is placed into the basolateral nucleus of the right amygdala at stereotaxic coordinates (AP: -2.8 mm; ML: 5.0 mm; DV: -8.7 mm) measured from bregma (Paxinos G and Watson C (1998) The rat brain in stereotaxic coordinates, 4th ed. Academic Press, Sydney). Dental
  • each rat is restrained and the infusion cannula is slowly inserted into the brain through the guide cannula
  • the tip of the infusion cannula extends to a depth 0 5 mm above the tips of the stimulating electrode wires and is maintained at the appropriate depth by a plastic stop at the top of the cannula
  • the rat is released and placed in a plastic cylinder for the entire infusion All infusions are performed in conscious and unrestrained animals
  • the brain tissue is allowed to seal around the cannula for a few minutes before initiation of the infusion A progressive infusion rate increment is used
  • the infusion rates used to administer 20 ⁇ L volume per hemisphere are 0 2 ⁇ l_/m ⁇ n for 15 mm, 0 5 ⁇ l_/m ⁇ n for 10 mm, and 0 8 ⁇ L/min for 15 mm Following infusion completion,
  • DM VAN/253729-17775/7468072 1 [0416] After the completion of testing, selected animals are perfused transcardially with 4% paraformaldehyde and the brains are removed for sectioning and cresyl violet and silver staining to assess cannula placement and evidence of neuronal damage The effects of each drug treatment are expressed as a change (in percent) from baseline calculated using the following formula 100 X[(value before treatment)-(value after treatment)]/(value before treatment) Treatment effects with respect to baseline for each rat are calculated separately and then averaged for a group Statistical analyses of the data from the kindling and locomotor-activity testing are performed by one-way (within a group) and two-way (between groups) repeated measures analysis of variance (ANOVA) after transformation of the percentage change data using arcsine-root transformation When appropriate, post hoc analysis is performed using Dunnett's test or Tukey's test Tremor data are expressed as frequencies analyzed by the Fisher's exact probability test

Landscapes

  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biomedical Technology (AREA)
  • Engineering & Computer Science (AREA)
  • Dispersion Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Dermatology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Pain & Pain Management (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

L'invention porte sur une administration améliorée par convection (CED) qui est utilisée en tant que procédé d'administration d'une infusion directe d'agents thérapeutiques au système nerveux central, contournant ainsi la barrière hémato-encéphalique. On utilise une composition liposomale non PEGylée comprenant au moins un phospholipide neutre saturé et au moins un phospholipide anionique saturé et un agent thérapeutique ou de diagnostic encapsulé dans celui-ci pour surmonter la toxicité associée à une concentration de médicament à pic élevé administrée localement (CED) ainsi que pour augmenter le volume d'administration au tissu pour une libération de médicament entretenue améliorée. Dans un mode de réalisation, la composition du liposome comprend un rapport molaire de DSPC:DSPG:CHOL de 7:2:1 et l'agent thérapeutique ou de diagnostic est choisi parmi le topotécane, la conotoxine, le gadodiamide ou la rhodamine, et on l'utilise dans le traitement de l'épilepsie.
PCT/CA2009/001708 2008-11-21 2009-11-23 Composition liposomale pour administration améliorée par convection vers le système nerveux central WO2010057317A1 (fr)

Priority Applications (7)

Application Number Priority Date Filing Date Title
AU2009317837A AU2009317837B2 (en) 2008-11-21 2009-11-23 Liposomal composition for convection-enhanced delivery to the central nervous centre
CN200980151445.7A CN102256596B (zh) 2008-11-21 2009-11-23 用于增强对流递送到中枢神经中心的脂质体组合物
BRPI0922100A BRPI0922100A2 (pt) 2008-11-21 2009-11-23 veiculo de liberação lipossômica não peguilado melhorado, composição terapêutica, método melhorado para tratar um distúrbio do sistema nervoso central, e, composição de diagnóstico.
JP2011536717A JP5795537B2 (ja) 2008-11-21 2009-11-23 中枢神経系への対流増進送達のためのリポソーム組成物
US13/130,525 US9295735B2 (en) 2008-11-21 2009-11-23 Liposomal composition for convection-enhanced delivery to the central nervous centre
EP09827097.8A EP2370058B1 (fr) 2008-11-21 2009-11-23 Composition liposomale pour administration améliorée par convection vers le système nerveux central
CA2743959A CA2743959C (fr) 2008-11-21 2009-11-23 Composition liposomale pour administration amelioree par convection vers le systeme nerveux central

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US11707608P 2008-11-21 2008-11-21
US61/117,076 2008-11-21

Publications (1)

Publication Number Publication Date
WO2010057317A1 true WO2010057317A1 (fr) 2010-05-27

Family

ID=42197794

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2009/001708 WO2010057317A1 (fr) 2008-11-21 2009-11-23 Composition liposomale pour administration améliorée par convection vers le système nerveux central

Country Status (9)

Country Link
US (1) US9295735B2 (fr)
EP (1) EP2370058B1 (fr)
JP (1) JP5795537B2 (fr)
CN (1) CN102256596B (fr)
AR (1) AR076634A1 (fr)
AU (1) AU2009317837B2 (fr)
BR (1) BRPI0922100A2 (fr)
CA (1) CA2743959C (fr)
WO (1) WO2010057317A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013135727A1 (fr) * 2012-03-12 2013-09-19 Renishaw Plc Traitement du gliome par un système de délivrance par convection (ced)
WO2019222441A1 (fr) 2018-05-16 2019-11-21 Voyager Therapeutics, Inc. Sérotypes de vaa pour l'administration de charge utile spécifique au cerveau
EP3632923A1 (fr) 2015-01-16 2020-04-08 Voyager Therapeutics, Inc. Polynucléotides de ciblage du système nerveux central

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104736201A (zh) * 2012-10-19 2015-06-24 加利福尼亚大学董事会 治疗中枢神经系统的肿瘤
US9993427B2 (en) 2013-03-14 2018-06-12 Biorest Ltd. Liposome formulation and manufacture
US20170049701A1 (en) * 2015-08-05 2017-02-23 Steven M. Kushner Clustoidal multilamellar soy lecithin phospholipid structures for transdermal, transmucosal, or oral delivery, improved intestinal absorption, and improved bioavailability of nutrients
JP6606600B2 (ja) 2016-04-15 2019-11-13 富士フイルム株式会社 マイクロニードルアレイ
BR112019020406B1 (pt) * 2017-03-31 2021-10-26 Fujifilm Corporation Composições lipossômicas, composição farmacêutica que compreende a dita composição lipossômica e uso da mesma para tratar câncer
US10722465B1 (en) * 2017-12-08 2020-07-28 Quicksilber Scientific, Inc. Transparent colloidal vitamin supplement
US11291702B1 (en) 2019-04-15 2022-04-05 Quicksilver Scientific, Inc. Liver activation nanoemulsion, solid binding composition, and toxin excretion enhancement method
US20240024220A1 (en) * 2019-05-11 2024-01-25 Youngsuk Yi Neurotoxin compositions and methods
CN114469863B (zh) * 2021-11-26 2023-09-26 南方医科大学南方医院 甾醇脂质体作为牙髓和牙本质药物传递系统的应用

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5077056A (en) 1984-08-08 1991-12-31 The Liposome Company, Inc. Encapsulation of antineoplastic agents in liposomes
US5171578A (en) 1985-06-26 1992-12-15 The Liposome Company, Inc. Composition for targeting, storing and loading of liposomes
US5192549A (en) 1988-09-28 1993-03-09 Yissum Research Development Company Of The Hebrew University Of Jerusalem Method of amphiphatic drug loading in liposomes by pH gradient
EP1385480A1 (fr) * 2001-05-10 2004-02-04 Amersham Health AS Liposomes
WO2004087115A2 (fr) * 2003-04-02 2004-10-14 Celator Pharmaceuticals, Inc. Compositions combinees de camptothecines et de fluoropyrimidines
WO2008100930A2 (fr) * 2007-02-13 2008-08-21 Cornell University Appareil, procédé et application d'administration améliorée par convection

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4925664A (en) * 1986-10-20 1990-05-15 University Of Utah Spider toxins and methods for their use as blockers of calcium channels and amino acid receptor function
DE69920425T2 (de) * 1998-04-09 2005-09-29 Amersham Health As Verwendung von teilchenförmigen kontrastmitteln in der diagnostischen bilderzeugung zur untersuchung physiologischer parameter
JP4555569B2 (ja) * 2001-11-13 2010-10-06 セレーター ファーマシューティカルズ, インコーポレイテッド 増強された血中安定性を有する脂質キャリア組成物
CN1391891A (zh) * 2002-04-22 2003-01-22 江苏佩沃特生物基因工程有限公司 紫杉醇隐形脂质体
WO2005079185A2 (fr) * 2003-09-02 2005-09-01 Board Of Regents, The University Of Texas System Composes encapsules dans des liposomes neutres et leurs procedes de fabrication
US8658203B2 (en) * 2004-05-03 2014-02-25 Merrimack Pharmaceuticals, Inc. Liposomes useful for drug delivery to the brain
CN100356919C (zh) * 2004-05-31 2007-12-26 上海医药工业研究院 羟基喜树碱脂质体及其制备方法
WO2006127962A2 (fr) * 2005-05-25 2006-11-30 Becton, Dickinson And Comapny Formulations particulaires pour une administration intradermique d'agents biologiquement actifs
WO2006126208A2 (fr) * 2005-05-26 2006-11-30 Yissum Research Development Company Of The Hebrew University Of Jerusalem Compositions et methodes d'utilisation desdites compositions dans l'administration d'agents dans un organe cible protege par une barriere sanguine
CN1823733A (zh) * 2006-01-06 2006-08-30 中国药科大学 一种含喜树碱类药物的自组装前体脂质体及其制备方法
WO2007106549A2 (fr) * 2006-03-15 2007-09-20 Alza Corporation Procédé de traitement de cancer du poumon
WO2007127839A2 (fr) * 2006-04-26 2007-11-08 The Regents Of The University Of California Compositions et méthodes pour l'administration de neurothérapeutiques de haut poids moléculaire améliorée par convection
WO2009023601A2 (fr) * 2007-08-11 2009-02-19 Argenis Llc Appareil et procédés pour traiter l'épilepsie moyennant une délivrance améliorée par convection

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5077056A (en) 1984-08-08 1991-12-31 The Liposome Company, Inc. Encapsulation of antineoplastic agents in liposomes
US5171578A (en) 1985-06-26 1992-12-15 The Liposome Company, Inc. Composition for targeting, storing and loading of liposomes
US5192549A (en) 1988-09-28 1993-03-09 Yissum Research Development Company Of The Hebrew University Of Jerusalem Method of amphiphatic drug loading in liposomes by pH gradient
EP1385480A1 (fr) * 2001-05-10 2004-02-04 Amersham Health AS Liposomes
WO2004087115A2 (fr) * 2003-04-02 2004-10-14 Celator Pharmaceuticals, Inc. Compositions combinees de camptothecines et de fluoropyrimidines
WO2008100930A2 (fr) * 2007-02-13 2008-08-21 Cornell University Appareil, procédé et application d'administration améliorée par convection

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
BOBO, R. H. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 91, 1994, pages 2076 - 80
CHEN, M.Y. ET AL., J. NEUROSURG., vol. 90, 1999, pages 315 - 20
DICHTER M.: "Basic mechanisms of epilepsy: targets for therapeutic intervention", EPILEPSIA, vol. 38, no. 9, 1997, pages 2 - 6
GRAHN, A. Y. ET AL.: "Non-PEGylated Liposomes for Convection-enhanced Delivevery of Topotecan and Gadodiamide in Malignant Glioma: Initial Experience", JOURNAL OF NEMOONCOLOY, vol. 95, 24 May 2009 (2009-05-24), pages 185 - 197 *
ISHIDA; KIWADA, INT J PHARM., vol. 354, 2008, pages 56 - 62
MCNAMARA J.: "The neurobiological basis of epilepsy", TRENDS NEUROSCI, vol. 15, no. 10, October 1992 (1992-10-01), pages 357 - 9, XP024346793, DOI: doi:10.1016/0166-2236(92)90178-B
MOGHIMI ET AL., FASEB J., vol. 20, 25 October 2006 (2006-10-25), pages 2591 - 3
PRINCE D. A. ET AL.: "Jasper's Basic Mechanisms of the Epilepsies", vol. 79, 1999, article "Epileptogenic neurons and circuits", pages: 665 - 684
SHORVON, S: "Epidemiology, classification, natural history, and genetics of epilepsy", LANCET, vol. 336, no. 8707, 14 July 1990 (1990-07-14), pages 93 - 6
SZEBENI ET AL., J. LIPOSOME RES., vol. 17, 2007, pages 107 - 117

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013135727A1 (fr) * 2012-03-12 2013-09-19 Renishaw Plc Traitement du gliome par un système de délivrance par convection (ced)
EP3632923A1 (fr) 2015-01-16 2020-04-08 Voyager Therapeutics, Inc. Polynucléotides de ciblage du système nerveux central
WO2019222441A1 (fr) 2018-05-16 2019-11-21 Voyager Therapeutics, Inc. Sérotypes de vaa pour l'administration de charge utile spécifique au cerveau

Also Published As

Publication number Publication date
EP2370058B1 (fr) 2017-10-18
EP2370058A4 (fr) 2013-11-20
BRPI0922100A2 (pt) 2016-02-10
US9295735B2 (en) 2016-03-29
AR076634A1 (es) 2011-06-29
AU2009317837A1 (en) 2011-06-30
CA2743959C (fr) 2017-09-26
US20110274625A1 (en) 2011-11-10
CA2743959A1 (fr) 2010-05-27
EP2370058A1 (fr) 2011-10-05
CN102256596A (zh) 2011-11-23
JP2012509284A (ja) 2012-04-19
AU2009317837B2 (en) 2016-02-25
CN102256596B (zh) 2014-10-22
JP5795537B2 (ja) 2015-10-14

Similar Documents

Publication Publication Date Title
AU2009317837B2 (en) Liposomal composition for convection-enhanced delivery to the central nervous centre
Krauze et al. Convection-enhanced delivery of nanoliposomal CPT-11 (irinotecan) and PEGylated liposomal doxorubicin (Doxil) in rodent intracranial brain tumor xenografts
Noble et al. Novel nanoliposomal CPT-11 infused by convection-enhanced delivery in intracranial tumors: pharmacology and efficacy
Yang et al. Focused ultrasound and interleukin-4 receptor-targeted liposomal doxorubicin for enhanced targeted drug delivery and antitumor effect in glioblastoma multiforme
Saito et al. Tissue affinity of the infusate affects the distribution volume during convection-enhanced delivery into rodent brains: implications for local drug delivery
US9655848B2 (en) Liposomes for in-vivo delivery
JP6941606B2 (ja) 安定化カンプトテシン医薬組成物
Inoue et al. Therapeutic efficacy of a polymeric micellar doxorubicin infused by convection-enhanced delivery against intracranial 9L brain tumor models
US20130052259A1 (en) Liposomes comprising amphipathic drugs and method for their preparation
CN103479578B (zh) 一种马来酸匹杉琼的脂质体制剂及其制备工艺
US20110190623A1 (en) Thermally-activatable liposome compositions and methods for imaging, diagnosis and therapy
Grahn et al. Non-PEGylated liposomes for convection-enhanced delivery of topotecan and gadodiamide in malignant glioma: initial experience
Nakamura et al. Local convection-enhanced delivery of chemotherapeutic agent transiently opens blood–brain barrier and improves efficacy of systemic chemotherapy in intracranial xenograft tumor model
US20050232984A1 (en) Non-vesicular cationic lipid formulations
US20110002851A1 (en) Cationic Colloidal Carriers for Delivery of Active Agents to the Blood-Brain Barrier in the Course of Neuroinflammatory Diseases
WO2012125486A1 (fr) Polychimiothérapie pour le traitement du cancer
Sugiyama et al. Safety and efficacy of convection-enhanced delivery of ACNU, a hydrophilic nitrosourea, in intracranial brain tumor models
EP3193871B1 (fr) Délivrance améliorée par convection de micelles chargées en sn-38 contre une tumeur cérébrale
US20220118116A1 (en) Adhesive/adsorption switch on nanoparticles to increase tumor uptake and delay tumor clearance
US20220071934A1 (en) Pharmaceutical compositions for use in treating pain
Sun Liposome-Based Delivery Systems for Use in Biomedical Applications

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200980151445.7

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09827097

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2743959

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2011536717

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2009827097

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2009827097

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2009317837

Country of ref document: AU

Date of ref document: 20091123

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 13130525

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0922100

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20110520