WO2010091192A2 - Nanoscale platinum compounds and methods of use thereof - Google Patents

Nanoscale platinum compounds and methods of use thereof Download PDF

Info

Publication number
WO2010091192A2
WO2010091192A2 PCT/US2010/023217 US2010023217W WO2010091192A2 WO 2010091192 A2 WO2010091192 A2 WO 2010091192A2 US 2010023217 W US2010023217 W US 2010023217W WO 2010091192 A2 WO2010091192 A2 WO 2010091192A2
Authority
WO
WIPO (PCT)
Prior art keywords
nanoparticle
cisplatin
backbone
sidechains
polymer
Prior art date
Application number
PCT/US2010/023217
Other languages
French (fr)
Other versions
WO2010091192A3 (en
Inventor
Shiladitya Sengupta
Shivani Soni
Abhimanyu Paraskar
Original Assignee
The Brigham And Women's Hospital, 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 The Brigham And Women's Hospital, Inc. filed Critical The Brigham And Women's Hospital, Inc.
Priority to JP2011549262A priority Critical patent/JP5844642B2/en
Priority to US13/147,751 priority patent/US9393227B2/en
Priority to EP10739130.2A priority patent/EP2393854B1/en
Priority to RU2011134316/04A priority patent/RU2538199C2/en
Priority to BRPI1008869A priority patent/BRPI1008869A2/en
Priority to CN201080015384.4A priority patent/CN102388073B/en
Priority to AU2010210593A priority patent/AU2010210593B2/en
Publication of WO2010091192A2 publication Critical patent/WO2010091192A2/en
Publication of WO2010091192A3 publication Critical patent/WO2010091192A3/en
Priority to HK12108633.2A priority patent/HK1167869A1/en
Priority to US15/182,967 priority patent/US10512696B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/58Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. poly[meth]acrylate, polyacrylamide, polystyrene, polyvinylpyrrolidone, polyvinylalcohol or polystyrene sulfonic acid resin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/282Platinum compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/382Heterocyclic compounds having sulfur as a ring hetero atom having six-membered rings, e.g. thioxanthenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/24Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing atoms other than carbon, hydrogen, oxygen, halogen, nitrogen or sulfur, e.g. cyclomethicone or phospholipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • A61K47/544Phospholipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • A61K47/6931Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer
    • A61K47/6933Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer the polymer being obtained by reactions only involving carbon to carbon, e.g. poly(meth)acrylate, polystyrene, polyvinylpyrrolidone or polyvinylalcohol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J41/00Normal steroids containing one or more nitrogen atoms not belonging to a hetero ring
    • C07J41/0033Normal steroids containing one or more nitrogen atoms not belonging to a hetero ring not covered by C07J41/0005
    • C07J41/0055Normal steroids containing one or more nitrogen atoms not belonging to a hetero ring not covered by C07J41/0005 the 17-beta position being substituted by an uninterrupted chain of at least three carbon atoms which may or may not be branched, e.g. cholane or cholestane derivatives, optionally cyclised, e.g. 17-beta-phenyl or 17-beta-furyl derivatives
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08FMACROMOLECULAR COMPOUNDS OBTAINED BY REACTIONS ONLY INVOLVING CARBON-TO-CARBON UNSATURATED BONDS
    • C08F22/00Homopolymers and copolymers of compounds having one or more unsaturated aliphatic radicals each having only one carbon-to-carbon double bond, and at least one being terminated by a carboxyl radical and containing at least one other carboxyl radical in the molecule; Salts, anhydrides, esters, amides, imides or nitriles thereof
    • C08F22/04Anhydrides, e.g. cyclic anhydrides
    • C08F22/06Maleic anhydride
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08FMACROMOLECULAR COMPOUNDS OBTAINED BY REACTIONS ONLY INVOLVING CARBON-TO-CARBON UNSATURATED BONDS
    • C08F222/00Copolymers of compounds having one or more unsaturated aliphatic radicals, each having only one carbon-to-carbon double bond, and at least one being terminated by a carboxyl radical and containing at least one other carboxyl radical in the molecule; Salts, anhydrides, esters, amides, imides, or nitriles thereof
    • C08F222/04Anhydrides, e.g. cyclic anhydrides
    • C08F222/06Maleic anhydride
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08FMACROMOLECULAR COMPOUNDS OBTAINED BY REACTIONS ONLY INVOLVING CARBON-TO-CARBON UNSATURATED BONDS
    • C08F8/00Chemical modification by after-treatment
    • C08F8/30Introducing nitrogen atoms or nitrogen-containing groups
    • C08F8/32Introducing nitrogen atoms or nitrogen-containing groups by reaction with amines

Definitions

  • This invention relates to biocompatible conjugated polymer nanoparticles including a copolymer backbone, a plurality of sidechains covalently linked to the backbone, and, a plurality of platinum compounds dissociably linked to the backbone.
  • Cancer is the second leading cause of mortality in the United States, with an estimated 1,444,180 new cases and 565,650 deaths in 2008. Cytotoxic agents, which are used in standard chemotherapy, non- specifically target all dividing cells resulting in dose- limiting toxicities. There is an urgent need to develop novel strategies that are more specifically targeted against the tumor.
  • nanovectors holds the potential to revolutionize cancer chemotherapy by specifically targeting the tumor.
  • a number of polymeric nanovectors are currently in development or in clinics, and are dramatically altering the pharmacodynamic and pharmacokinetic profile of the active agent.
  • most of these polymeric constructs decrease the potency of the conjugated active agent, relying on increased uptake into the tumor for the improved therapeutic index.
  • Cisplatin is one of the mainstays in chemotherapy regimes for most types of
  • the invention is directed to a biocompatible conjugated polymer nanoparticle including a copolymer backbone, a plurality of sidechains covalently linked to said backbone, and, a plurality of platinum compounds dissociably linked to said backbone.
  • the platinum compound is dissociably linked to backbone via linkage through the sidechain.
  • the platinum compound is linked to the sidechain through at least one coordination bond.
  • Another aspect of the invention is directed to biocompatible conjugated polymer nanoparticles including a polymaleic acid (PMA) such as a poly(isobutylene- ⁇ /£- maleic acid) (PIMA) backbone.
  • the backbone consists of from 25 to 50 monomers.
  • a plurality of PEG sidechains covalently linked to said backbone have a molecular weight of from 200 to 3000 Dalton.
  • the PEG sidechains number between 50% and 100%, inclusive, of the number of monomeric units of the polymer backbone.
  • cisplatin or oxaliplatin side groups dissociably linked to the backbone.
  • the cisplatin side groups number between 25% and 75%, inclusive, of the number of monomeric units of the polymer backbone.
  • Yet another aspect of the invention is directed to biocompatible conjugated polymer nanoparticles including a poly(isobutylene- ⁇ /Mnaleic acid) backbone.
  • the backbone consist of about 40 monomers.
  • a plurality of PEG sidechains covalently linked to the backbone The PEG sidechains have a molecular weight of about 2000 Dalton.
  • the PEG sidechains number greater than 90% of monomeric units of said polymer backbone.
  • a plurality of cisplatin or oxaliplatin side groups dissociably linked to the backbone.
  • the cisplatin or oxaliplatin side groups number between 25% and 75%, inclusive, of the number of monomeric units of the polymer backbone.
  • Still another aspect of the invention is directed to biocompatible conjugated polymer nanoparticles including a poly(isobutylene- ⁇ /Mnaleic acid) backbone.
  • the backbone consists of from 25 to 50 monomers.
  • a plurality of glucosamine sidechains covalently linked to said backbone The glucosamine sidechains number between 50% and 100%, inclusive, of monomeric units of said polymer backbone.
  • a plurality of cisplatin or oxaliplatin side groups dissociably linked to the backbone.
  • the cisplatin or oxaliplatin side groups number between 25% and 75%, inclusive, of the number of monomeric units of the polymer backbone.
  • Another aspect of the invention is directed to biocompatible conjugated polymer nanoparticles including a poly(isobutylene- ⁇ /Mnaleic acid) backbone.
  • the backbone consists of from 25 to 50 monomers.
  • a plurality of glucosamine sidechains covalently linked to said backbone The glucosamine sidechains number greater than 75% of monomeric units of said polymer backbone.
  • a plurality of cisplatin or oxaliplatin side groups dissociably linked to the backbone.
  • the cisplatin or oxaliplatin side groups number between 25% and 75%, inclusive, of the number of monomeric units of the polymer backbone.
  • Yet another aspect of the invention is directed to carboxylic acid-platinum II
  • (Pt(II)) complex conjugated nanoparticles including a carboxylic acid-(Pt(II)) complex and a plurality of lipid-polymer chains.
  • the carboxylic acid portion of said carboxylic acid- cisplatin/oxaliplatin complex is covalently bound to said lipid-polymer chains.
  • Another aspect of the invention is directed to a vesicle, micelle, or liposome compound comprising a plurality of nanoparticles of claim as described herein.
  • Still another aspect of the invention is directed to pharmaceutical compositions including any of the nanoparticles or compounds described herein and a pharmaceutically acceptable carrier.
  • Yet another aspect of the invention is directed to a method of treating cancer or metastasis.
  • the method includes selecting a subject in need of treatment for cancer or metastasis and administering to the subject an effective amount of any of the nanoparticles, compounds, or compositions described herein.
  • Figure 1 shows a schematic of synthesis of PMA-Cisplatin. Loading of different numbers of cisplatin per polymer affects the size of the nanoparticles as measured using DLS or TEM. And a graph of Cytotoxicity study of Polyisobutylene maleic acid carrier
  • Figure 2 shows a scheme showing derivatization of PMA with EDA.
  • the derivatized polymer was used to synthesize the cisplatin-complex.
  • the graph shows the effect of different treatments on LLC viability following 48 hours of incubation.
  • Figure 3 shows a scheme of the synthesis of PMA-GA-Cisplatin.
  • FIG. 4 The graph on the left shows the amount of active cisplatin that is released from the PMA-GA-Cisplatin nanoparticle when incubated with LLC lysate.
  • the concentration-effect graph of the right shows the effect of different treatments on the viability of Lewis Lung Carcinoma cells when incubated with the active agents for 48 hours. Cell viability was measured using an MTS assay.
  • Figures 5A-5E are line graphs (Figs.5A and 5B) and bar graphs (Figs. 5C-5E) showing efficacy and toxicity profile of free and nanoparticle cisplatin in Lewis Lung carcinoma model.
  • Tumors were induced by injecting LLC cells in c57/BL6 mouse.
  • the effect of treatments on tumor volume (Fig, 15A) and body weight (Fig. 5B) over the treatment period is shown.
  • the effect of treatment on the organ weight of kidneys Fig.
  • the images on top of each graph show representative organs from each treatment group.
  • Fig. 5E shows the biodistribution of Pt in kidney and tumor as measured using ICP-spectroscopy 24 h hours following administration of free or cisplatin nanoparticles
  • Figure 6 is a scheme showing synthesis of PMA-GA-Cisplatin (8).
  • Figure 7 Effect of PMA-PEG-Cisplatin on Lewis Lung Carcinoma. Cell were incubated for 48 hours with the drugs or vehicles and then tested for viability using an MTS assay.
  • Figure 8 Effect of different treatments on tumor growth and body weight in vivo. Tumors were induced by injecting LLC cells in c57/BL6 mouse.
  • Figure 9 is a graph of the amount of platination of the polymer was quantified using a uv-vis spectroscopy method.
  • FIG. 10 Scheme showing synthesis of lipid maleic acid-cipslatin complex, which can form micelles in water.
  • FIG. HA and HB aree schematics showing SAR-inspired engineering of a cisplatin nanoparticle.
  • Fig. HA shows the mechanism underlying the intracellular activation of cisplatin through aquation. The leaving groups of cisplatin and analogues (shown with blue lines) are replaced with OH prior to DNA-binding.
  • FIG. 12A shows that derivatization of all the monomeric units of PDVIA with glucosamine and subsequent complexation with Pt forms nanoparticles smaller than 150 nm.
  • Fig. 12B shows the total platinum loaded per mg of polymer at this ratio.
  • Figures 13A-13E are line graphs showing in vitro characterization of cisplatin nanoparticles.
  • Figs. 13A and 13B show the concentration-effect of different treatments on cellular viability as measured using MTS assay.
  • X-axis shows the equivalent concentrations of cisplatin.
  • dose of polymer used was equivalent to that used to deliver that specific dose of cisplatin in the complexed form.
  • PDVIA was also derivatized with ethylene diamine to generate PDVIA-EDA, which offers a similar complexation environment to platinum as PDVIA-GA. Unlike PIMA-GA, PIMA-EDA exerted inherent toxicity.
  • PIMA-GA-Cisplatinfacidic refers to the isomer formed under acidic complexation environment while PDVIA-GA-Cisplatin[basic] refers to the isomer formed under alkaline environment.
  • Figs. 13C-13E show effect of PIMA-GA-cisplatin nanoparticles on LLC cells viability, when the PDVIA backbone (40 monomeric units) is derivatized to different degrees.
  • PIMA-30GA-Cisplatin has 30 of the 40 monomeric units derivatized with glucosamine while PIMA-GA-40 and PIMA-GA-200 have all the monomeric units derivatized.
  • [a] and [b] refers to isomers formed in acidic and basic environments when the polymers are complexed with cisplatin.
  • Table 1 shows the corresponding IC50 values.
  • Figures 14A-14J show FACS images (Figs. 14A- 14H) and bar graphs (Figs.
  • Figurel5 is a schematic showing the labeling of the PEVIA-GA polymer with
  • FIG. 16 is a line graph showing the effect of pH and Pt complexation environment on release kinetics.
  • the nanoparticles were incubated at pH 5.5 or pH8.5 in a dialysis bag, and release over time was quantified.
  • the nanoparticles [PIMAGA-Cisplatin (0->Pt)] used were generated by complexing the polymer and cisplatin in an acidic pH [6.4] unless in the case of PIMA-GA-Cisplatin (Pt->N), where the complexation was carried out in a basic pH to generate the stable isomer [PEVIA-GA-Cisplatin (N->Pt)].
  • Figures 17A-17D are line graphs (Figs. 17A and 17B) and bar graphs (Figs.
  • FIG. 17C and 17D showing PIMA-GA-cisplatin nanoparticle exerts similar anti-tumor effect with reduced systemic toxicity compared to free cisplatin in a 4Tl breast cancer model.
  • Figures 18A and 18B are a bar graph (Fig. 18A) and a line graph (Fig. 18B) showing PIMA-GA-cisplatin nanoparticle inhibits tumor growth in a K-ras LSI7+ /Pten fl/fl ovarian cancer model.
  • bioluminescence quantification indicated a significantly decreased tumor luciferase signal in mice treated with cisplatin-NP compared to vehicle (p ⁇ 0.05, one-way ANOVA analysis).
  • Fig. 18B shows drug toxicity assessed by measurements in overall body weight.
  • FIGS. 19A and 19B are bar graphs showing the distribution of Pt following administration of cisplatin, cisplatin-nanoparticle ([PIMA-GA-Cisplatin (0->Pt)] or carboplatin in breast cancer or ovarian cancer. Treatment was administered as described in Figs. 17 and 18. The level of Pt in different tissues harvested following necropsy was quantified using inductively coupled plasma- spectrometry (ICP).
  • ICP inductively coupled plasma- spectrometry
  • Figures 2OA and 2OB are schematic showing SAR- inspired engineering of a oxaliplatin nanoparticle.
  • Figure 2OA shows the mechanism underlying the intracellular activation of oxaliplatin through aquation.
  • Figure 2OB shows the chemical synthesis of PIMA-oxaliplatin and PIMA-glucosamine (PIMA-GA)-oxaliplatin complex.
  • Oxaliplatin-OH can be complexed with PIMA through dicarboxylato bonds. Derivatization of one arm of PIMA with glucosamine, and complexation with oxaliplatin can lead to two isomers depending on pH.
  • Figures 21 A and 21B are line graphs showing the concentration-effect of different treatments on cellular viability as measured using MTS assay.
  • Breast cancer cell line, Lewis lung cancer (Fig. 21A) and 4Tl (Fig. 21B) cell lines were used for this study.
  • X- axis shows the equivalent concentrations of platinum. Where blank polymeric controls were used, dose of polymer used was equivalent to that used to deliver that specific dose of oxaliplatin in the complexed form.
  • PIMA-GA-Ox refers to the isomer [PIMA-GA- Oxaliplatin (0->Pt)] formed under acidic complexation environment. The PIMA-GA- oxaliplatin curve shifted to the left, indicating that the nanoparticles are more effective than free oxaliplatin in anti-tumor efficacy.
  • Figures 22A-22E are line graphs (Figs. 26A and 26B) and bargraphs (Figs.
  • Figure 23 is a line graph showing concentration-effect of different oxaliplatin complexes on cellular viability as measured using MTS assay.
  • Figure 24 is a line graph showing the effect of cisplatin, carboplatin and
  • Figure 25A is a scheme showing the synthesis of cholesterol- succinic acid conjugate and the complexation of Pt to the conjugate.
  • Figure 25B shows a dynamic laser light scatter of liponanoparticles.
  • the size of the liponanoparticles is less than 150 nm.
  • Figure 26 is a line graph showing the release kinetics of Pt from the liponanoparticle with time and the influence of pH. The rate of release is faster in an acidic pH, which facilitates selective release of active platinate in the tumor, consistent with the acidic intratumoral pH.
  • Figures 27A-27C are line graphs showing the effect of cisplatin- liponanoparticle on viability of 4Tl breast cancer cells. Cell viability was measured using
  • Figures 28A and 28B are line graphs showing the effect of cisplatin- liponanoparticle on viability of a cisplatin-resistant hepatocellular cancer cell line (CP20) and on a Lewis lung cancer cell line (LLC). Cisplatin acts on CP20 only at the highest concentration, while the cells are susceptible to the cisplatin-liponanoparticle (Fig. 28A).
  • Cisplatinliponanoparticle exerted superior anticancer effect than free cisplatin on LLCs (Fig.
  • Figures 29A-29E are line graphs (Figs. 29A and 29B) and bar graphs (Figs.
  • the invention is directed to biocompatible conjugated polymer nanoparticles including a copolymer backbone, a plurality of sidechains covalently linked to said backbone, and, a plurality of platinum compounds dissociably linked to said backbone.
  • the platinum compounds are linked to the backbone via a linkage to the sidechains.
  • the copolymer comprises maleic acid monomers.
  • the copolymer is poly(isobutylene- ⁇ /Mnaleic acid)
  • the copolymer comprises from 2 to 100 monomeric units. In some embodiments, the copolymer comprises from 25 to 50 monomeric uints.
  • the sidechains are selected from the group consisting of polymers, monosaccharides, carboxylic acids, dicarboxylic acids, amides, and combinations thereof.
  • the sidechains are polyethylene glycol (PEG).
  • PEG sidechains can be represented by -C(O)-NH-PEG.
  • the PEG sidechains have a molecular weight of from
  • the PEG sidechains have a molecular weight of from 1000 to 3000 Dalton. In a preferred embodiment, the PEG sidechains have a molecular weight of about 2000 Dalton.
  • the sidechains are monosaccharides.
  • the monosaccharides are glucosamine.
  • the monosaccharide sidechains can be represented by -C(O)-saccharide.
  • platinum compound is a platinum(II) or platinum (IV) compound.
  • the platinum (II) compound is selected from the group consisting of cisplatin, oxaliplatin, carboplatin, paraplatin, sartraplatin, and combinations thereof.
  • the platinum (II) compound side group is cisplatin or oxalipaltin.
  • the platinum(II) compound is selected from the group consisting of:
  • the platinum (II) compound is Pt(NHs) 2 .
  • the platinum (II) compound is " p , wherein p is
  • the platinum (II) compound comprises at least two nitrogen atoms, where said nitrogen atoms are directly linked to platinum.
  • the two nitrogen atoms are linked to each other via an optionally substituted linker, e.g. acyclic or cyclic linker.
  • a cyclic linker means a linking moiety that comprises at least one ring structure. Cyclic linkers can be aryl, heteroaryl, cyclyl or heterocyclyl.
  • At least one nitrogen that is linked to platinum is a ring atom of a heteroaryl or a heterocyclyl.
  • heteroaryl is optionally substituted pyridine, e.g., 2-methylpyridine.
  • the plurality of sidechains corresponds to a number between 50% and 100%, inclusive, of the number of monomeric units of said polymer backbone. This means that between 50% to 100% of the monomeric units have at least one sidechain linked to the monomeric unit.
  • the total number of sidechains can be greater than the total number of the monomeric units. For example, two sidechains can be attached to a maleic acid monomer.
  • the plurality of sidechains corresponds to a number greater than 90% of the number of monomeric units of said polymer backbone.
  • the plurality of platinum compounds corresponds to number between 10% and 100%, inclusive, of the number of monomeric units of said polymer backbone. Generally there is a one-to-one relationship between platinum compounds and monomeric subunits. Thus, the percent refers to the number of monomeric units that are linked to a platinum compound to the total number of monomeric units present in the polymer backbone.
  • the plurality of platinum compounds corresponds to number between 25% and 75%, inclusive, of the number of monomeric units of said polymer backbone.
  • the sidechains comprise dicarboxylic acids.
  • the dicarboxylic acids are of the formula HOOC-R-COOH, wherein R is a Cl- C6 alkyl, C2-C6 alkenyl, or C2-C6 alkynyl.
  • the dicarboxylic acid is maleic acid.
  • the copolymer comprises at least one monomer having the formula -CH(CO 2 H)-R-CH(C(O)R')- , wherein R is a bond, Ci-C 6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
  • R is a bond.
  • between 50% to 100%, inclusive, of the monomeric subunits in the polymeric backbone are -CH(CO 2 H)-R-CH(C(O)R')-, wherein R is a bond, Ci-C 6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
  • At least 90% or more of the monomeric subunits in the polymeric backbone are -CH(CO 2 H)-R-CH(C(O)R')-, wherein R is a bond, Ci-C 6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
  • the copolymer comprises at least one monomer having the formula -CH(CO 2 H)-R-CH(C(O)R' )CH 2 C(Me 2 )- or -CH(C(O)R' )-R-CH(CO 2 H)- CH 2 C(Me 2 )-, wherein R is a bond, C 1 -C 6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
  • R is a bond.
  • the copolymer comprises between 50% to 100%, inclusive of monomers having the formula -CH(CO 2 H)-R-CH(C(O)R' )CH 2 C(Me 2 )- or - CH(C(O)R')-R-CH(CO 2 H)-CH 2 C(Me 2 )-, wherein R is a bond, C 1 -C 6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
  • the copolymer comprises at least 90% of monomers having the formula -CH(CO 2 H)-R-CH(C(O)R' )CH 2 C(Me 2 )- or -CH(C(O)R' )-R- CH(CO 2 H)-CH 2 C(Me 2 )-, wherein R is a bond, C 1 -C 6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
  • R' is o O r -
  • At least one monomer of the polymer comprises two sidechains selected from the group consisting of carboxylic acid, amide, and ester. Said sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof.
  • said amide and ester sidechains are separated from each other by two carbon atoms.
  • at least one of the sidechains is not a carboxylic acid.
  • At least one monomer of the polymer comprises two carboxylic acid sidechains.
  • Said carboxylic acid sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof.
  • said carboxylic acid sidechains are separated from each other by two carbon atoms.
  • the said carbon atoms can be linked to each other through a single bond or a double bond.
  • At least one monomer of the polymer comprises a carboxylic acid and an amide sidechains.
  • Said carboxylic acid sidechains and amide sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof.
  • said carboxylic acid and amide sidechains are separated from each other by two carbon atoms. The said carbon atoms can be linked to each other through a single bond or a double bond.
  • At least one monomer of the polymer comprises a carboxylic acid and an ester sidechains.
  • Said carboxylic acid sidechains and ester sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof.
  • said carboxylic acid and ester sidechains are separated from each other by two carbon atoms.
  • the said carbon atoms can be linked to each other through a single bond or a double bond.
  • At least one monomer of the polymer comprises an amide and an ester sidechains.
  • Said amide sidechains and ester sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof.
  • said amide and ester sidechains are separated from each other by two carbon atoms.
  • the said carbon atoms can be linked to each other through a single bond or a double bond.
  • At least one monomer of the polymer comprises two amide sidechains. Said amide sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof. Preferably, said amide and ester sidechains are separated from each other by two carbon atoms. [0079] In some embodiments, at least one monomer of the polymer comprises two ester sidechains. Said ester sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7,
  • said amide and ester sidechains are separated from each other by two carbon atoms.
  • the polymer comprises two sidechains selected from the group consisting of carboxylic acid, amide, and ester. Said sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof.
  • said amide and ester sidechains are separated from each other by two carbon atoms.
  • at least least one of the sidechains is not a carboxylic acid.
  • the polymer comprises at least two carboxylic acid sidechains. Said carboxylic acid sidechains being separated from each other by 1, 2, 3, 4, 5,
  • carboxylic acid sidechains are separated from each other by two carbon atoms.
  • the polymer comprises a carboxylic acid and an amide sidechains.
  • Said carboxylic acid sidechains and amide sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof.
  • said carboxylic acid and amide sidechains are separated from each other by two carbon atoms.
  • the polymer comprises a carboxylic acid and an ester sidechains.
  • Said carboxylic acid sidechains and ester sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof.
  • said carboxylic acid and ester sidechains are separated from each other by two carbon atoms.
  • the polymer comprises an amide and an ester sidechains. Said amide sidechains and ester sidechains being separated from each other by
  • said amide and ester sidechains are separated from each other by two carbon atoms.
  • the said carbon atoms can be linked to each other through a single bond or a double bond.
  • the polymer comprises two amide sidechains. Said amide sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof. Preferably, said amide and ester sidechains are separated from each other by two carbon atoms. [0086] In some embodiments, the polymer comprises two ester sidechains. Said ester sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof. Preferably, said amide and ester sidechains are separated from each other by two carbon atoms.
  • the nanoparticles of the invention can range in size from 25-250nm, preferably 50-200nm, more preferably 80-160nm, and most preferably 90-1 IOnm.
  • nanoparticles in the size range of 80-160 home preferentially into tumors resulting from the enhanced permeability and retention. See for example,
  • the platinum compound is dissociably linked to said backbone through at least one coordination bond.
  • the coordination bond is more liable and thus releases the platinum compound more easily.
  • linking of the platinum compound to the biopolymer backbone further comprises a carboxylato bond.
  • the platinum compound is linked to the backbone through a coordination bond and a carboxylato bond.
  • the coordination bond is between platinum atom of the platinum compound and an oxygen of the sidechain.
  • the coordination bond is between platinum and a carbonyl oxygen.
  • the coordination bond is between platinum atom of the platinum compound and an amide oxygen of the sidechain. In some embodiments, the coordination bond is between platinum atom of the platinum compound and an ester carbonyl oxygen of the sidechain.
  • the copolymer comprises at least one maleic acid monomer, wherein at least one carboxylic acid of said at least maleic acid is derivatized to an amide.
  • between 50% to 100%, inclusive, of the monomeric units in the polymer backbone are maleic acid monomer and wherein at least one carboxylic acid of said maleic acid monomer is derivatized to an amide.
  • at least 90% of the monomeric units in the polymer backbone are maleic acid monomer and wherein at least one carboxylic acid of said maleic acid monomer is derivatized to an amide.
  • the loading of platinum compound onto the polymer can be represented by as percent mg of platinum compound per mg of polymer.
  • a maximum of 0.375 mg of cisplatin can be loaded onto the PIMA-GA polymer therefore a loading of 37.5% represents the maximum loading for that particular polymer.
  • the loading can range from about 1% to the theoretical total loading for a polymer.
  • the platinum compound loading is from l%-37.5%.
  • the percent loading represent mg of platinum compound linked with per mg of polymer.
  • the platinum compound loading is from l%-6%.
  • the Pt(II) compound loading is from 0.01% to 1%.
  • Another aspect of the invention is directed to biocompatible conjugated polymer nanoparticles including a poly(isobutylene- ⁇ /Mnaleic acid) backbone.
  • the backbone consists of from 25 to 50 monomers.
  • the PEG sidechains have a molecular weight of from 1000 to 3000 Dalton.
  • the PEG sidechains number between 50% and 100%, inclusive, of the number of monomeric units of the polymer backbone.
  • a plurality of cisplatin side groups dissociably linked to the backbone.
  • the cisplatin side groups number between 25% and 75%, inclusive, of the number of monomeric units of the polymer backbone.
  • Yet another aspect of the invention is directed to biocompatible conjugated polymer nanoparticles including a poly(isobutylene- ⁇ /Mnaleic acid) backbone.
  • the backbone consist of 40 monomers.
  • a plurality of PEG sidechains covalently linked to the backbone The PEG sidechains have a molecular weight of 2000 Dalton.
  • the PEG sidechains number greater than 90% of monomeric units of said polymer backbone.
  • cisplatin side groups dissociably linked to the backbone. The cisplatin side groups number between 25% and 75%, inclusive, of the number of monomeric units of the polymer backbone.
  • Still another aspect of the invention is directed to biocompatible conjugated polymer nanoparticles including a poly(isobutylene- ⁇ /Mnaleic acid) backbone.
  • the backbone consists of from 25 to 50 monomers.
  • a plurality of glucosamine sidechains covalently linked to said backbone The glucosamine sidechains number between 50% and 100%, inclusive, of monomeric units of said polymer backbone.
  • a plurality of cisplatin side groups dissociably linked to the backbone. The cisplatin side groups number between 25% and 75%, inclusive, of the number of monomeric units of the polymer backbone.
  • Another aspect of the invention is directed to biocompatible conjugated polymer nanoparticles including a poly(isobutylene- ⁇ /Mnaleic acid) backbone.
  • the backbone consists of from 25 to 50 monomers.
  • a plurality of glucosamine sidechains covalently linked to said backbone The glucosamine sidechains number greater than 90% of monomeric units of said polymer backbone.
  • a plurality of cisplatin side groups dissociably linked to the backbone. The cisplatin side groups number between 25% and 75%, inclusive, of the number of monomeric units of the polymer backbone.
  • Yet another aspect of the invention is directed to carboxylic acid-platinum compound complex conjugated nanoparticles including a carboxylic acid-platinum compound complex and a plurality of lipid-polymer chains.
  • the carboxylic acid portion of said carboxylic acid-platinum complex is covalently bound to said lipid-polymer chains.
  • the carboxylic acid is maleic acid.
  • the polymer is PEG.
  • the platinum compound loading is from l%-37.5%.
  • the platinum compound loading is from l%-6%.
  • the platinum compound can be Pt(II) compound or a Pt(IV) compound.
  • the Pt(II) compound is selected from the group consisting of cisplatin, oxaliplatin, carboplatin, paraplatin, sartraplatin, and combinations thereof.
  • the Pt(II) compound is cisplatin.
  • Another aspect of the invention is directed to a vesicle, micelle, or liposome compound comprising a plurality of nanoparticles of claim as described herein.
  • Still another aspect of the invention is directed to a pharmaceutical composition including any of the nanoparticles or compounds described herein and a pharmaceutically acceptable carrier.
  • Yet another aspect of the invention is directed to a method of treating cancer or metastasis.
  • the method includes selecting a subject in need of treatment for cancer or metastasis and administering to the subject an effective amount of any of the nanoparticles, compounds, or compositions described herein.
  • the cancer or metastasis is selected from the group consisting of platinum susceptible or resistant tumors including breast, head and neck, ovarian, testicular, pancreatic, oral-esophageal, gastrointestinal, liver, gall bladder, lung, melanoma, skin cancer, sarcomas, blood cancers, brain tumors including glioblastomas, and tumors of neuroectodermal origin.
  • the invention provide for methods of formulating platinum compound polymer nanoparticles, method comprising conjugation of platinum compound with a biocompatible polymer or biocompatible copolymer.
  • conjugation of platinum compound with the biocompatible polymer at acidic pH results in nanoparticles that are more active in vivo than when conjugation is done at basic pH.
  • conjugation is done at pH below 7, preferably a pH between 1 and 6.9. In some more preferred embodiments, conjugation is carried out at a pH of 6.5.
  • Pt(II) compound to the polymer is used. In some embodiments, 5-25 mole access of Pt(II) compound to polymer is used. Preferably 10-20 mole access of platinum(II) compound to polymer is used . In one preferred 15 mole access of Pt(II) compound to polymer is used.
  • the invention provides a dicarbonyl molecule linked to a lipid molecule.
  • a dicarbonyl molecule linked to a lipid molecule.
  • a compound can be represented by the structure lipid-linker- dicarbnoyl.
  • These molecules can be used to complex platinum compounds such as cisplatin, oxaliplatin or other platinates and platinum compounds described herein through carboxylato- linkage and/or coordination bonds. These can then be mixed with appropriate lipids/phospholipids to nanoparticles of less than 150 nm, which release Pt in a pH-dependent manner. Once formulated these nanoparticles exhibit improved efficacy and toxicity profile as compared with carboplatin and cisplatin, and are active in a cisplatin-resistant cancer.
  • nanoparticles can be formulated to comprise pharmaceutically active agents for delivery.
  • lipid is used in the conventional sense to refer to molecules that are soluble to a greater or lesser degree in organic solvents, like alcohols, and relatively insoluble in aqueous media.
  • lipid includes compounds of varying chain length, from as short as about 2 carbon atoms to as long as about 28 carbon atoms. Additionally, the compounds may be saturated or unsaturated, and in the form of straight- or branched-chains or in the form of unfused or fused ring structures.
  • Exemplary lipids include, but are not limited to, fats, waxes, sterols, steroids, bile acids, fat-soluble vitamins (such as A, D, E, and K), monoglycerides, diglycerides, phospholipids, glycolipids, sulpholipids, aminolipids, chromolipids (lipochromes), glycerophospholipids, sphingolipids, prenol lipids, saccharolipids, polyketides, and fatty acids.
  • the lipid is cholesterol or distearoylphosphatidylethanolamine.
  • the dicarbonyl molecule is a dicarboxylic acid, or a keto-carboxylic acid. In some preferred embodiments, the dicarbonyl molecule is succinic acid. [00119] In some embodiments, the dicarbonyl molecule is ROC(O)-R-C(O)-, wherein
  • R is Ci-C 6 alkylene, where the alkylene can comprise one or more double or triple bonds and/or the backbone of the alkylene can be interrupted with one or more of O, S, S(O), SO 2 , NH, C(O); and R' is H, alkyl, alkenyl, alkynyl, aryl, heteroaryl, acylcy, heterocyclyl, each of which can be optionally substituted.
  • R' is H.
  • the dicarbonyl molecule can be linked with the lipid molecule directly or through a linker molecule.
  • linker means an organic moiety that connects two parts of a compound.
  • Linkers typically comprise a direct bond or an atom such as oxygen or sulfur, a unit such as NH, C(O), C(O)NH, SO, SO 2 , SO 2 NH or a chain of atoms, such as substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, heterocyclylalkyl, heterocyclylalkenyl, heterocyclylalkynyl, aryl, heteroaryl, heterocyclyl, cycloalkyl, cycloalkenyl, alkyl,
  • linker is a diamine such as ethylene diamine. In some embodiments, linker is PEG-NH 2 .
  • linker is -NHCH 2 CH 2 C(O)-. In another preferred embodiment, linker is -CH 2 CH 2 NHC(O)-[OCH 2 CH 2 ] Z -NH-, where z is 1-50.
  • z is 45.
  • the lipid-dicarbonyl compound is as shown in Figs. 10
  • the invention provide a biocompatible polymer comprising at least one monomer having the formula -CH(CO 2 H)-R-CH(C(O)R')- , wherein R is a bond,
  • Ci-C 6 alkylene where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
  • R is a bond.
  • the polymer comprises from 2 to 100 monomeric units having the formula -CH(CO 2 H)-R-CH(C(O)R')-, wherein R is a bond, Ci-C 6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
  • the polymer comprises from 25 to 50 monomeric units having the formula -CH(CO 2 H)-R-CH(C(O)R')-, wherein R is a bond, Ci-C 6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
  • between 50% to 100%, inclusive, of the monomeric subunits in the polymeric backbone are -CH(CO 2 H)-R-CH(C(O)R')-, wherein R is a bond,
  • Ci-C 6 alkylene where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
  • At least 90% or more of the monomeric subunits in the polymeric backbone are -CH(CO 2 H)-R-CH(C(O)R')-, wherein R is a bond, Ci-C 6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
  • the copolymer comprises at least one monomer having the formula -CH(CO 2 H)-R-CH(C(O)R' )CH 2 C(Me 2 )- or -CH(C(O)R' )-R-CH(CO 2 H)- CH 2 C(Me 2 )-, wherein R is a bond, Ci-C 6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
  • R is a bond.
  • the copolymer comprises between 50% to 100%, inclusive of monomers having the formula -CH(CO 2 H)-R-CH(C(O)R' )CH 2 C(Me 2 )- or - CH(C(O)R')-R-CH(CO 2 H)-CH 2 C(Me 2 )-, wherein R is a bond, Ci-C 6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
  • the copolymer comprises at least 90% of monomers having the formula -CH(CO 2 H)-R-CH(C(O)R' )CH 2 C(Me 2 )- or -CH(C(O)R' )-R- CH(CO 2 H)-CH 2 C(Me 2 )-, wherein R is a bond, Ci-C 6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
  • R' is " ° or -
  • compositions described herein can be used in methods for sustained release of bioactive active agents.
  • the method comprising: (a) providing or administering to a subject a composition described herein, wherein the composition contains the bioactive agent.
  • bioactive agents refer to naturally occurring biological materials, for example, extracellular matrix materials such as fibronectin, vitronection, and laminin; cytokins; and growth factors and differentiation factors.
  • Bioactive agents also refer to artificially synthesized materials, molecules or compounds that have a biological effect on a biological cell, tissue or organ.
  • Suitable growth factors and cytokines include, but are not limited, to stem cell factor (SCF), granulocyte-colony stimulating factor (G-CSF), granulocyte-macrophage stimulating factor (GM-CSF), stromal cell-derived factor- 1, steel factor, VEGF, TGF ⁇ , platelet derived growth factor (PDGF), angiopoeitins (Ang), epidermal growth factor (EGF), bFGF, HNF, NGF, bone morphogenic protein (BMP), fibroblast growth factor (FGF), hepatocye growth factor, insulin-like growth factor (IGF-I), interleukin (IL)-3, IL-l ⁇ , IL-l ⁇ , IL-6, IL-7, IL-8, IL-Il, and IL-13, colony- stimulating factors, thrombopoietin, erythropoietin, fit3-ligand, and tumor necrosis factor ⁇ (TNFg).
  • SCF stem cell factor
  • suitable bioactive agents include but not limited to therapeutic agents.
  • therapeutic agent refers to a substance used in the diagnosis, treatment, or prevention of a disease. Any therapeutic agent known to those of ordinary skill in the art to be of benefit in the diagnosis, treatment or prevention of a disease is contemplated as a therapeutic agent in the context of the present invention.
  • Therapeutic agents include pharmaceutically active compounds, hormones, growth factors, enzymes, DNA, plasmid DNA, RNA, siRNA, viruses, proteins, lipids, pro-inflammatory molecules, antibodies, antibiotics, anti-inflammatory agents, anti-sense nucleotides and transforming nucleic acids or combinations thereof. Any of the therapeutic agents may be combined to the extent such combination is biologically compatible.
  • Exemplary therapeutic agents include, but are not limited to, those found in
  • therapeutic agents include but are not limited to, narcotic analgesic drugs; salts of gold; corticosteroids; hormones; antimalarial drugs; indole derivatives; pharmaceuticals for arthritis treatment; antibiotics, including Tetracyclines, Penicillin, Streptomycin and Aureomycin; antihelmintic and canine distemper drugs, applied to domestic animals and large cattle, such, as, for example, phenothiazine; drugs based on sulfur, such, as sulfioxazole; antitumor drugs; pharmaceuticals supervising addictions, such as agents controlling alcohol addiction and agents controlling tobacco addiction; antagonists of drug addiction, such, as methadone; weightcontrolling drugs; thyroid gland controlling drugs; analgesics; drugs controlling fertilization or contraception hormones; amphetamines; antihypertensive drugs; antiinflammatories agents; antitussives; sedatives; neuromuscular relaxants; antiepileptic drugs; antide
  • the amount of therapeutic agent distributed in a composition depends on various factors including, for example, specific agent; function which it should carry out; required period of time for release of a the agent; quantity to be administered.
  • dosage of a therapeutic agent i.e. amount of therapeutic agent in composition, is selected from the range about from 0.001% (w/w) up to 95% (w/w), preferably, from about 5% (w/w) to about 75% (w/w), and, most preferably, from about 10% (w/w) to about 60% (w/w).
  • Cisplatin [cis-dichlorodiammineplatinum(II)] (CDDP) has emerged as an important class of antitumor agents, and is widely used for the treatment of many malignancies including testicular , ovarian , cervical, head and neck, and non-small cell lung cancer (Jamieson, et al, Chem. Rev. (1999), 99(9): 2467-2498). It was also shown to be active in triple negative breast cancer (Leong, et al., J. Clin. Invest. (2007), 117(5): 1370-80). Its use is however dose-limited mainly because of nephrotoxicity or toxicity to the kidney (Madias, NE and Harrington, JT, Am. J.
  • a nanoliposomal formulation of cisplatin was found to deliver 50-200 times more drug to the tumor as compared to administration of free cisplatin (Harrington, et al. Ann. Oncol. (2001) 12: 493- 496). Although there was minimal toxicity with the nanoliposomal formulation, it had only modest antitumor activity as compared to cisplatin; reflecting the challenges of not only delivering platinum in a relatively inactive form, but the subsequent need to achieve significant release and activation within the tumor.
  • a second strategy of encapsulating cisplatin into polymeric systems has been a challenge as a result of its insolubility in organic solvents and partial solubility in water, which resulted in poor loading or inability to maintain sustained release.
  • Cisplatin gets activated through intracellular aquation of one of the two chloride leaving groups to form [Pt(NH 3 ) 2 Cl(OH 2 )] + and [Pt(NH 3 ) 2 (OH 2 )] 2+ , following which the Pt forms covalent bonds to the N 7 position of purine bases to form intrastrand and interstrand crosslinks (Huifang Huang, Leiming Zhu, Brian R. Reid, Gary P. Drobny, Paul B. Hopkins.
  • Cisplatin-Induced DNA Interstrand Cross-Link Science 1995: 270. 1842 - 1845.
  • carboplatin and oxaloplatin have a cyclobutane-l,l-decarboxylate and an oxalate respectively as the leaving groups, which chelate the platinum more strongly thus confering greater stability to the leaving group-PT complex and as a result exhibit fewer side effects than cisplatin but also lower efficacy than cisplatin (Richard J. Knox, Frank Friedlos, David A. Lydall and John J.
  • the inventors selected a 40-mer Poly(isobutylene-alt-maleic acid) (PIMA or PMA) as the polymer because each monomer exhibits dicarboxylato groups that can be complexed with cisplatin(OH) 2 . allowed the loading of a cisplatin molecule. Furthermore, hydrogenation of maleic acid generates succinic acid, which is a component of the Krebs cycle.
  • Poly(isobutylene-alt-maleic acid) 2 was synthesized from Poly(isobutylene-alt-maleic anhydride) 1 by reaction with water in DMF in one step as shown in Figure 1.
  • Cisplatin is a first line therapy for lung cancer, and as a result the inventors studied the effect of PMA-Cisplatin on the viability of Lewis lung cancer cells. Treatment with both cisplatin and PMA-cisplatin induced identical cell kill (Fig. 1C). However, PMA also induced tumor cell death. The inventors discovered that this can be overcome through derivatization of PMA. The inventors derivatized the polymer with ethylene diamine under basic conditions (Fig. 2). Interestingly, although the derivatization failed to remove the cytotoxicity of PMA, it increased the cytotoxicity of the PMA-cisplatin complex. This could potentially arise from the fact that the leaving group is less tightly bound as compared to underivatized PMA.
  • the increased efficacy with the derivatized chelated polymers as compared with the native polymer indicates that the monocarboxylato-chelated release drug much easily and showed superior activity over dicarboxylato-chelated (6).
  • the inventors discovered that the polymeric monocarboxylato-chelated platinum compounds represent a sizeable advantage over the conjugates in which the metal is bound via dicarboxylic acid. Smooth hydrolytic drug liberation from the carrier in the monocarboxylato-chelated derivatized PMA conjugates, as compared to the more retarded hydrolytic fission of the dicarboxylato-chelated in PMA, may explain this enormous difference in cell killing performance.
  • compositions described herein can be formulated into gels and used for sustained released delivery of bioactive agents at specific locations in a subject.
  • the composition can be used for sustained release delivery of platinum compounds at site of tumors.
  • the composition is used for sustain delivery of a platinum compound after a tumor has been removed.
  • the polymer linked platinum compound can be provided in pharmaceutically acceptable compositions.
  • These pharmaceutically acceptable compositions comprise a therapeutically-effective amount of one or more of the platinum compounds described herein, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents.
  • compositions of the present invention can be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), lozenges, dragees, capsules, pills, tablets (e.g., those targeted for buccal, sublingual, and systemic absorption), boluses, powders, granules, pastes for application to the tongue; (2) parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; (3) topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin; (4) intravaginally or intrarectally, for example, as a pessary, cream or foam; (5) sublingually; (6) ocularly; (7) transdermally; (8) transmucosally; or (9) nasal administration, for example, d
  • compounds can be implanted into a patient or injected using a drug delivery system. See, for example, Urquhart, et al., Ann. Rev. Pharmacol. Toxicol. 24: 199-236 (1984); Lewis, ed. "Controlled Release of Pesticides and Pharmaceuticals” (Plenum Press, New York, 1981); U.S. Pat. No. 3,773,919; and U.S. Pat. No. 35 3,270,960.
  • the term "pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • the term "pharmaceutically- acceptable carrier” means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid), or solvent encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body.
  • manufacturing aid e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid
  • solvent encapsulating material involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials which can serve as pharmaceutically- acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, methylcellulose, ethyl cellulose, microcrystalline cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) lubricating agents, such as magnesium stearate, sodium lauryl sulfate and talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol (PEG); (12) esters, such as ethyl
  • wetting agents, coloring agents, release agents, coating agents, sweetening agents, flavoring agents, perfuming agents, preservative and antioxidants can also be present in the formulation.
  • excipient e.g., pharmaceutically acceptable carrier or the like are used interchangeably herein.
  • terapéuticaally-effective amount means that amount of a compound, material, or composition comprising a compound of the present invention which is effective for producing some desired therapeutic effect in at least a sub- population of cells in an animal at a reasonable benefit/risk ratio applicable to any medical treatment.
  • an amount of a compound administered to a subject that is sufficient to produce a statistically significant, measurable change in at least one symptom of cancer or metastasis.
  • a therapeutically effective amount is well within the capability of those skilled in the art. Generally, a therapeutically effective amount can vary with the subject's history, age, condition, sex, as well as the severity and type of the medical condition in the subject, and administration of other pharmaceutically active agents.
  • administer refers to the placement of a composition into a subject by a method or route which results in at least partial localization of the composition at a desired site such that desired effect is produced.
  • a compound or composition described herein can be administered by any appropriate route known in the art including, but not limited to, oral or parenteral routes, including intravenous, intramuscular, subcutaneous, transdermal, airway (aerosol), pulmonary, nasal, rectal, and topical (including buccal and sublingual) administration.
  • oral or parenteral routes including intravenous, intramuscular, subcutaneous, transdermal, airway (aerosol), pulmonary, nasal, rectal, and topical (including buccal and sublingual) administration.
  • Exemplary modes of administration include, but are not limited to, injection, infusion, instillation, inhalation, or ingestion.
  • injection includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intraventricular, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, sub capsular, subarachnoid, intraspinal, intracerebro spinal, and intrasternal injection and infusion.
  • the compositions are administered by intravenous infusion or injection.
  • treatment means a human or animal. Usually the animal is a vertebrate such as a primate, rodent, domestic animal or game animal.
  • Primates include chimpanzees, cynomologous monkeys, spider monkeys, and macaques, e.g., Rhesus.
  • Rodents include mice, rats, woodchucks, ferrets, rabbits and hamsters.
  • Domestic and game animals include cows, horses, pigs, deer, bison, buffalo, feline species, e.g., domestic cat, canine species, e.g., dog, fox, wolf, avian species, e.g., chicken, emu, ostrich, and fish, e.g., trout, catfish and salmon.
  • Patient or subject includes any subset of the foregoing, e.g., all of the above, but excluding one or more groups or species such as humans, primates or rodents.
  • the subject is a mammal, e.g., a primate, e.g., a human.
  • the terms, "patient” and “subject” are used interchangeably herein.
  • the terms, “patient” and “subject” are used interchangeably herein.
  • the subject is a mammal.
  • the mammal can be a human, non- human primate, mouse, rat, dog, cat, horse, or cow, but are not limited to these examples. Mammals other than humans can be advantageously used as subjects that represent animal models of disorders associated with inflammation.
  • a subject can be male or female.
  • a subject can be one who has been previously diagnosed with or identified as suffering from or having a disorder a cancer or metastasis, but need not have already undergone treatment.
  • cancer includes, but is not limited to, solid tumors and blood born tumors.
  • the term cancer refers to disease of skin, tissues, organs, bone, cartilage, blood and vessels.
  • the term “cancer” further encompasses primary and metastatic cancers.
  • cancers that can be treated with the compounds of the invention include, but are not limited to, carcinoma, including that of the bladder, breast, colon, kidney, lung, ovary, pancreas, stomach, cervix, thyroid, and skin, including squamous cell carcinoma; hematopoietic tumors of lymphoid lineage, including, but not limited to, leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B -cell lymphoma, T-cell lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, hairy cell lymphoma, and Burketts lymphoma; hematopoietic tumors of myeloid lineage including, but not limited to, acute and chronic myelogenous leukemias and promyelocytic leukemia; tumors of mesenchymal origin including, but not limited to, fibrosarcoma, rhabdomyosarcoma, and osteosarcoma;
  • the compounds of the invention are also useful in combination with known anti-cancer treatments, including radiation.
  • the methods of the invention are especially useful in combination with anti-cancer treatments that involve administering a second drug that acts in a different phase of the cell cycle, e.g., S phase, than the epothilones of Formula (Ia) or (Ib), which exert their effects at the G2-M phase.
  • alkyl refers to saturated non-aromatic hydrocarbon chains that may be a straight chain or branched chain, containing the indicated number of carbon atoms (these include without limitation methyl, ethyl, propyl, iso-propyl, butyl, 2-methyl-ethyl, t-butyl, allyl, or propargyl), which may be optionally inserted with N, O, or S.
  • Ci-C 6 indicates that the group may have from 1 to 6 (inclusive) carbon atoms in it.
  • alkenyl refers to an alkyl that comprises at least one double bond.
  • alkenyl groups include, but are not limited to, for example, ethenyl, propenyl, butenyl, l-methyl-2-buten-l-yl and the like.
  • alkynyl refers to an alkyl that comprises at least one triple bond.
  • aryl refers to monocyclic, bicyclic, or tricyclic aromatic ring system wherein 0, 1, 2, 3, or 4 atoms of each ring may be substituted by a substituent.
  • Examplary aryl groups include, but are not limited to, benzyl, phenyl, naphthyl, anthracenyl, azulenyl, fluorenyl, indanyl, indenyl, naphthyl, phenyl, tetrahydronaphthyl, and the like.
  • cyclyl or "cycloalkyl” refers to saturated and partially unsaturated cyclic hydrocarbon groups having 3 to 12 carbons, for example, 3 to 8 carbons, and, for example, 3 to 6 carbons, wherein the cycloalkyl group additionally may be optionally substituted.
  • exemplary cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, cyclooctyl, and the like.
  • heteroaryl refers to an aromatic 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2, 3, or 4 atoms of each ring may be substituted by a substituent.
  • heteroaryl groups include, but are not limited to, pyridyl, furyl or furanyl, imidazolyl, benzimidazolyl, pyrimidinyl, thiophenyl or thienyl, pyridazinyl, pyrazinyl, quinolinyl, indolyl, thiazolyl, naphthyridinyl, and the like.
  • heterocyclyl refers to a nonaromatic 5-8 membered monocyclic, 8-
  • Examplary heterocyclyl groups include, but are not limited to piperazinyl, pyrrolidinyl, dioxanyl, morpholinyl, tetrahydrofuranyl, and the like.
  • the term "optionally substituted” means that the specified group or moiety, such as an alkyl group, alkenyl group, and the like, is unsubstituted or is substituted with one or more (typically 1-4 substituents) independently selected from the group of substituents listed below in the definition for "substituents" or otherwise specified.
  • substituted refers to a group “substituted” on an alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heterocyclyl, or heteroaryl group at any atom of that group.
  • Suitable substituents include, without limitation, halogen, hydroxy, oxo, nitro, haloalkyl, alkyl, alkenyl, alkynyl, alkaryl, aryl, aralkyl, alkoxy, aryloxy, amino, acylamino, alkylcarbanoyl, arylcarbanoyl, aminoalkyl, alkoxycarbonyl, carboxy, hydroxyalkyl, alkanesulfonyl, arenesulfonyl, alkanesulfonamido, arenesulfonamido, aralkylsulfonamido, alkylcarbonyl, acyloxy, cyano or ureido.
  • two substituents, together with the carbons to which they are attached to can form a ring.
  • polymer refers to the product of a polymerization reaction, and is inclusive of homopolymers, copolymers, terpolymers, tetrapolymers, etc.
  • polymer is also inclusive of random polymers, block polymers, graft polymers, copolymers, block copolymers, and graft copolymers.
  • copolymer refers to polymers formed by the polymerization reaction of at least two different monomers.
  • copolymer backbone refers to that portion of the polymer which is a continuous chain comprising the bonds formed between monomers upon polymerization.
  • composition of the copolymner backbone can be described in terms of the identity of the monomers from which it is formed without regard to the composition of branches, or sidechains, of the polymer backbone.
  • sidechain refers to portions of the monomer which, following polymerization, forms an extension of the copolymer backbone.
  • biocompatible refers to a material that is capable of interacting with a biological system without causing cytotoxicity, undesired protein or nucleic acid modification or activation of an undesired immune response.
  • Biocompatibility also includes essentially no interactions with recognition proteins, e.g., naturally occurring antibodies, cell proteins, cells and other components of biological systems.
  • an ester sidechains means a sidechains of the formula -
  • R"'C(O)-OR E where RE is independently Cl-C6alkyl, Cl-C6alkenyl, Cl-C6alkynyl, cyclyl, heterocycly, aryl, or heteroaryl, each of which can be optionally substituted; and R'" is a bond or C1-C6 alkylene, were the alkylene can comprise one or more double or triple bonds and/or the backbone of the alkylene can be interrupted by O, S, S(O), NH, or C(O).
  • R'" is a bond.
  • an amide sidechains means a sidechains of the formula -
  • R"C(O)-N(R A ) 2 where RA is independently H, Cl-C6alkyl, Cl-C6alkenyl, Cl-C6alkynyl, cyclyl, heterocycly, aryl, heteroaryl, saccharide, disaccharide, or trisaccharide, each of which can be optionally substituted; and R" is a bond or C1-C6 alkylene, were the alkylene can comprise one or more double or triple bonds and/or the backbone of the alkylene can be interrupted by O, S, S(O), NH, or C(O).
  • R" is a bond.
  • a carboxylic acid chain means a sidechains of the formula -
  • R""C(O)OH where R"" is a bond or C1-C6 alkylene, were the alkylene can comprise one or more double or triple bonds and/or the backbone of the alkylene can be interrupted by O, S, S(O), NH, or C(O).
  • R"" is a bond.
  • biocompatible polymers include polyamides, polycarbonates, polyalkylenes, polyalkylene glycols, polyalkylene oxides, polyalkylene terepthalates, polyvinyl alcohols, polyvinyl ethers, polyvinyl esters, polyvinyl halides, polyvinylpyrrolidone, polyglycolides, polysiloxanes, polyurethanes and copolymers thereof, alkyl cellulose, hydroxyalkyl celluloses, cellulose ethers, cellulose esters, nitro celluloses, polymers of acrylic and methacrylic esters, methyl cellulose, ethyl cellulose, hydroxypropyl cellulose, hydroxy-propyl methyl cellulose, hydroxybutyl methyl cellulose, cellulose acetate, cellulose propionate, cellulose acetate butyrate, cellulose acetate phthalate, carboxylethyl cellulose, cellulose triacetate, cellulose
  • biocompatible polymers and copolymers that have been modified for desirable enzymatic degradation, or change upon application of light, ultrasonic energy, radiation, a change in temperature, pH, osmolality, solute or solvent concentration are also amenable to the present invention.
  • a biocompatible conjugated polymer nanoparticle comprising: a copolymer backbone; a plurality of sidechains covalently linked to said backbone; and a plurality of platinum compounds dissociably linked to said sidechains.
  • platinum compound is a Pt(II) compound selected from the group consisting of cisplatin, oxaliplatin, carboplatin, paraplatin, sartraplatin, and combinations thereof.
  • a biocompatible conjugated polymer nanoparticle comprising: a poly(isobutylene- ⁇ /Mnaleic acid) backbone, wherein said backbone contains 25 to
  • PEG sidechains covalently linked to said backbone, wherein said PEG sidechains have a molecular weight of from 1000 to 3000 Dalton and wherein the number of said PEG sidechains corresponds to between 50% and 100% of the number of monomeric units of said polymer backbone; and a plurality of cisplatin sidegroups dissociably linked to said backbone wherein the number of said cisplatin sidegroups is between 25% and 75% of the number of monomeric units of said polymer backbone.
  • a biocompatible conjugated polymer nanoparticle comprising: a poly(isobutylene- ⁇ /Mnaleic acid) backbone, wherein said backbone consist of 40 monomers; a plurality of PEG sidechains covalently linked to said backbone, wherein said PEG sidechains have a molecular weight of 2000 Dalton and wherein the number of said
  • PEG sidechains is greater than 90% of monomeric units of said polymer backbone; and a plurality of cisplatin sidegroups dissociably linked to said backbone, wherein the number of said cisplatin sidegroups is between 25% and 75% of the number of monomeric units of said polymer backbone.
  • a biocompatible conjugated polymer nanoparticle comprising: a poly(isobutylene- ⁇ /Mnaleic acid) backbone, wherein said backbone comprises from
  • glucosamine sidechains covalently linked to said backbone and wherein the number of said glucosamine sidechains is between 50% and 100% of monomeric units of said polymer backbone; and a plurality of cisplatin sidegroups dissociably linked to said backbone, wherein the number of said cisplatin sidegroups is between 25% and 75% of the number of monomeric units of said polymer backbone.
  • a biocompatible conjugated polymer nanoparticle comprising: a poly(isobutylene- ⁇ /Mnaleic acid) backbone, wherein said backbone comprises from
  • glucosamine sidechains covalently linked to said backbone and wherein the number of said glucosamine sidechains is greater than 90% of monomeric units of said polymer backbone; and a plurality of cisplatin sidegroups dissociably linked to said backbone, wherein the number of said cisplatin sidegroups is between 25% and 75%, inclusive, of the number of monomeric units of said polymer backbone.
  • a carboxylic acid-platinum compound complex conjugated nanoparticle comprising: a carboxylic acid-platinum compound complex; and a plurality of lipid-polymer chains, wherein the carboxylic acid portion of said carboxylic acid-platinum compound complex is covalently bound to said lipid- polymer chains.
  • platinum compound is a Pt(II) compound selected from the group consisting of cisplatin, oxaliplatin, carboplatin, paraplatin, sartraplatin, and combinations thereof.
  • a vesicle, micelle, or liposome compound comprising a plurality of nanoparticles of any of paragraphs 33-39.
  • a vesicle, micelle, liposome or nanoparticle compound comprising a dicarbonyl-lipid compound of paragraph 41 and a platinum compound, wherein the platinum compound is dis sociably linked to the compound of paragraph 41.
  • platinum compound is selected from Pt(II) compounds, Pt(IV) compounds, and any combinations thereof.
  • platinum compound is a Pt(II) compound selected from the group consisting of cisplatin, oxaliplatin, carboplatin, paraplatin, sartraplatin, and combinations thereof.
  • a nanoparticle compound comprising a biocompatible polymer, wherein the polymer comprises at least one monomer having the formula -CH(CO 2 H)-R-CH(C(O)R')- , wherein R is a bond, Ci-C 6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
  • R is a bond.
  • a pharmaceutical composition comprising: the nanoparticle or compound of paragraphs 1-51; and a pharmaceutically acceptable carrier.
  • a method of treating cancer or metastasis comprising: administering to a subject in need thereof an effective amount of the composition of any of paragraphs 1-52.
  • cancer or metastasis is selected from the group consisting of breast, head and neck, ovarian, testicular, pancreatic, oral- esophageal, gastrointestinal, liver, gall bladder, lung, melanoma, skin cancer, sarcomas, blood cancers, brain tumors, glioblastomas, tumors of neuroectodermal origin and any combinations thereof.
  • a method of sustain release of a platinum compound at a specific location in a subject comprising: providing at the location a composition of paragraph the composition of any of paragraphs 1-52.
  • composition is in the form of a gel.
  • CellTiter 96 AQueous One Solution Cell Proliferation Assay [3-(4,5- dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt (MTS) assay] reagent was from Promega (Madison, WI). All Polymer solutions were dialyzed in cellulose membrane tubing, types Spectra/Por 4 and Spectra/Por 6 (wet tubing), with mass-average molecular mass cut-off limits of 1000 and 3500 respectively. Operations were performed against several batches of stirred deionized H2O.
  • (4Tl) were purchased from American Type Culture Collection (ATCC, Rockville, MD, USA). Lewis Lung Carcinoma cells were cultured in Dulbecco's Modified Eagle's Medium supplemented with 10% FBS, 50 unit/ml penicillin and 50 unit/ml streptomycin. The 4Tl cells were cultured in RPMI medium supplemented with 10% FBS, 50 unit/ml penicillin and 50 unit/ml streptomycin. Trypsinized cultured LLC and 4Tl cells were washed twice with PBS and seeded into 96-well flat bottomed plates at a density of 2xlO 3 cells in 100 ⁇ l of medium. Different concentrations of conjugates were tested in triplicate in the same 96-well plate for each experiment.
  • High resolution TEM images were obtained on a JEOL 2011 high contrast digital TEM.
  • Samples were prepared on carbon 300 mesh copper grids (Electron Microscopy Sciences) by adding drops of aqueous nanoparticles at different concentrations, and allowed to air-dry.
  • the size distribution of nanoparticles was studied by dynamic light scattering (DLS), which was performed at 25°C on a DLS-system (Malvern NanoZetasizer) equipped with a He Ne laser.
  • DLS dynamic light scattering
  • concentrated PIMA-GA-cisplatin conjugate was resuspended into 100 ⁇ L of double distilled water with pH adjusted to 8.5 or 5.5 using IN sodium hydroxide or IN nitric acid and transferred to a dialysis tube (MWCO: 1000 KD, Spectrapor).
  • the dialysis tube was put into a tube containing magnetic pallet and 2 mL solutions of different pH phosphate-buffered saline. Cisplatin release was studied by gently stirring the dialysis bag at 300 rpm using IKA stirrer at 25oC.
  • Cells were grown in 6-well plates incubated in the presence of cisplatin nanoparticle or free cisplatin at 37 0 C for 24 h. After 24 h, the cells were washed with PBS and collected at O 0 C. The cells were then treated with annexin V-Alexa Fluor 488 conjugate (Molecular Probes, Invitrogen) and incubated in the dark, at room temperature, for 15 min. The cells were then washed with PBS and incubated with propidium iodide (PI) solution (50 g/mL; Sigma) containing RNase (1 mg/mL; Sigma). The cell suspensions were then transferred to FACS tubes and analyzed for Annexin V/PI staining on a BD FACS Calibur instrument. Data were analyzed using a CellQuestPro software (BD Biosciences).
  • PI propidium iodide
  • [00191] LLC and 4Tl cells were seeded on glass coverslips in 24-well plates, 50000 cells per well. When cells reached 70% confluency, they were treated with fluorescein isothiocyanate (FITC)-conjudated cisplatin nanoparticles for different durations of 30 min, 2 h, 6 h, 12 h, and 24 h, respectively. For colocalization studies, at indicated time points, the cells were washed with PBS and incubated with Lysotracker Red (Molecular Probes) at 37 0 C for 30 min to allow internalization.
  • FITC fluorescein isothiocyanate
  • the cells were then fixed with 4% paraformaldehyde for 20 min at room temperature, then washed twice with PBS and mounted on glass slides using Prolong Gold Antifade Reagent (Molecular Probes). Images were obtained using a Nikon Eclipse TE2000 fluorescence microscope equipped with green and red filters for FITC and Lysotracker Red, respectively.
  • LLC Lung Cancer cells and 4Tl Breast Cancer cells were implanted subcutaneously in the flanks of 4- week-old C57/ BL6 and BALB/c mice (weighing 20 g, Charles River Laboratories, MA) respectively.
  • the drug therapy was started after the tumors attained volume of 50 mm 3 .
  • the tumor therapy consisted of administration of cisplatin nanoparticle and free cisplatin or oxaliplatin and free oxaliplatin.
  • the formulations were prepared and validated such that 100 ⁇ L of cisplatin nanoparticle and free cisplatin contained 1.25 and 3 mg/kg of cisplatin or 100 ⁇ L of oxaliplatin nanoparticle and free oxaliplatin contained 5 and 15 mg/kg of oxaliplatin Administration was by tail vein injection.
  • PBS 100 ⁇ L administered by tail-vein injection was used as a control for drug treatment.
  • the tumor volumes and body weights were monitored on a daily basis. The animals were sacrificed when the average tumor size of the control exceeded 2000 mm 3 in the control group.
  • the tumors were harvested immediately following sacrifice and stored in 10% formalin for further analysis. All animal procedures were approved by Harvard institutional IUCAC.
  • Ovarian adenocarcinomas were induced in genetically-engineered K- ras LSL/+ /Pten fl/fl mice via intrabursal delivery of adenovirus carrying Cre recombinase, as described previously.
  • Tumor cells were engineered to express luciferase once activated by Adeno-Cre, in order to make tumor imaging feasible before and after drug treatment.
  • mice Once mice developed medium to large tumors they were placed into one of four treatment groups (controls, cisplatin NPl.25 mg/kg, cisplatin NP-3 mg/kg, and free cisplatin), with all drugs administered intravenously (Lv.).
  • Cisplatin-nanoparticles and free cisplatin were injected i.v. (dose equivalent to
  • mice 8 mg/Kg of cisplatin in mice to study its distribution.
  • the animals were sacrificed and necropsy was performed to harvest the tumor and kidney.
  • the animals were dosed repeatedly following the efficacy study protocols, and the animals were sacrificed at the end of the multiple dosing study.
  • the organs were then weighed and dissolved in Cone. HNO 3 (approx. 10 mL) by shaking for 24 hours at room temperature and then heating at 100 0 C for 12 hours. To these mixtures were then added 30% H 2 O 2 , the resulting solutions were stirred for 24 hours at room temperature and then heated for another 12 hours to evaporate the liquids. All solid residues were re-dissolved in 1 mL water and then amount of platinum was measured by inductively coupled plasma- spectrometry (ICP).
  • ICP inductively coupled plasma- spectrometry
  • Toxicity Assessment of Drug Treatment The tissues were fixed in 10% formalin, paraffin embedded, sectioned and stained with H&E at the Harvard Medical School Core Facility. Tumor and Kidney paraffin sections were deparaffinized and stained with standard TMR-red fluorescent terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) kit following the manufacturer's protocol (In situ Cell Death Detection Kit, TMR Red, Roche). Images were obtained using a Nikon Eclipse TE2000 fluorescence microscope equipped with red filter. Toxicity Assessment of Drug Treatment
  • Body weights were recorded daily to assess toxicity.
  • livers and spleens were removed at the end of treatment to record weights and perform extensive pathological examination to assess toxicity of vital organs.
  • Cell apoptosis in vital organs was measured using TUNNEL assay.
  • Statistical analysis of the toxicity data was performed using a two-way ANOVA test with the Prism 5TM software.
  • CDDP (30mg) and AgNO 3 (17mg) was added to 10ml double distilled water.
  • the resulting solution was stirred in dark at room temperature for 24h. AgCl precipitates were found after reaction. AgCl precipitates are removed from reaction by centrifugation at 10000 rpm for 10 min. The supernatant was further purified by passing through 0.2 ⁇ m filter.
  • the brush polymer PIMA-PEG 5 (0.019 g, 0.00007 mmol) was taken in 10 niL RB flask mixed with CDDP (0.0002 g, 0.0007 mmol) dissolved in 0.3 mL double distilled water. After stirring for 3 days at room temperature (25°C) the resulting turbid reaction mixture was dialyzed.
  • the solution containing PIMA-PEG-CDDP (2) conjugate was further purified by dialyzing it in cellulose membrane tubing, types Spectra/Por 4 and with mass-average molecular mass cut-off limits of 1000 for 2-3 hours to remove free CDDP.
  • PIMA-PEG-CDDP (9) conjugate was then lyophilized to get white colored solid. The conjugate was re- suspended in double distilled water for cell culture experiments.
  • the resulting orange solid was purified by dialysis for 3 days using dialysis bag of molecular cut off of 3.5KD. Lyophilization gave fluorescent orange PIMA-GA-FITC polymer.
  • PIMA-GA-FITC 0.004 g
  • ImI double distilled water containing cisplatin 0.001 g
  • the PIMA-GA-FITC-cisplatin conjugate formed in solution was further purified by dialysis to remove unattached cisplatin with mass-average molecular mass cut-off limits of 1000. Lyophilization of the dialyzed solution resulted in orange colored FITC labeled PEVIA-GAFITC-cisplatin conjugate nanoparticles.
  • PIMA Poly(isobutylene-alt-maleic acid)
  • ImI double distilled water containing oxaliplatin-OH (lmg) was dissolved in ImI double distilled water containing oxaliplatin-OH (lmg) in a round bottom flask to and then resulting reaction mixture was stirred at room temperature (25oC) for 48h.
  • the PIMA-oxaliplatin conjugate was further purified by dialyzing it in cellulose membrane tubing, types Spectra/Por 4 and with mass-average molecular mass cut-off limits of 1000. The resulting turbid solution was then lyophilized to get PIMA-oxaliplatin conjugate.
  • the conjugate was re-suspended for cell culture experiments.
  • Glucosamine hydrochloride 360 mg, 1.66 mmol, 200 equiv was suspended in 5 mL DMF and treated with DBU (250 ⁇ L, 1.66 mmol, 200 equiv) at room temperature for Ih. After Ih glucosamine/DBU (in DMF) solution was added drop wise into poly (isobutylene-alt-maleic anhydride) (50 mg, 0.008 mmol, 1 equiv) solution in 5 mL DMF and the reaction mixture was stirred for 72h at room temperature. The reaction mixture was quenched with 3 mL of dd-water. The PIMA-GA conjugate was purified by dialysis using 2000 MWCO dialysis bag for 72h.
  • the product was lyophilized for 48h to obtain 100 mg cream yellow powder.
  • the product was characterized by 1 H NMR spectroscopy (300 MHz). Solubility: product was soluble in water but not soluble in organic solvent e.g. acetone, methanol or acetonitrile.
  • Glucosamine hydrochloride (179 mg, 0.83 mmol, 100 equiv) was suspended in 2 mL DMF and treated with DIPEA (145 ⁇ L, 0.83 mmol, 100 equiv) at room temperature for Ih. After Ih poly (isobutylene-alt-maleic anhydride) (50 mg, 0.008 mmol, 1 equiv) (dissolved in 3 mL DMF) was added into the reaction mixture and stirred for 24h at room temperature. The reaction mixture was quenched with 3 mL of dd- water. The PIMA-GA conjugate was purified by dialysis using 1000 MWCO dialysis bag for 24h.
  • the product was lyophilized for 48h to obtain 106 mg white powder.
  • the product was characterized by 1 H NMR spectroscopy (300 MHz). Solubility: product was soluble in water but not soluble in organic solvent e.g. acetone, methanol or acetonitrile.
  • Example 4 Time dependent loading efficiency of PIMA-GA-CDDP.
  • the figure shows that loading of platinum in the polymer conjugate increases with time from 190 ⁇ g/mg (5h) to 210 ⁇ g/mg (3Ih) and reaches maximum 347 ⁇ g/mg at 48h. This indicates almost 100% of the Pt is complexed with the polymer at this time point as the maximal predicted loading is 37.5% per polymer unit, and we attain 34.7% Pt per polymer.
  • Example 5 Rational optimization of the polymer based on structure-activity relationship.
  • the inventors derivatized one arm of each monomer of the polymer with biocompatible glucosamine to generate PIMA- glucosamine conjugate (PIMA-GA) (Fig.1 IB). This converted the dicarboxylato bonds with Pt to a monocarboxylato bond and a coordinate bond, which can release Pt more easily given that a coordinate bond is less stable than a monocarboxylato linkage (Fig. 11 B).
  • Nuclear magnetic resonance (NMR) characterization of the Pt environment revealed that complexation of PIMA-GA and cisplatin in an acidic pH (pH 6.5) generated an isomeric state [PEVIA-GA-Cisplatin (0->Pt)] (8) characterized by the monocarboxylato and a 0->Pt coordination complex as characterized by a single Pt NMR peak at -1611.54 (Fig. HB).
  • Tagging the polymer with fluorescein enabled the temporal tracking of uptake of the nanoparticles into the cells, which were co-labeled with a lysotracker-red dye to label the endolysosomal compartments.
  • a rapid uptake of the nanoparticles was observed in the LLC cells within 15 min of treatment with internalization into the endolysosomal compartment as evident by colocalization of the FITC-nanoparticles and the Lysotracker-Red dye (data not shown).
  • the uptake into 4Tl cells was delayed, with internalization into the endolysosomal compartment evident only after 2 hours post-incubation. Over a 12 hour period, the fluorescent signals from the lysosomal compartment and the FITC- conjugated dissociate, suggesting a cytosolic distribution of the polymer after processing within the lysosome (data not shown).
  • Example 7 The release of active cisplatin from nanoparticle is pH-dependent [00224] Given that the nanoparticles localized to the lysosomal compartment, the inventors tested the release of Pt from the nanoparticles at pH 5.5, mimicking the acidic pH of the endolysosomal compartment of the tumor (Lin, et al., Eur. J. Cancer, 2004 40(2):291- 297). The inventors also selected pH8.5 as a reference pH in the alkaline range. As shown in Fig. 16, at pH5.5 PIMA-GA-cisplatin (0->Pt) nanoparticles resulted in a sustained but significant release of cisplatin monitored over a 70 hour period.
  • Example 8 - Nanoparticle induces tumor growth delay and regression with reduced nephrotoxicity
  • mice randomly sorted mice bearing established Lewis lung carcinoma or 4Tl breast cancer into five groups respectively and treated each group with three doses of (i) PBS (control); (ii) Cisplatin (1.25 mg/kg); (iii) Cisplatin(3mg/kg); (iv) PIMA-GA-Cisplatin (0->Pt) nanoparticles (1.25 mg/kg); (v) PIMA- GA-Cisplatin (0->Pt) nanoparticles (3 mg/kg).
  • the mice injected with PBS formed large tumors by day 16 (day after the last injection), and consequently, were euthanized.
  • the animals in the other groups were also sacrificed at the same time point to evaluate the effect of the treatments on tumor pathology. As shown in Fig.
  • the inventors labeled the tumor cross sections for TUNEL, which revealed a significant induction of apoptosis following treatment with both free cisplatin and PIMA- GAcisplatin(O->Pt) nanoparticles (data not shown).
  • labeling the kidney sections for TUNEL demonstrated significant apoptosis in the animals treated with free cisplatin as opposed to minimal nephrotoxicity in the nanoparticle-treated group (data not shown).
  • biodistribution studies using inductively coupled plasma-spectrometry (ICP) revealed that the concentration of Pt in the kidney following administration of the cisplatin nanoparticle is 50% of that attained following administration of free drug (Fig. 5E), which can explain the reduction in nephrotoxicity.
  • the increased therapeutic index can arise from a preferential accumulation of the nanoparticles in the tumors arising from the well-studied EPR effect, and circumventing the kidney as it exceeds the size limit for clearance, which in a previous study was shown to be less than 5 nm (Choi HS, Liu W, Misra P, Tanaka E, Zimmer JP, Itty Ipe B, Bawendi MG, Frangioni JV. Renal clearance of quantum dots. Nat Biotechnol. 2007 ;25: 1165-70).
  • the inventors further evaluated the PIMA-GA-cisplatin(O->Pt) nanoparticle in an ovarian cancer model.
  • Epithelial ovarian cancer is the deadliest malignancies of the female reproductive cycle.
  • the discovery of frequent somatic PTEN mutations and loss of heterozygosity at the 10q23 PTEN locus in endometrioid ovarian cancer implicates a key role for PTEN in the etiology of this epithelial ovarian cancer subtype (Obata, K. et al. Frequent PTEN/MMAC1 mutations in endometrioid but not serous or mucinous epithelial ovarian tumors. Cancer Res.
  • K-RAS oncogene is also mutated in endometrioid ovarian cancer, albeit at a lesser frequency (Cuatrecasas, et al., Cancer (1998) 82:1088-1095).
  • the combination of these two mutations in the ovarian surface epithelium was found to induce invasive and widely metastatic endometrioid ovarian adenocarcinomas with complete penetrance, making it a good model for mimicking human tumor progression.
  • TUNEL staining revealed significant apoptosis in the kidney at 3 mg/kg of free cisplatin while the cisplatinnanoparticles at equivalent Pt concentration did not induce apoptosis of the nephrons.
  • the inventors have also engineered an analog, where maleic acid is conjugated to PEG end of a pegylated lipid (PEG2000-DSPE).
  • PEG2000-DSPE pegylated lipid
  • the inventors complexed Pt to the maleic acid, resulting in the formation of a platinated lipid derivative where the Pt is at the hydrophilic end and the lipid forms the hydrophobic end.
  • a measured amount of the extruded lipo-nanoparticle was heated at 100 0 C in
  • mice were inoculated s.c. with IXlO 5 of 4Tl breast tumor cells inlOO ⁇ L PBS on right flank of mice. Treatment with different anticancer agents either free or entrapped in nanop articles was started on day when tumor volume reached 200 mm . Typically the animals received free drug alone or in nanoparticles through i.v route every alternate day for total of three dosages. Once the tumor volume reached 2000 mm3 in control group, mice were sacrificed. Tumor, kidney, spleen, lung, liver were harvested and processed for paraffin embedding and sectioning.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Molecular Biology (AREA)
  • Polymers & Plastics (AREA)
  • Biophysics (AREA)
  • Nanotechnology (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Inorganic Chemistry (AREA)
  • Immunology (AREA)
  • Oncology (AREA)
  • Pulmonology (AREA)
  • Endocrinology (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Reproductive Health (AREA)
  • Hematology (AREA)
  • Neurology (AREA)
  • Dermatology (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Addition Polymer Or Copolymer, Post-Treatments, Or Chemical Modifications (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Steroid Compounds (AREA)

Abstract

The invention is directed to biocompatible conjugated polymer nanoparticles including a copolymer backbone, a plurality of sidechains covalently linked to said backbone, and a plurality of platinum compounds dissociably linked to said backbone. The invention is also directed to dicarbonyl-lipid compounds wherein a platinum compound is dissociably linked to the dicarbonyl compound. The invention is also directed to methods of treating cancer or metastasis. The methods includes selecting a subject in need of treatment for cancer or metastasis and administering to the subject an effective amount of any of the nanoparticles, compounds, or compositions of the invention.

Description

NANOSCALE PLATINUM COMPOUNDS AND METHODS OF USE THEREOF
RELATED APPLICATIONS
[0001] This application claims priority to and benefit under 35 U. S. C. § 119(e) of the
U.S. Provisional Application Nos. 61/149,725, filed February 4, 2009 and 61/240,007, filed September 4, 2009, the content of both of which are incorporated herein by reference in their entirety.
GOVERNMENT SUPPORT
[0002] The subject matter of this application was made with support of a Department of Defense Era of Hope Scholar Award W81XWH-07- 1-0482 and Department of Defense Postdoctoral Award W81XWH-09- 1-0728. The U.S. Government has certain rights in this invention.
FIELD OF THE INVENTION
[0003] This invention relates to biocompatible conjugated polymer nanoparticles including a copolymer backbone, a plurality of sidechains covalently linked to the backbone, and, a plurality of platinum compounds dissociably linked to the backbone.
BACKGROUND OF THE INVENTION
[0004] Cancer is the second leading cause of mortality in the United States, with an estimated 1,444,180 new cases and 565,650 deaths in 2008. Cytotoxic agents, which are used in standard chemotherapy, non- specifically target all dividing cells resulting in dose- limiting toxicities. There is an urgent need to develop novel strategies that are more specifically targeted against the tumor.
[0005] The use of nanovectors holds the potential to revolutionize cancer chemotherapy by specifically targeting the tumor. A number of polymeric nanovectors are currently in development or in clinics, and are dramatically altering the pharmacodynamic and pharmacokinetic profile of the active agent. However, most of these polymeric constructs decrease the potency of the conjugated active agent, relying on increased uptake into the tumor for the improved therapeutic index. [0006] Cisplatin is one of the mainstays in chemotherapy regimes for most types of
Cancer (Kelland L. The resurgence of platinum-based cancer chemotherapy. Nat Rev Cancer. 2007 Aug;7(8):573-84). However, its use is does-limited due to severe nephrotoxicity . Furthermore, the nanovector formulation of cisplatin, which is a first-line therapy for multiple cancers, has been a challenge.
SUMMARY OF THE INVENTION
[0007] Reported herein is a rational engineering of a polymeric construct of platinum-based chemotherapeutics such as cisplatin and oxaliplatin, which results in self- assembly into a nanoparticle. The nanoparticle sustains the potency of the active agent, and compared with cisplatin or oxaliplatin or carboplatin, exhibits increased anti-tumor effects with reduced systemic- and nephro-toxicity when administered intravenously to tumor- bearing mice. This nanotechnology-enabled improvement in the therapeutic index of cisplatin or oxaliplatin can be utilized for using nanoplatinates in the clinical management of multiple types of cancer.
[0008] The invention is directed to a biocompatible conjugated polymer nanoparticle including a copolymer backbone, a plurality of sidechains covalently linked to said backbone, and, a plurality of platinum compounds dissociably linked to said backbone. Generally, the platinum compound is dissociably linked to backbone via linkage through the sidechain. In some embodiments, the platinum compound is linked to the sidechain through at least one coordination bond.
[0009] Another aspect of the invention is directed to biocompatible conjugated polymer nanoparticles including a polymaleic acid (PMA) such as a poly(isobutylene-α/£- maleic acid) (PIMA) backbone. The backbone consists of from 25 to 50 monomers. Also included are a plurality of PEG sidechains covalently linked to said backbone. The PEG sidechains have a molecular weight of from 200 to 3000 Dalton. The PEG sidechains number between 50% and 100%, inclusive, of the number of monomeric units of the polymer backbone. Also included are a plurality of cisplatin or oxaliplatin side groups dissociably linked to the backbone. The cisplatin side groups number between 25% and 75%, inclusive, of the number of monomeric units of the polymer backbone.
[0010] Yet another aspect of the invention is directed to biocompatible conjugated polymer nanoparticles including a poly(isobutylene-α/Mnaleic acid) backbone. The backbone consist of about 40 monomers. Also included are a plurality of PEG sidechains covalently linked to the backbone. The PEG sidechains have a molecular weight of about 2000 Dalton. The PEG sidechains number greater than 90% of monomeric units of said polymer backbone. Also included are a plurality of cisplatin or oxaliplatin side groups dissociably linked to the backbone. The cisplatin or oxaliplatin side groups number between 25% and 75%, inclusive, of the number of monomeric units of the polymer backbone. [0011] Still another aspect of the invention is directed to biocompatible conjugated polymer nanoparticles including a poly(isobutylene-α/Mnaleic acid) backbone. The backbone consists of from 25 to 50 monomers. Also included are a plurality of glucosamine sidechains covalently linked to said backbone. The glucosamine sidechains number between 50% and 100%, inclusive, of monomeric units of said polymer backbone. Also included are a plurality of cisplatin or oxaliplatin side groups dissociably linked to the backbone. The cisplatin or oxaliplatin side groups number between 25% and 75%, inclusive, of the number of monomeric units of the polymer backbone.
[0012] Another aspect of the invention is directed to biocompatible conjugated polymer nanoparticles including a poly(isobutylene-α/Mnaleic acid) backbone. The backbone consists of from 25 to 50 monomers. Also included are a plurality of glucosamine sidechains covalently linked to said backbone. The glucosamine sidechains number greater than 75% of monomeric units of said polymer backbone. Also included are a plurality of cisplatin or oxaliplatin side groups dissociably linked to the backbone. The cisplatin or oxaliplatin side groups number between 25% and 75%, inclusive, of the number of monomeric units of the polymer backbone.
[0013] Yet another aspect of the invention is directed to carboxylic acid-platinum II
(Pt(II)) complex conjugated nanoparticles including a carboxylic acid-(Pt(II)) complex and a plurality of lipid-polymer chains. The carboxylic acid portion of said carboxylic acid- cisplatin/oxaliplatin complex is covalently bound to said lipid-polymer chains. [0014] Another aspect of the invention is directed to a vesicle, micelle, or liposome compound comprising a plurality of nanoparticles of claim as described herein. [0015] Still another aspect of the invention is directed to pharmaceutical compositions including any of the nanoparticles or compounds described herein and a pharmaceutically acceptable carrier.
[0016] Yet another aspect of the invention is directed to a method of treating cancer or metastasis. The method includes selecting a subject in need of treatment for cancer or metastasis and administering to the subject an effective amount of any of the nanoparticles, compounds, or compositions described herein. BRIEF DESCRIPTION OF THE DRAWINGS
[0017] Figure 1 shows a schematic of synthesis of PMA-Cisplatin. Loading of different numbers of cisplatin per polymer affects the size of the nanoparticles as measured using DLS or TEM. And a graph of Cytotoxicity study of Polyisobutylene maleic acid carrier
(PIMA) 2 and conjugate (PIM A-CISPLATIN) (6).
[0018] Figure 2 shows a scheme showing derivatization of PMA with EDA. The derivatized polymer was used to synthesize the cisplatin-complex. The graph shows the effect of different treatments on LLC viability following 48 hours of incubation.
[0019] Figure 3 shows a scheme of the synthesis of PMA-GA-Cisplatin.
Complexation with cisplatin was carried out over a 48 hour period. This resulted in the formation of nanoparticles in the size range of around 100 nm as seen from the DLS measurements.
[0020] Figure 4 The graph on the left shows the amount of active cisplatin that is released from the PMA-GA-Cisplatin nanoparticle when incubated with LLC lysate. The concentration-effect graph of the right shows the effect of different treatments on the viability of Lewis Lung Carcinoma cells when incubated with the active agents for 48 hours. Cell viability was measured using an MTS assay.
[0021] Figures 5A-5E are line graphs (Figs.5A and 5B) and bar graphs (Figs. 5C-5E) showing efficacy and toxicity profile of free and nanoparticle cisplatin in Lewis Lung carcinoma model. Tumors were induced by injecting LLC cells in c57/BL6 mouse. The effect of treatments on tumor volume (Fig, 15A) and body weight (Fig. 5B) over the treatment period is shown. The animals were dosed thrice (shown by arrows on x-axis). Data shown are mean+SE, n=4-8. The effect of treatment on the organ weight of kidneys (Fig.
5C) and spleen (Fig. 5D) as a marker for nephrotoxicity and hematological toxicity is also shown (n=4-6). The images on top of each graph show representative organs from each treatment group. Fig. 5E shows the biodistribution of Pt in kidney and tumor as measured using ICP-spectroscopy 24 h hours following administration of free or cisplatin nanoparticles
(8mg/kg cisplatin dose)
[0022] Figure 6 is a scheme showing synthesis of PMA-GA-Cisplatin (8).
[0023] Figure 7 Effect of PMA-PEG-Cisplatin on Lewis Lung Carcinoma. Cell were incubated for 48 hours with the drugs or vehicles and then tested for viability using an MTS assay. [0024] Figure 8 Effect of different treatments on tumor growth and body weight in vivo. Tumors were induced by injecting LLC cells in c57/BL6 mouse. [0025] Figure 9 is a graph of the amount of platination of the polymer was quantified using a uv-vis spectroscopy method.
[0026] Figure 10 Scheme showing synthesis of lipid maleic acid-cipslatin complex, which can form micelles in water.
[0027] Figures HA and HB aree schematics showing SAR-inspired engineering of a cisplatin nanoparticle. Fig. HA shows the mechanism underlying the intracellular activation of cisplatin through aquation. The leaving groups of cisplatin and analogues (shown with blue lines) are replaced with OH prior to DNA-binding. Fig. HB shows the chemical synthesis of PIMA-cisplatin and PIMA-glucosamine (PIMA-GA)-cisplatin complex. Transformation of polymaleic anhydride (n=40) (1) to polymaleic acid [PIMA] (2) enables complexation of [NHi]2Pt[OH]2 through dicarboxylato bond (6). Derivatization of one arm of PEVIA with glucosamine (4), and complexation with [NH2]2Pt[OH]2 can lead to two isomers (8) and (10) depending on pH, characterized by unique Pt NMR signatures (Fig. 11B). [0028] Figures 12A and 12B show characterization of cisPt-NP. Increasing the number of Pt on a PDVIA (n=40) backbone increased the size of nanoparticle formed. At an optimal Pt to polymer ratio, the inventors obtained nanoparticles smaller than 150 nm, the size cut-off below which enables preferential homing to tumors. Fig. 12A shows that derivatization of all the monomeric units of PDVIA with glucosamine and subsequent complexation with Pt forms nanoparticles smaller than 150 nm. Fig. 12B shows the total platinum loaded per mg of polymer at this ratio.
[0029] Figures 13A-13E are line graphs showing in vitro characterization of cisplatin nanoparticles. Figs. 13A and 13B show the concentration-effect of different treatments on cellular viability as measured using MTS assay. X-axis shows the equivalent concentrations of cisplatin. Where blank polymeric controls were used, dose of polymer used was equivalent to that used to deliver that specific dose of cisplatin in the complexed form. PDVIA was also derivatized with ethylene diamine to generate PDVIA-EDA, which offers a similar complexation environment to platinum as PDVIA-GA. Unlike PIMA-GA, PIMA-EDA exerted inherent toxicity. PIMA-GA-Cisplatinfacidic] refers to the isomer formed under acidic complexation environment while PDVIA-GA-Cisplatin[basic] refers to the isomer formed under alkaline environment. Figs. 13C-13E show effect of PIMA-GA-cisplatin nanoparticles on LLC cells viability, when the PDVIA backbone (40 monomeric units) is derivatized to different degrees. PIMA-30GA-Cisplatin has 30 of the 40 monomeric units derivatized with glucosamine while PIMA-GA-40 and PIMA-GA-200 have all the monomeric units derivatized. [a] and [b] refers to isomers formed in acidic and basic environments when the polymers are complexed with cisplatin. Table 1 shows the corresponding IC50 values. [0030] Figures 14A-14J show FACS images (Figs. 14A- 14H) and bar graphs (Figs.
141 and 14J) showing that treatment with PJJVIA-GA-Cisplatin induces cell death. Representative FACS images of 4Tl (Figs. 14A-14D) and LLC (Figs. 14E- 14H) cells showing the percentage in each quadrant following treatments with free or nanoparticle - cisplatin. Carboplatin was used as a control for comparison (Figs. 14D and 14H). The cells were incubated with the drugs for 24h, following which they were labeled with Annexin-V FITC and counter stained with propidium iodide.
[0031] Figurel5 is a schematic showing the labeling of the PEVIA-GA polymer with
FITC to enable the tracking of cellular uptake of the nanoparticles. [0032] Figure 16 is a line graph showing the effect of pH and Pt complexation environment on release kinetics. The nanoparticles were incubated at pH 5.5 or pH8.5 in a dialysis bag, and release over time was quantified. The nanoparticles [PIMAGA-Cisplatin (0->Pt)] used were generated by complexing the polymer and cisplatin in an acidic pH [6.4] unless in the case of PIMA-GA-Cisplatin (Pt->N), where the complexation was carried out in a basic pH to generate the stable isomer [PEVIA-GA-Cisplatin (N->Pt)]. The data shown are mean+SE from n=3.
[0033] Figures 17A-17D are line graphs (Figs. 17A and 17B) and bar graphs (Figs.
17C and 17D) showing PIMA-GA-cisplatin nanoparticle exerts similar anti-tumor effect with reduced systemic toxicity compared to free cisplatin in a 4Tl breast cancer model. Line graphs show the effect of treatments on tumor volume (Fig. 17A) and body weight (Fig. 17B) over the treatment period. The animals were dosed thrice (shown by arrows on x-axis). Data shown are mean+SE, n=4-8. Bar graphs show the effect of treatment on the organ weight of spleen (Fig. 18C) and kidneys (Fig. 17D) as a marker for nephrotoxicity and hematological toxicity (n=4-6)*P<0.05 vs vehicle-treated group [ANOVA followed by Newman Keuls Post Hoc test]. Carboplatin [3mg/kg] was used as a control.
[0034] Figures 18A and 18B are a bar graph (Fig. 18A) and a line graph (Fig. 18B) showing PIMA-GA-cisplatin nanoparticle inhibits tumor growth in a K-rasLSI7+/Ptenfl/fl ovarian cancer model. As shown in Fig. 18A, bioluminescence quantification indicated a significantly decreased tumor luciferase signal in mice treated with cisplatin-NP compared to vehicle (p< 0.05, one-way ANOVA analysis). Fig. 18B shows drug toxicity assessed by measurements in overall body weight. Daily recording of body weights indicated a significant loss of body weight in the free cisplatin group as compared to both cisplatin-NP (1.25 mg/kg and 3 mg/kg) treated groups (P < 0.05, two-way ANOVA analysis). [0035] Figures 19A and 19B are bar graphs showing the distribution of Pt following administration of cisplatin, cisplatin-nanoparticle ([PIMA-GA-Cisplatin (0->Pt)] or carboplatin in breast cancer or ovarian cancer. Treatment was administered as described in Figs. 17 and 18. The level of Pt in different tissues harvested following necropsy was quantified using inductively coupled plasma- spectrometry (ICP).
[0036] Figures 2OA and 2OB are schematic showing SAR- inspired engineering of a oxaliplatin nanoparticle. Figure 2OA shows the mechanism underlying the intracellular activation of oxaliplatin through aquation. Figure 2OB shows the chemical synthesis of PIMA-oxaliplatin and PIMA-glucosamine (PIMA-GA)-oxaliplatin complex. Oxaliplatin-OH can be complexed with PIMA through dicarboxylato bonds. Derivatization of one arm of PIMA with glucosamine, and complexation with oxaliplatin can lead to two isomers depending on pH.
[0037] Figures 21 A and 21B are line graphs showing the concentration-effect of different treatments on cellular viability as measured using MTS assay. Breast cancer cell line, Lewis lung cancer (Fig. 21A) and 4Tl (Fig. 21B) cell lines were used for this study. X- axis shows the equivalent concentrations of platinum. Where blank polymeric controls were used, dose of polymer used was equivalent to that used to deliver that specific dose of oxaliplatin in the complexed form. PIMA-GA-Ox refers to the isomer [PIMA-GA- Oxaliplatin (0->Pt)] formed under acidic complexation environment. The PIMA-GA- oxaliplatin curve shifted to the left, indicating that the nanoparticles are more effective than free oxaliplatin in anti-tumor efficacy.
[0038] Figures 22A-22E are line graphs (Figs. 26A and 26B) and bargraphs (Figs.
22C-22E), showing PIMA-GA-oxaliplatin nanoparticle exerts similar anti-tumor effect with reduced systemic toxicity compared to free oxaliplatin in a 4Tl breast cancer model. Line graphs show the effect of treatments on tumor volume (Fig. 22A) and body weight (Fig. 22B) over the treatment period. The animals were dosed thrice. Data shown are mean + SE, n=4-8. Bar graphs show the effect of treatment on the weight of tumor (Fig. 22C), kidney (Fig. 22D), and spleen (Fig. 22E) as a marker for nephrotoxicity and hematological toxicity (n=4- 6).
[0039] Figure 23 is a line graph showing concentration-effect of different oxaliplatin complexes on cellular viability as measured using MTS assay. [0040] Figure 24 is a line graph showing the effect of cisplatin, carboplatin and
PIMA-GA-200(A) on cell viability.
[0041] Figure 25A is a scheme showing the synthesis of cholesterol- succinic acid conjugate and the complexation of Pt to the conjugate.
[0042] Figure 25B shows a dynamic laser light scatter of liponanoparticles. The size of the liponanoparticles is less than 150 nm.
[0043] Figure 26 is a line graph showing the release kinetics of Pt from the liponanoparticle with time and the influence of pH. The rate of release is faster in an acidic pH, which facilitates selective release of active platinate in the tumor, consistent with the acidic intratumoral pH.
[0044] Figures 27A-27C are line graphs showing the effect of cisplatin- liponanoparticle on viability of 4Tl breast cancer cells. Cell viability was measured using
MTS assay. The treatment with liponanoparticles results in rapid cell kill within 12 hours as compared with either cisplatin or carboplatin (Fig. 27A). At all three time points cisplatin- liponanoparticle was found to be more effective than cisplatin. Carboplatin is the least effective of all platinates tested (Fis. 27A-27C).
[0045] Figures 28A and 28B are line graphs showing the effect of cisplatin- liponanoparticle on viability of a cisplatin-resistant hepatocellular cancer cell line (CP20) and on a Lewis lung cancer cell line (LLC). Cisplatin acts on CP20 only at the highest concentration, while the cells are susceptible to the cisplatin-liponanoparticle (Fig. 28A).
Carboplatin has no effect at this concentration range (Figs. 28A and 28B).
Cisplatinliponanoparticle exerted superior anticancer effect than free cisplatin on LLCs (Fig.
28B).
[0046] Figures 29A-29E are line graphs (Figs. 29A and 29B) and bar graphs (Figs.
29C-29E) showing the efficacy of cisplatin-liponanoparticle in a 4Tl syngeneic tumor model in vivo. Line graphs show the effect of different treatments on tumor growth (Figs. 29 A and
29C) and body weight (as a marker for systemic toxicity, Fig. 29B). Bar graphs show kidney
(Fig. 29D) and spleen (Fig. 29E) weights as markers for nephrotoxicity and hematological toxicity. As seen in the figure, cisplatin-liponanoparticle induced greater antitumor activity with reduced systemic, nephrotoxicity. DETAILED DESCRIPTION OF THE INVENTION
[0047] The invention is directed to biocompatible conjugated polymer nanoparticles including a copolymer backbone, a plurality of sidechains covalently linked to said backbone, and, a plurality of platinum compounds dissociably linked to said backbone. Generally, the platinum compounds are linked to the backbone via a linkage to the sidechains. [0048] In some embodiments, the copolymer comprises maleic acid monomers.
[0049] In a preferred embodiment, the copolymer is poly(isobutylene-α/Mnaleic acid)
(PIMA or PMA).
[0050] In some embodiments, the copolymer comprises from 2 to 100 monomeric units. In some embodiments, the copolymer comprises from 25 to 50 monomeric uints. [0051] In some embodiments, the sidechains are selected from the group consisting of polymers, monosaccharides, carboxylic acids, dicarboxylic acids, amides, and combinations thereof.
[0052] In preferred embodiments, the sidechains are polyethylene glycol (PEG). PEG sidechains can be represented by -C(O)-NH-PEG.
[0053] In some embodiments, the PEG sidechains have a molecular weight of from
100 to 5000 Dalton. In some embodiments, the PEG sidechains have a molecular weight of from 1000 to 3000 Dalton. In a preferred embodiment, the PEG sidechains have a molecular weight of about 2000 Dalton.
[0054] In some embodiments, the sidechains are monosaccharides. In a preferred embodiment, the monosaccharides are glucosamine. The monosaccharide sidechains can be represented by -C(O)-saccharide.
[0055] Any platinum compound can be used in the invention. Preferably, the platinum compound is a platinum(II) or platinum (IV) compound. In some embodiments, the platinum (II) compound is selected from the group consisting of cisplatin, oxaliplatin, carboplatin, paraplatin, sartraplatin, and combinations thereof. In a preferred embodiment, the platinum (II) compound side group is cisplatin or oxalipaltin.
[0056] In some embodiments, the platinum(II) compound is selected from the group
consisting of Pt(NHs)2, Pt(NH3)(2-methylpyridine), and
Figure imgf000011_0001
, wherein p is 0 - 3. In a preferred embodiment, the platinum (II) compound is Pt(NHs)2.
Figure imgf000012_0001
[0057] In some embodiments, the platinum (II) compound is "p , wherein p is
0 - 3.
[0058] In some embodiments, the platinum (II) compound comprises at least two nitrogen atoms, where said nitrogen atoms are directly linked to platinum. In a further embodiment, the two nitrogen atoms are linked to each other via an optionally substituted linker, e.g. acyclic or cyclic linker. A cyclic linker means a linking moiety that comprises at least one ring structure. Cyclic linkers can be aryl, heteroaryl, cyclyl or heterocyclyl.
[0059] In some embodiments, at least one nitrogen that is linked to platinum is a ring atom of a heteroaryl or a heterocyclyl. In a preferred embodiment, heteroaryl is optionally substituted pyridine, e.g., 2-methylpyridine.
[0060] In some embodiments, the plurality of sidechains corresponds to a number between 50% and 100%, inclusive, of the number of monomeric units of said polymer backbone. This means that between 50% to 100% of the monomeric units have at least one sidechain linked to the monomeric unit. The total number of sidechains can be greater than the total number of the monomeric units. For example, two sidechains can be attached to a maleic acid monomer.
[0061] In some embodiments, the plurality of sidechains corresponds to a number greater than 90% of the number of monomeric units of said polymer backbone.
[0062] In some embodiments, the plurality of platinum compounds corresponds to number between 10% and 100%, inclusive, of the number of monomeric units of said polymer backbone. Generally there is a one-to-one relationship between platinum compounds and monomeric subunits. Thus, the percent refers to the number of monomeric units that are linked to a platinum compound to the total number of monomeric units present in the polymer backbone.
[0063] In some embodiments, the plurality of platinum compounds corresponds to number between 25% and 75%, inclusive, of the number of monomeric units of said polymer backbone.
[0064] Generally from 10 to 500 ug of the platinum compound can be loaded on 1 mg of the polymer. Preferably, 50 to 250 ug, more preferably 150 to 200 ug of the platinum compound is loaded on 1 mg of the polymer. In some experiments, the inventors obtained a loading of 175 + 5 ug/mg of polymer. [0065] In some embodiments, the sidechains comprise dicarboxylic acids. In some embodiments, the dicarboxylic acids are of the formula HOOC-R-COOH, wherein R is a Cl- C6 alkyl, C2-C6 alkenyl, or C2-C6 alkynyl. In a preferred embodiment, the dicarboxylic acid is maleic acid.
[0066] In some embodiments, the copolymer comprises at least one monomer having the formula -CH(CO2H)-R-CH(C(O)R')- , wherein R is a bond, Ci-C6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom. Preferably, R is a bond.
[0067] In some embodiments, between 50% to 100%, inclusive, of the monomeric subunits in the polymeric backbone are -CH(CO2H)-R-CH(C(O)R')-, wherein R is a bond, Ci-C6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
[0068] In some embodiments, at least 90% or more of the monomeric subunits in the polymeric backbone are -CH(CO2H)-R-CH(C(O)R')-, wherein R is a bond, Ci-C6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
[0069] In some embodiments, the copolymer comprises at least one monomer having the formula -CH(CO2H)-R-CH(C(O)R' )CH2C(Me2)- or -CH(C(O)R' )-R-CH(CO2H)- CH2C(Me2)-, wherein R is a bond, C1-C6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom. Preferably, R is a bond. [0070] In some embodiments, the copolymer comprises between 50% to 100%, inclusive of monomers having the formula -CH(CO2H)-R-CH(C(O)R' )CH2C(Me2)- or - CH(C(O)R')-R-CH(CO2H)-CH2C(Me2)-, wherein R is a bond, C1-C6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
[0071] In some embodiments, the copolymer comprises at least 90% of monomers having the formula -CH(CO2H)-R-CH(C(O)R' )CH2C(Me2)- or -CH(C(O)R' )-R- CH(CO2H)-CH2C(Me2)-, wherein R is a bond, C1-C6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
[0072] In some embodiments, R' is
Figure imgf000013_0001
o Or -
NH(CH2CH2O)1nCH3, wherein m is 1-150. [0073] In some embodiments, at least one monomer of the polymer comprises two sidechains selected from the group consisting of carboxylic acid, amide, and ester. Said sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof. Preferably, said amide and ester sidechains are separated from each other by two carbon atoms. Preferably, at least one of the sidechains is not a carboxylic acid.
[0074] In some embodiments, at least one monomer of the polymer comprises two carboxylic acid sidechains. Said carboxylic acid sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof. Preferably, said carboxylic acid sidechains are separated from each other by two carbon atoms. The said carbon atoms can be linked to each other through a single bond or a double bond.
[0075] In some embodiments, at least one monomer of the polymer comprises a carboxylic acid and an amide sidechains. Said carboxylic acid sidechains and amide sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof. Preferably, said carboxylic acid and amide sidechains are separated from each other by two carbon atoms. The said carbon atoms can be linked to each other through a single bond or a double bond.
[0076] In some embodiments, at least one monomer of the polymer comprises a carboxylic acid and an ester sidechains. Said carboxylic acid sidechains and ester sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof. Preferably, said carboxylic acid and ester sidechains are separated from each other by two carbon atoms. The said carbon atoms can be linked to each other through a single bond or a double bond.
[0077] In some embodiments, at least one monomer of the polymer comprises an amide and an ester sidechains. Said amide sidechains and ester sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof. Preferably, said amide and ester sidechains are separated from each other by two carbon atoms. The said carbon atoms can be linked to each other through a single bond or a double bond.
[0078] In some embodiments, at least one monomer of the polymer comprises two amide sidechains. Said amide sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof. Preferably, said amide and ester sidechains are separated from each other by two carbon atoms. [0079] In some embodiments, at least one monomer of the polymer comprises two ester sidechains. Said ester sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7,
8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof. Preferably, said amide and ester sidechains are separated from each other by two carbon atoms.
[0080] In some embodiments, the polymer comprises two sidechains selected from the group consisting of carboxylic acid, amide, and ester. Said sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof. Preferably, said amide and ester sidechains are separated from each other by two carbon atoms. Preferably, at least least one of the sidechains is not a carboxylic acid.
[0081] In some embodiments, the polymer comprises at least two carboxylic acid sidechains. Said carboxylic acid sidechains being separated from each other by 1, 2, 3, 4, 5,
6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof. Preferably, said carboxylic acid sidechains are separated from each other by two carbon atoms.
[0082] In some embodiments, the polymer comprises a carboxylic acid and an amide sidechains. Said carboxylic acid sidechains and amide sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof. Preferably, said carboxylic acid and amide sidechains are separated from each other by two carbon atoms.
[0083] In some embodiments, the polymer comprises a carboxylic acid and an ester sidechains. Said carboxylic acid sidechains and ester sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof. Preferably, said carboxylic acid and ester sidechains are separated from each other by two carbon atoms.
[0084] In some embodiments, the polymer comprises an amide and an ester sidechains. Said amide sidechains and ester sidechains being separated from each other by
1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof.
Preferably, said amide and ester sidechains are separated from each other by two carbon atoms. The said carbon atoms can be linked to each other through a single bond or a double bond.
[0085] In some embodiments, the polymer comprises two amide sidechains. Said amide sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof. Preferably, said amide and ester sidechains are separated from each other by two carbon atoms. [0086] In some embodiments, the polymer comprises two ester sidechains. Said ester sidechains being separated from each other by 1, 2, 3, 4, 5, 6, 7, 8, 9 or ten carbon, oxygen, nitrogen, sulfur atoms, or combination thereof. Preferably, said amide and ester sidechains are separated from each other by two carbon atoms.
[0087] The nanoparticles of the invention can range in size from 25-250nm, preferably 50-200nm, more preferably 80-160nm, and most preferably 90-1 IOnm. Without wishing to be bound by theory, nanoparticles in the size range of 80-160 home preferentially into tumors resulting from the enhanced permeability and retention. See for example,
Moghimi, et al., Pharmacol Rev. 2001 Jun; 53(2):283-318.
[0088] In some embodiments, the platinum compound is dissociably linked to said backbone through at least one coordination bond. Without wishing to be bound by theory, the coordination bond is more liable and thus releases the platinum compound more easily.
[0089] In some embodiments, linking of the platinum compound to the biopolymer backbone further comprises a carboxylato bond. In some embodiments, the platinum compound is linked to the backbone through a coordination bond and a carboxylato bond.
[0090] It is to be understood that although linkage to backbone is recited, one of skill in the art understands that the platinum compound is generally linked to one or more sidechains which themselves are linked to the backbone. So any recitation of linking of a platinum compound to backbone encompasses the situations where the platinum compound is linked to a sidechain which is then linked to the backbone.
[0091] In some embodiments, the coordination bond is between platinum atom of the platinum compound and an oxygen of the sidechain. Preferably the coordination bond is between platinum and a carbonyl oxygen.
[0092] In some embodiments, the coordination bond is between platinum atom of the platinum compound and an amide oxygen of the sidechain. In some embodiments, the coordination bond is between platinum atom of the platinum compound and an ester carbonyl oxygen of the sidechain.
[0093] In some embodiments, the copolymer comprises at least one maleic acid monomer, wherein at least one carboxylic acid of said at least maleic acid is derivatized to an amide.
[0094] In some embodiments, between 50% to 100%, inclusive, of the monomeric units in the polymer backbone are maleic acid monomer and wherein at least one carboxylic acid of said maleic acid monomer is derivatized to an amide. [0095] In some embodiments, at least 90% of the monomeric units in the polymer backbone are maleic acid monomer and wherein at least one carboxylic acid of said maleic acid monomer is derivatized to an amide.
[0096] The loading of platinum compound onto the polymer can be represented by as percent mg of platinum compound per mg of polymer. For example, a maximum of 0.375 mg of cisplatin can be loaded onto the PIMA-GA polymer therefore a loading of 37.5% represents the maximum loading for that particular polymer. The loading can range from about 1% to the theoretical total loading for a polymer.
[0097] In some embodiments, the platinum compound loading is from l%-37.5%.
The percent loading represent mg of platinum compound linked with per mg of polymer. [0098] In some embodiments, the platinum compound loading is from l%-6%. In some embodiments, the Pt(II) compound loading is from 0.01% to 1%. [0099] Another aspect of the invention is directed to biocompatible conjugated polymer nanoparticles including a poly(isobutylene-α/Mnaleic acid) backbone. The backbone consists of from 25 to 50 monomers. Also included are a plurality of PEG sidechains covalently linked to said backbone. The PEG sidechains have a molecular weight of from 1000 to 3000 Dalton. The PEG sidechains number between 50% and 100%, inclusive, of the number of monomeric units of the polymer backbone. Also included are a plurality of cisplatin side groups dissociably linked to the backbone. The cisplatin side groups number between 25% and 75%, inclusive, of the number of monomeric units of the polymer backbone.
[00100] Yet another aspect of the invention is directed to biocompatible conjugated polymer nanoparticles including a poly(isobutylene-α/Mnaleic acid) backbone. The backbone consist of 40 monomers. Also included are a plurality of PEG sidechains covalently linked to the backbone. The PEG sidechains have a molecular weight of 2000 Dalton. The PEG sidechains number greater than 90% of monomeric units of said polymer backbone. Also included are a plurality of cisplatin side groups dissociably linked to the backbone. The cisplatin side groups number between 25% and 75%, inclusive, of the number of monomeric units of the polymer backbone.
[00101] Still another aspect of the invention is directed to biocompatible conjugated polymer nanoparticles including a poly(isobutylene-α/Mnaleic acid) backbone. The backbone consists of from 25 to 50 monomers. Also included are a plurality of glucosamine sidechains covalently linked to said backbone. The glucosamine sidechains number between 50% and 100%, inclusive, of monomeric units of said polymer backbone. Also included are a plurality of cisplatin side groups dissociably linked to the backbone. The cisplatin side groups number between 25% and 75%, inclusive, of the number of monomeric units of the polymer backbone.
[00102] Another aspect of the invention is directed to biocompatible conjugated polymer nanoparticles including a poly(isobutylene-α/Mnaleic acid) backbone. The backbone consists of from 25 to 50 monomers. Also included are a plurality of glucosamine sidechains covalently linked to said backbone. The glucosamine sidechains number greater than 90% of monomeric units of said polymer backbone. Also included are a plurality of cisplatin side groups dissociably linked to the backbone. The cisplatin side groups number between 25% and 75%, inclusive, of the number of monomeric units of the polymer backbone.
[00103] Yet another aspect of the invention is directed to carboxylic acid-platinum compound complex conjugated nanoparticles including a carboxylic acid-platinum compound complex and a plurality of lipid-polymer chains. The carboxylic acid portion of said carboxylic acid-platinum complex is covalently bound to said lipid-polymer chains.
[00104] In a preferred embodiment, the carboxylic acid is maleic acid. In some embodiments, the polymer is PEG.
[00105] In certain embodiments, the platinum compound loading is from l%-37.5%.
In certain embodiments, the platinum compound loading is from l%-6%.
[00106] The platinum compound can be Pt(II) compound or a Pt(IV) compound. In some embodiments, the Pt(II) compound is selected from the group consisting of cisplatin, oxaliplatin, carboplatin, paraplatin, sartraplatin, and combinations thereof. In a preferred embodiment, the Pt(II) compound is cisplatin.
[00107] Another aspect of the invention is directed to a vesicle, micelle, or liposome compound comprising a plurality of nanoparticles of claim as described herein.
[00108] Still another aspect of the invention is directed to a pharmaceutical composition including any of the nanoparticles or compounds described herein and a pharmaceutically acceptable carrier.
[00109] Yet another aspect of the invention is directed to a method of treating cancer or metastasis. The method includes selecting a subject in need of treatment for cancer or metastasis and administering to the subject an effective amount of any of the nanoparticles, compounds, or compositions described herein.
[00110] In some embodiments, the cancer or metastasis is selected from the group consisting of platinum susceptible or resistant tumors including breast, head and neck, ovarian, testicular, pancreatic, oral-esophageal, gastrointestinal, liver, gall bladder, lung, melanoma, skin cancer, sarcomas, blood cancers, brain tumors including glioblastomas, and tumors of neuroectodermal origin.
[00111] In yet another aspect, the invention provide for methods of formulating platinum compound polymer nanoparticles, method comprising conjugation of platinum compound with a biocompatible polymer or biocompatible copolymer. Without wishing to be bound by theory, conjugation of platinum compound with the biocompatible polymer at acidic pH results in nanoparticles that are more active in vivo than when conjugation is done at basic pH.
[00112] Accordingly, in some embodiments, conjugation is done at pH below 7, preferably a pH between 1 and 6.9. In some more preferred embodiments, conjugation is carried out at a pH of 6.5.
[00113] The inventors have observed that conjugation under basic conditions favor the formation of an isomeric PIMA-GA_Cisplantin complex with a monocarboxylato and a more stable Pt<->N coordinate bond. In contrast, complexxation PIMA-GA and cisplatin in an acidic pH generates the isomeric state characterized by the monocarboxylato bond and Pt<-
>O coordinate bond. Thus, conjugations conditions that lead to the formation of a Pt<->0 coordinate bond over a Pt<->N coordinate bond are preferred for conjugation.
[00114] Generally an excess of the Pt(II) compound to the polymer is used. In some embodiments, 5-25 mole access of Pt(II) compound to polymer is used. Preferably 10-20 mole access of platinum(II) compound to polymer is used . In one preferred 15 mole access of Pt(II) compound to polymer is used.
[00115] In yet another aspect, the invention provides a dicarbonyl molecule linked to a lipid molecule. Such a compound can be represented by the structure lipid-linker- dicarbnoyl. These molecules can be used to complex platinum compounds such as cisplatin, oxaliplatin or other platinates and platinum compounds described herein through carboxylato- linkage and/or coordination bonds. These can then be mixed with appropriate lipids/phospholipids to nanoparticles of less than 150 nm, which release Pt in a pH-dependent manner. Once formulated these nanoparticles exhibit improved efficacy and toxicity profile as compared with carboplatin and cisplatin, and are active in a cisplatin-resistant cancer.
[00116] These nanoparticles can be formulated to comprise pharmaceutically active agents for delivery.
[00117] The term "Lipid" is used in the conventional sense to refer to molecules that are soluble to a greater or lesser degree in organic solvents, like alcohols, and relatively insoluble in aqueous media. Thus, the term "lipid" includes compounds of varying chain length, from as short as about 2 carbon atoms to as long as about 28 carbon atoms. Additionally, the compounds may be saturated or unsaturated, and in the form of straight- or branched-chains or in the form of unfused or fused ring structures. Exemplary lipids include, but are not limited to, fats, waxes, sterols, steroids, bile acids, fat-soluble vitamins (such as A, D, E, and K), monoglycerides, diglycerides, phospholipids, glycolipids, sulpholipids, aminolipids, chromolipids (lipochromes), glycerophospholipids, sphingolipids, prenol lipids, saccharolipids, polyketides, and fatty acids. In some embodiments, the lipid is cholesterol or distearoylphosphatidylethanolamine.
[00118] Generally any molecule that has two carbonyl groups can be used. In some embodiments, the dicarbonyl molecule is a dicarboxylic acid, or a keto-carboxylic acid. In some preferred embodiments, the dicarbonyl molecule is succinic acid. [00119] In some embodiments, the dicarbonyl molecule is ROC(O)-R-C(O)-, wherein
R is Ci-C6 alkylene, where the alkylene can comprise one or more double or triple bonds and/or the backbone of the alkylene can be interrupted with one or more of O, S, S(O), SO2, NH, C(O); and R' is H, alkyl, alkenyl, alkynyl, aryl, heteroaryl, acylcy, heterocyclyl, each of which can be optionally substituted.. Preferably R is CH2, -CH2CH2-, -CH2CH2-CH2- or - CH=CH-. Preferably R' is H.
[00120] The dicarbonyl molecule can be linked with the lipid molecule directly or through a linker molecule. The term "linker" means an organic moiety that connects two parts of a compound. Linkers typically comprise a direct bond or an atom such as oxygen or sulfur, a unit such as NH, C(O), C(O)NH, SO, SO2, SO2NH or a chain of atoms, such as substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, heterocyclylalkyl, heterocyclylalkenyl, heterocyclylalkynyl, aryl, heteroaryl, heterocyclyl, cycloalkyl, cycloalkenyl, alkylarylalkyl, alkylarylalkenyl, alkylarylalkynyl, alkenylarylalkyl, alkenylarylalkenyl, alkenylarylalkynyl, alkynylarylalkyl, alkynylarylalkenyl, alkynylarylalkynyl, alkylheteroarylalkyl, alkylheteroarylalkenyl, alkylheteroarylalkynyl, alkenylheteroarylalkyl, alkenylheteroarylalkenyl, alkenylhetero arylalkynyl, alkynylheteroarylalkyl, alkynylheteroarylalkenyl, alkynylheteroarylalkynyl, alkylheterocyclylalkyl, alkylheterocyclylalkenyl, alkylhererocyclylalkynyl, alkenylheterocyclylalkyl, alkenylheterocyclylalkenyl, alkenylheterocyclylalkynyl, alkynylheterocyclylalkyl, alkynylheterocyclylalkenyl, alkynylheterocyclylalkynyl, alkylaryl, alkenylaryl, alkynylaryl, alkylheteroaryl, alkenylheteroaryl, alkynylhereroaryl, where one or more methylenes can be interrupted or terminated by O, S, S(O), SO2, NH, C(O). It is to be understood that the diacarbonyl molecule and/or the lipid can be modified to comprise functional groups for linking to each other or to the linker.
[00121] In some embodiments, linker is a diamine such as ethylene diamine. In some embodiments, linker is PEG-NH2.
[00122] In one preferred embodiment, linker is -NHCH2CH2C(O)-. In another preferred embodiment, linker is -CH2CH2NHC(O)-[OCH2CH2]Z-NH-, where z is 1-50.
Preferably z is 45.
[00123] In some embodiments, the lipid-dicarbonyl compound is as shown in Figs. 10
(compound 2) and 25 (compound 5).
[00124] In another aspect, the invention provide a biocompatible polymer comprising at least one monomer having the formula -CH(CO2H)-R-CH(C(O)R')- , wherein R is a bond,
Ci-C6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom. Preferably, R is a bond.
[00125] In some embodiments, the polymer comprises from 2 to 100 monomeric units having the formula -CH(CO2H)-R-CH(C(O)R')-, wherein R is a bond, Ci-C6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
[00126] In some embodiments, the polymer comprises from 25 to 50 monomeric units having the formula -CH(CO2H)-R-CH(C(O)R')-, wherein R is a bond, Ci-C6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
[00127] In some embodiments, between 50% to 100%, inclusive, of the monomeric subunits in the polymeric backbone are -CH(CO2H)-R-CH(C(O)R')-, wherein R is a bond,
Ci-C6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
[00128] In some embodiments, at least 90% or more of the monomeric subunits in the polymeric backbone are -CH(CO2H)-R-CH(C(O)R')-, wherein R is a bond, Ci-C6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
[00129] In some embodiments, the copolymer comprises at least one monomer having the formula -CH(CO2H)-R-CH(C(O)R' )CH2C(Me2)- or -CH(C(O)R' )-R-CH(CO2H)- CH2C(Me2)-, wherein R is a bond, Ci-C6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom. Preferably, R is a bond. [00130] In some embodiments, the copolymer comprises between 50% to 100%, inclusive of monomers having the formula -CH(CO2H)-R-CH(C(O)R' )CH2C(Me2)- or - CH(C(O)R')-R-CH(CO2H)-CH2C(Me2)-, wherein R is a bond, Ci-C6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
[00131] In some embodiments, the copolymer comprises at least 90% of monomers having the formula -CH(CO2H)-R-CH(C(O)R' )CH2C(Me2)- or -CH(C(O)R' )-R- CH(CO2H)-CH2C(Me2)-, wherein R is a bond, Ci-C6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom.
[00132] In some embodiments, R' is
Figure imgf000022_0001
"° or -
NH(CH2CH2O)1nCH3, wherein m is 1-150.
[00133] These polymers can be used for formulating nanoparticles and gels which can be used for drug delivery. Thus, the invention also provides nanoparticles comprising a polymer described herein and one or more bioactive active agent ("bioactive agent"). [00134] Compositions described herein can be used in methods for sustained release of bioactive active agents. In one embodiment, the method comprising: (a) providing or administering to a subject a composition described herein, wherein the composition contains the bioactive agent. As used herein, "bioactive agents" refer to naturally occurring biological materials, for example, extracellular matrix materials such as fibronectin, vitronection, and laminin; cytokins; and growth factors and differentiation factors. "Bioactive agents" also refer to artificially synthesized materials, molecules or compounds that have a biological effect on a biological cell, tissue or organ.
[00135] Suitable growth factors and cytokines include, but are not limited, to stem cell factor (SCF), granulocyte-colony stimulating factor (G-CSF), granulocyte-macrophage stimulating factor (GM-CSF), stromal cell-derived factor- 1, steel factor, VEGF, TGFβ, platelet derived growth factor (PDGF), angiopoeitins (Ang), epidermal growth factor (EGF), bFGF, HNF, NGF, bone morphogenic protein (BMP), fibroblast growth factor (FGF), hepatocye growth factor, insulin-like growth factor (IGF-I), interleukin (IL)-3, IL-lα, IL-lβ, IL-6, IL-7, IL-8, IL-Il, and IL-13, colony- stimulating factors, thrombopoietin, erythropoietin, fit3-ligand, and tumor necrosis factor α (TNFg). Other examples are described in Dijke et al., "Growth Factors for Wound Healing", Bio/Technology, 7:793-798 (1989); Mulder GD, Haberer PA, Jeter KF, eds. Clinicians' Pocket Guide to Chronic Wound Repair. 4th ed. Springhouse, PA: Springhouse Corporation; 1998:85; Ziegler T.R., Pierce, G.F., and Herndon, D. N., 1997, International Symposium on Growth Factors and Wound Healing: Basic Science & Potential Clinical Applications (Boston, 1995, Serono Symposia USA), Publisher: Springer Verlag.
[00136] In some embodiments, suitable bioactive agents include but not limited to therapeutic agents. As used herein, the term "therapeutic agent" refers to a substance used in the diagnosis, treatment, or prevention of a disease. Any therapeutic agent known to those of ordinary skill in the art to be of benefit in the diagnosis, treatment or prevention of a disease is contemplated as a therapeutic agent in the context of the present invention. Therapeutic agents include pharmaceutically active compounds, hormones, growth factors, enzymes, DNA, plasmid DNA, RNA, siRNA, viruses, proteins, lipids, pro-inflammatory molecules, antibodies, antibiotics, anti-inflammatory agents, anti-sense nucleotides and transforming nucleic acids or combinations thereof. Any of the therapeutic agents may be combined to the extent such combination is biologically compatible.
[00137] Exemplary therapeutic agents include, but are not limited to, those found in
Harrison's Principles of Internal Medicine, 13th Edition, Eds. T.R. Harrison et al. McGraw- Hill N.Y., NY; Physicians Desk Reference, 50th Edition, 1997, Oradell New Jersey, Medical Economics Co.; Pharmacological Basis of Therapeutics, 8th Edition, Goodman and Gilman, 1990; United States Pharmacopeia, The National Formulary, USP XII NF XVII, 1990; current edition of Goodman and Oilman's The Pharmacological Basis of Therapeutics; and current edition of The Merck Index, the complete contents of all of which are incorporated herein by reference.
[00138] Examples of therapeutic agents which may be incorporated in the composition, include but are not limited to, narcotic analgesic drugs; salts of gold; corticosteroids; hormones; antimalarial drugs; indole derivatives; pharmaceuticals for arthritis treatment; antibiotics, including Tetracyclines, Penicillin, Streptomycin and Aureomycin; antihelmintic and canine distemper drugs, applied to domestic animals and large cattle, such, as, for example, phenothiazine; drugs based on sulfur, such, as sulfioxazole; antitumor drugs; pharmaceuticals supervising addictions, such as agents controlling alcohol addiction and agents controlling tobacco addiction; antagonists of drug addiction, such, as methadone; weightcontrolling drugs; thyroid gland controlling drugs; analgesics; drugs controlling fertilization or contraception hormones; amphetamines; antihypertensive drugs; antiinflammatories agents; antitussives; sedatives; neuromuscular relaxants; antiepileptic drugs; antidepressants; antidisrhythmic drugs; vasodilating drugs; antihypertensive diuretics; antidiabetic agents; anticoagulants; antituberculous agents; antipsyhotic agents; hormones and peptides. It is understood that above list is not full and simply represents the wide diversification of therapeutic agents that may be included in the compositions. In some embodiments, therapeutic agent is Mitoxantrone, protein (e.g. VEGF) or plasmid DNA.
[00139] The amount of therapeutic agent distributed in a composition depends on various factors including, for example, specific agent; function which it should carry out; required period of time for release of a the agent; quantity to be administered. Generally, dosage of a therapeutic agent i.e. amount of therapeutic agent in composition, is selected from the range about from 0.001% (w/w) up to 95% (w/w), preferably, from about 5% (w/w) to about 75% (w/w), and, most preferably, from about 10% (w/w) to about 60% (w/w).
[00140] Cisplatin [cis-dichlorodiammineplatinum(II)] (CDDP) has emerged as an important class of antitumor agents, and is widely used for the treatment of many malignancies including testicular , ovarian , cervical, head and neck, and non-small cell lung cancer (Jamieson, et al, Chem. Rev. (1999), 99(9): 2467-2498). It was also shown to be active in triple negative breast cancer (Leong, et al., J. Clin. Invest. (2007), 117(5): 1370-80). Its use is however dose-limited mainly because of nephrotoxicity or toxicity to the kidney (Madias, NE and Harrington, JT, Am. J. (1978), 65(2): 307-14). To address this limitation, two directions of research has evolved, the first focused on the synthesis of platinum analogues, the second is to engineer novel nanodelivery systems as a mean to target the drug directly to the tumor site. It is now well established that nanoparticles in the size range 80- 120 nm home preferentially into tumors resulting from the enhanced permeability and retention (EPR) effect (Moghimi, et al., Pharmacol. Rev. (2001), 53(2): 283-318). This can reduce systemic side effects and exhibit increased intratumoral delivery. A nanoliposomal formulation of cisplatin was found to deliver 50-200 times more drug to the tumor as compared to administration of free cisplatin (Harrington, et al. Ann. Oncol. (2001) 12: 493- 496). Although there was minimal toxicity with the nanoliposomal formulation, it had only modest antitumor activity as compared to cisplatin; reflecting the challenges of not only delivering platinum in a relatively inactive form, but the subsequent need to achieve significant release and activation within the tumor. A second strategy of encapsulating cisplatin into polymeric systems has been a challenge as a result of its insolubility in organic solvents and partial solubility in water, which resulted in poor loading or inability to maintain sustained release. This has required the development of platinum (IV) prodrugs that can be modified to increase hydrophobicity, and increase loading in polylactide-polyglycolide copolymer nanoparticles (Dhar et al., 2009). Alternatively, cisplatin was conjugated to N-(2- hydroxypropyl) methacrylamide (HPMA) through peptidyl side-chains, and were shown to be biologically active(Lin X, Zhang Q, Rice JR, Stewart DR, Nowotnik DP, Ho well SB. Improved targeting of platinum chemotherapeutics. The antitumour activity of the HPMA copolymer platinum agent AP5280 in murine tumour models. Eur J Cancer. 2004 Jan;40(2):291-7). However, such approaches require processing through enzymatic cleavage or intracellular reduction for activation of the drug. Similarly, a PAMAM dendrimers- platinum complex, which increased drug loading, was found to be 200-550-fold less toxic than cisplatin as a result of strong bonds that are formed between the polymer and Pt (Haxton KJ, Burt HM. Polymeric drug delivery of platinum-based anticancer agents. J Pharm Sci. 2009 Jul;98(7):2299-316).
[00141] To engineer a nanoformulation of cisplatin that is facile but overcomes the challenges associated with current approaches, the inventors integrated the existing information on the biotransformation of cisplatin and understanding of the structure activity relationship that has emerged through the development of cisplatin analogues. Cisplatin gets activated through intracellular aquation of one of the two chloride leaving groups to form [Pt(NH3)2Cl(OH2)]+ and [Pt(NH3)2(OH2)]2+, following which the Pt forms covalent bonds to the N7 position of purine bases to form intrastrand and interstrand crosslinks (Huifang Huang, Leiming Zhu, Brian R. Reid, Gary P. Drobny, Paul B. Hopkins. Solution Structure of a Cisplatin-Induced DNA Interstrand Cross-Link. Science 1995: 270. 1842 - 1845). In comparison, carboplatin and oxaloplatin, have a cyclobutane-l,l-decarboxylate and an oxalate respectively as the leaving groups, which chelate the platinum more strongly thus confering greater stability to the leaving group-PT complex and as a result exhibit fewer side effects than cisplatin but also lower efficacy than cisplatin (Richard J. Knox, Frank Friedlos, David A. Lydall and John J. Roberts Mechanism of Cytotoxicity of Anticancer Platinum Drugs: Evidence That ds-Diamminedichloroplatinum(II) and ds-Diammine-(l,l- cyclobutanedicarboxylato) platinum(II) Differ Only in the Kinetics of Their Interaction with DNA. Cancer Research 46, 1972-1979, April 1, 1986; and Ronald S. Go, Alex A. Adjei. Review of the Comparative Pharmacology and Clinical Activity of Cisplatin and Carboplatin. Journal of Clinical Oncology, VoI 17, Issue 1 (January), 1999: 409). The inventors selected a 40-mer Poly(isobutylene-alt-maleic acid) (PIMA or PMA) as the polymer because each monomer exhibits dicarboxylato groups that can be complexed with cisplatin(OH)2. allowed the loading of a cisplatin molecule. Furthermore, hydrogenation of maleic acid generates succinic acid, which is a component of the Krebs cycle. Poly(isobutylene-alt-maleic acid) 2 was synthesized from Poly(isobutylene-alt-maleic anhydride) 1 by reaction with water in DMF in one step as shown in Figure 1. Further conjugation of cisplatin to Poly(isobutylene- alt-maleic acid) (PIMA) 2 was achieved by stirring hydrated cisplatin for 48 hours gave PMA-Cisplatin 6. The non-conjugated cisplatin was removed by dialysis and amount of loading was determined by NMR and spectrophotometry. Interestingly, the complexation process led to the generation of nanoparticles through a self-assembly process, with the size defined by the number of cisplatin molecules loaded per polymer. Measurement using dynamic laser light scatter revealed that saturating all the complexation sites with cisplatin resulted in a gel formation while loading 15 molecules of cisplatin per polymer resulted in a nanoparticle in the size range of 100 nm. This was validated by transmission electron microscopy (data not shown).
[00142] Cisplatin is a first line therapy for lung cancer, and as a result the inventors studied the effect of PMA-Cisplatin on the viability of Lewis lung cancer cells. Treatment with both cisplatin and PMA-cisplatin induced identical cell kill (Fig. 1C). However, PMA also induced tumor cell death. The inventors discovered that this can be overcome through derivatization of PMA. The inventors derivatized the polymer with ethylene diamine under basic conditions (Fig. 2). Interestingly, although the derivatization failed to remove the cytotoxicity of PMA, it increased the cytotoxicity of the PMA-cisplatin complex. This could potentially arise from the fact that the leaving group is less tightly bound as compared to underivatized PMA. Indeed, such an effect has been seen in the case of carboplatin, which has a lower rate constant for aquation than cisplatin, and as a result is also less cytotoxic. The native PMA-cisplatin may be tightly held as compared with PMA-EDA because of strong chelation formed by two carboxy groups. To further make the polymer more biocompatible the inventors modified the polymer with glucosamine (GA). PMA-GA-cisplatin was synthesized starting with PMA (1) by reacting with Glucosamine and then with aqueous cisplatin (Figs. 3 and HB). All the carrier polymers synthesized were platinated in aqueous phase at room temperature 250C for 2 days, with aquated cisplatin as platination agent, giving conjugates. At different time points, the inventors aliquoted out a small fraction and quantified the total loading of cisplatin on the polymer. The inventors observed a loading efficiency of -60% by 5 hours of complexation, -80% by 30 hours and 100% by 48 hours of platination. The total drug loaded was 6mg/15 mg of polymer. Aquation of cisplatin was achieved using equimolar cisplatin and AgNO3 under dark for 48h. All carriers were routinely fractionated by dialysis and isolated by freeze-drying for spectroscopic characterization. Using DBU resulted in the synthesis of the glucosamine-PMA conjugate as seen in the distinct polymer and sugar peaks in the NMR results that matches with the predicted NMR values. However, treatments with bases, triethylamine or DIPEA, failed to give the predicted product, but the NMR traces provided valuable clue to defining the final functional product.
[00143] Complexation of cisplatin with PMA-GA resulted in the self assembly of the complex into nanoparticles. In certain cases, passing the nanoparticles through a 0.22 micron filter resulted in the generation of nanoparticles that were in the sub lOOnm range, which is critical for the particles to home in specifically to the tumor using the EPR effect. Interestingly, cell viability studies revealed that the PMA-GA derivative was devoid of any inherent toxicity to the cells. In contrast, it retained the efficacy of the aquated Cisplatin (Fig. 4B). Furthermore, derivatization of PMA with polyethylene glycol also removed the inherent toxicity associated with PMA. Additionally, the same goal can be achieved by conjugating maleic acid to a polymeric backbone that is biocompatible.
[00144] The increased efficacy with the derivatized chelated polymers as compared with the native polymer indicates that the monocarboxylato-chelated release drug much easily and showed superior activity over dicarboxylato-chelated (6). The inventors discovered that the polymeric monocarboxylato-chelated platinum compounds represent a sizeable advantage over the conjugates in which the metal is bound via dicarboxylic acid. Smooth hydrolytic drug liberation from the carrier in the monocarboxylato-chelated derivatized PMA conjugates, as compared to the more retarded hydrolytic fission of the dicarboxylato-chelated in PMA, may explain this enormous difference in cell killing performance. To study this further, the inventors incubated the drug-polymer conjugate with Lewis Lung Cancer cell lysate in a dialysis chamber, and quantified the release of free drug using a calorimetric assay. The inventors obtained a rapid and sustained release of the active agent (Fig. 4A). It should be noted that the same formulation had been dialyzed in water for 48 hours to remove any free cisplatin and the inventors had obtained 100% loading efficiency, suggesting that the active agent is not released in neutral conditions but is rapidly released in the presence of tumor cell lysate. [00145] The compositions described herein can be formulated into gels and used for sustained released delivery of bioactive agents at specific locations in a subject. For example, the composition can be used for sustained release delivery of platinum compounds at site of tumors. In some embodiments, the composition is used for sustain delivery of a platinum compound after a tumor has been removed.
Pharmaceutical Compositions
[00146] For administration to a subject, the polymer linked platinum compound can be provided in pharmaceutically acceptable compositions. These pharmaceutically acceptable compositions comprise a therapeutically-effective amount of one or more of the platinum compounds described herein, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents. As described in detail below, the pharmaceutical compositions of the present invention can be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), lozenges, dragees, capsules, pills, tablets (e.g., those targeted for buccal, sublingual, and systemic absorption), boluses, powders, granules, pastes for application to the tongue; (2) parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; (3) topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin; (4) intravaginally or intrarectally, for example, as a pessary, cream or foam; (5) sublingually; (6) ocularly; (7) transdermally; (8) transmucosally; or (9) nasally. Additionally, compounds can be implanted into a patient or injected using a drug delivery system. See, for example, Urquhart, et al., Ann. Rev. Pharmacol. Toxicol. 24: 199-236 (1984); Lewis, ed. "Controlled Release of Pesticides and Pharmaceuticals" (Plenum Press, New York, 1981); U.S. Pat. No. 3,773,919; and U.S. Pat. No. 35 3,270,960. [00147] As used here, the term "pharmaceutically acceptable" refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. [00148] As used here, the term "pharmaceutically- acceptable carrier" means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid), or solvent encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically- acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, methylcellulose, ethyl cellulose, microcrystalline cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) lubricating agents, such as magnesium stearate, sodium lauryl sulfate and talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol (PEG); (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) pH buffered solutions; (21) polyesters, polycarbonates and/or polyanhydrides; (22) bulking agents, such as polypeptides and amino acids (23) serum component, such as serum albumin, HDL and LDL; (22) C2-C12 alchols, such as ethanol; and (23) other non-toxic compatible substances employed in pharmaceutical formulations. Wetting agents, coloring agents, release agents, coating agents, sweetening agents, flavoring agents, perfuming agents, preservative and antioxidants can also be present in the formulation. The terms such as "excipient", "carrier", "pharmaceutically acceptable carrier" or the like are used interchangeably herein.
[00149] The phrase "therapeutically-effective amount" as used herein means that amount of a compound, material, or composition comprising a compound of the present invention which is effective for producing some desired therapeutic effect in at least a sub- population of cells in an animal at a reasonable benefit/risk ratio applicable to any medical treatment. For example, an amount of a compound administered to a subject that is sufficient to produce a statistically significant, measurable change in at least one symptom of cancer or metastasis.
[00150] Determination of a therapeutically effective amount is well within the capability of those skilled in the art. Generally, a therapeutically effective amount can vary with the subject's history, age, condition, sex, as well as the severity and type of the medical condition in the subject, and administration of other pharmaceutically active agents. [00151] As used herein, the term "administer" refers to the placement of a composition into a subject by a method or route which results in at least partial localization of the composition at a desired site such that desired effect is produced. A compound or composition described herein can be administered by any appropriate route known in the art including, but not limited to, oral or parenteral routes, including intravenous, intramuscular, subcutaneous, transdermal, airway (aerosol), pulmonary, nasal, rectal, and topical (including buccal and sublingual) administration.
[00152] Exemplary modes of administration include, but are not limited to, injection, infusion, instillation, inhalation, or ingestion. "Injection" includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intraventricular, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, sub capsular, subarachnoid, intraspinal, intracerebro spinal, and intrasternal injection and infusion. In preferred embodiments, the compositions are administered by intravenous infusion or injection.
[00153] By "treatment", "prevention" or "amelioration" of a disease or disorder is meant delaying or preventing the onset of such a disease or disorder, reversing, alleviating, ameliorating, inhibiting, slowing down or stopping the progression, aggravation or deterioration the progression or severity of a condition associated with such a disease or disorder. In one embodiment, at least one symptom of a disease or disorder is alleviated by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, or at least 50%. [00154] As used herein, a "subject" means a human or animal. Usually the animal is a vertebrate such as a primate, rodent, domestic animal or game animal. Primates include chimpanzees, cynomologous monkeys, spider monkeys, and macaques, e.g., Rhesus. Rodents include mice, rats, woodchucks, ferrets, rabbits and hamsters. Domestic and game animals include cows, horses, pigs, deer, bison, buffalo, feline species, e.g., domestic cat, canine species, e.g., dog, fox, wolf, avian species, e.g., chicken, emu, ostrich, and fish, e.g., trout, catfish and salmon. Patient or subject includes any subset of the foregoing, e.g., all of the above, but excluding one or more groups or species such as humans, primates or rodents. In certain embodiments, the subject is a mammal, e.g., a primate, e.g., a human. The terms, "patient" and "subject" are used interchangeably herein. The terms, "patient" and "subject" are used interchangeably herein.
[00155] Preferably, the subject is a mammal. The mammal can be a human, non- human primate, mouse, rat, dog, cat, horse, or cow, but are not limited to these examples. Mammals other than humans can be advantageously used as subjects that represent animal models of disorders associated with inflammation.
[00156] In addition, the methods described herein can be used to treat domesticated animals and/or pets. A subject can be male or female. A subject can be one who has been previously diagnosed with or identified as suffering from or having a disorder a cancer or metastasis, but need not have already undergone treatment.
[00157] As used herein, the term "cancer" includes, but is not limited to, solid tumors and blood born tumors. The term cancer refers to disease of skin, tissues, organs, bone, cartilage, blood and vessels. The term "cancer" further encompasses primary and metastatic cancers. Examples of cancers that can be treated with the compounds of the invention include, but are not limited to, carcinoma, including that of the bladder, breast, colon, kidney, lung, ovary, pancreas, stomach, cervix, thyroid, and skin, including squamous cell carcinoma; hematopoietic tumors of lymphoid lineage, including, but not limited to, leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B -cell lymphoma, T-cell lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, hairy cell lymphoma, and Burketts lymphoma; hematopoietic tumors of myeloid lineage including, but not limited to, acute and chronic myelogenous leukemias and promyelocytic leukemia; tumors of mesenchymal origin including, but not limited to, fibrosarcoma, rhabdomyosarcoma, and osteosarcoma; other tumors including melanoma, seminoma, tetratocarcinoma, neuroblastoma, and glioma; tumors of the central and peripheral nervous system including, but not limited to, astrocytoma, neuroblastoma, glioma, and schwannomas; and other tumors including, but not limited to, xenoderma, pigmentosum, keratoactanthoma, thyroid follicular cancer, and teratocarcinoma. The compounds of the invention are useful for treating patients who have been previously treated for cancer, as well as those who have not previously been treated for cancer. Indeed, the methods and compositions of this invention can be used in first-line and second- line cancer treatments.
[00158] The compounds of the invention are also useful in combination with known anti-cancer treatments, including radiation. The methods of the invention are especially useful in combination with anti-cancer treatments that involve administering a second drug that acts in a different phase of the cell cycle, e.g., S phase, than the epothilones of Formula (Ia) or (Ib), which exert their effects at the G2-M phase.
Definitions [00159] Unless stated otherwise, or implicit from context, the following terms and phrases include the meanings provided below. Unless explicitly stated otherwise, or apparent from context, the terms and phrases below do not exclude the meaning that the term or phrase has acquired in the art to which it pertains. The definitions are provided to aid in describing particular embodiments, and are not intended to limit the claimed invention, because the scope of the invention is limited only by the claims. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular. [00160] As used herein the term "comprising" or "comprises" is used in reference to compositions, methods, and respective component(s) thereof, that are essential to the invention, yet open to the inclusion of unspecified elements, whether essential or not. [00161] Other than in the operating examples, or where otherwise indicated, all numbers expressing quantities of ingredients or reaction conditions used herein should be understood as modified in all instances by the term "about." The term "about" when used in connection with percentages may mean +1%.
[00162] The singular terms "a," "an," and "the" include plural referents unless context clearly indicates otherwise. Similarly, the word "or" is intended to include "and" unless the context clearly indicates otherwise.
[00163] Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of this disclosure, suitable methods and materials are described below. The term "comprises" means "includes." The abbreviation, "e.g." is derived from the Latin exempli gratia, and is used herein to indicate a non-limiting example. Thus, the abbreviation "e.g." is synonymous with the term "for example." [00164] The term "alkyl" refers to saturated non-aromatic hydrocarbon chains that may be a straight chain or branched chain, containing the indicated number of carbon atoms (these include without limitation methyl, ethyl, propyl, iso-propyl, butyl, 2-methyl-ethyl, t-butyl, allyl, or propargyl), which may be optionally inserted with N, O, or S. For example, Ci-C6 indicates that the group may have from 1 to 6 (inclusive) carbon atoms in it. [00165] The term "alkenyl" refers to an alkyl that comprises at least one double bond.
Exemplary alkenyl groups include, but are not limited to, for example, ethenyl, propenyl, butenyl, l-methyl-2-buten-l-yl and the like.
[00166] The term "alkynyl" refers to an alkyl that comprises at least one triple bond.
[00167] The term "aryl" refers to monocyclic, bicyclic, or tricyclic aromatic ring system wherein 0, 1, 2, 3, or 4 atoms of each ring may be substituted by a substituent. Examplary aryl groups include, but are not limited to, benzyl, phenyl, naphthyl, anthracenyl, azulenyl, fluorenyl, indanyl, indenyl, naphthyl, phenyl, tetrahydronaphthyl, and the like. [00168] The term "cyclyl" or "cycloalkyl" refers to saturated and partially unsaturated cyclic hydrocarbon groups having 3 to 12 carbons, for example, 3 to 8 carbons, and, for example, 3 to 6 carbons, wherein the cycloalkyl group additionally may be optionally substituted. Exemplary cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, cyclooctyl, and the like.
[00169] The term "heteroaryl" refers to an aromatic 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2, 3, or 4 atoms of each ring may be substituted by a substituent. Examplary heteroaryl groups include, but are not limited to, pyridyl, furyl or furanyl, imidazolyl, benzimidazolyl, pyrimidinyl, thiophenyl or thienyl, pyridazinyl, pyrazinyl, quinolinyl, indolyl, thiazolyl, naphthyridinyl, and the like. [00170] The term "heterocyclyl" refers to a nonaromatic 5-8 membered monocyclic, 8-
12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2 or 3 atoms of each ring may be substituted by a substituent. Examplary heterocyclyl groups include, but are not limited to piperazinyl, pyrrolidinyl, dioxanyl, morpholinyl, tetrahydrofuranyl, and the like. [00171] The term "optionally substituted" means that the specified group or moiety, such as an alkyl group, alkenyl group, and the like, is unsubstituted or is substituted with one or more (typically 1-4 substituents) independently selected from the group of substituents listed below in the definition for "substituents" or otherwise specified. [00172] The term "substituents" refers to a group "substituted" on an alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heterocyclyl, or heteroaryl group at any atom of that group. Suitable substituents include, without limitation, halogen, hydroxy, oxo, nitro, haloalkyl, alkyl, alkenyl, alkynyl, alkaryl, aryl, aralkyl, alkoxy, aryloxy, amino, acylamino, alkylcarbanoyl, arylcarbanoyl, aminoalkyl, alkoxycarbonyl, carboxy, hydroxyalkyl, alkanesulfonyl, arenesulfonyl, alkanesulfonamido, arenesulfonamido, aralkylsulfonamido, alkylcarbonyl, acyloxy, cyano or ureido. In some cases, two substituents, together with the carbons to which they are attached to can form a ring.
[00173] As used herein, the term "polymer" refers to the product of a polymerization reaction, and is inclusive of homopolymers, copolymers, terpolymers, tetrapolymers, etc. The term "polymer" is also inclusive of random polymers, block polymers, graft polymers, copolymers, block copolymers, and graft copolymers. As used herein, the term "copolymer" refers to polymers formed by the polymerization reaction of at least two different monomers. [00174] The term "copolymer backbone" as used herein refers to that portion of the polymer which is a continuous chain comprising the bonds formed between monomers upon polymerization. The composition of the copolymner backbone can be described in terms of the identity of the monomers from which it is formed without regard to the composition of branches, or sidechains, of the polymer backbone. The term "sidechain" refers to portions of the monomer which, following polymerization, forms an extension of the copolymer backbone.
[00175] As used herein, the term "biocompatible" refers to a material that is capable of interacting with a biological system without causing cytotoxicity, undesired protein or nucleic acid modification or activation of an undesired immune response. "Biocompatibility" also includes essentially no interactions with recognition proteins, e.g., naturally occurring antibodies, cell proteins, cells and other components of biological systems. [00176] As used herein an ester sidechains means a sidechains of the formula -
R"'C(O)-ORE, where RE is independently Cl-C6alkyl, Cl-C6alkenyl, Cl-C6alkynyl, cyclyl, heterocycly, aryl, or heteroaryl, each of which can be optionally substituted; and R'" is a bond or C1-C6 alkylene, were the alkylene can comprise one or more double or triple bonds and/or the backbone of the alkylene can be interrupted by O, S, S(O), NH, or C(O). Preferably R'" is a bond. [00177] As used herein an amide sidechains means a sidechains of the formula -
R"C(O)-N(RA)2, where RA is independently H, Cl-C6alkyl, Cl-C6alkenyl, Cl-C6alkynyl, cyclyl, heterocycly, aryl, heteroaryl, saccharide, disaccharide, or trisaccharide, each of which can be optionally substituted; and R" is a bond or C1-C6 alkylene, were the alkylene can comprise one or more double or triple bonds and/or the backbone of the alkylene can be interrupted by O, S, S(O), NH, or C(O). Preferably R" is a bond.
[00178] As used herein a carboxylic acid chain means a sidechains of the formula -
R""C(O)OH where R"" is a bond or C1-C6 alkylene, were the alkylene can comprise one or more double or triple bonds and/or the backbone of the alkylene can be interrupted by O, S, S(O), NH, or C(O). Preferably R"" is a bond.
[00179] Some non-exhaustive examples of biocompatible polymers include polyamides, polycarbonates, polyalkylenes, polyalkylene glycols, polyalkylene oxides, polyalkylene terepthalates, polyvinyl alcohols, polyvinyl ethers, polyvinyl esters, polyvinyl halides, polyvinylpyrrolidone, polyglycolides, polysiloxanes, polyurethanes and copolymers thereof, alkyl cellulose, hydroxyalkyl celluloses, cellulose ethers, cellulose esters, nitro celluloses, polymers of acrylic and methacrylic esters, methyl cellulose, ethyl cellulose, hydroxypropyl cellulose, hydroxy-propyl methyl cellulose, hydroxybutyl methyl cellulose, cellulose acetate, cellulose propionate, cellulose acetate butyrate, cellulose acetate phthalate, carboxylethyl cellulose, cellulose triacetate, cellulose sulphate sodium salt, poly(methylmethacryl ate), poly (ethylmethacrylate), poly(butylmethacrylate), poly (isobutylmethacrylate), poly(hexlmethacrylate), poly (isodecylmethacrylate), poly(laurylmethacrylate), poly (phenylmethacrylate), poly(methacrylate), poly (isopropacrylate), poly(isobutacrylate), poly (octadecacrylate), polyethylene, polypropylene poly (ethylene glycol), poly(ethylene oxide), poly(ethylene terephthalate), poly( vinyl alcohols), poly( vinyl acetate), poly vinyl chloride, polystyrene, polyhyaluronic acids, casein, gelatin, gluten, polyanhydrides, polyacrylic acid, alginate, chitosan, any copolymers thereof, and any combination of any of these. Additionally, biocompatible polymers and copolymers that have been modified for desirable enzymatic degradation, or change upon application of light, ultrasonic energy, radiation, a change in temperature, pH, osmolality, solute or solvent concentration are also amenable to the present invention.
[00180] The present invention may be defined in any of the following numbered paragraphs:
1. A biocompatible conjugated polymer nanoparticle comprising: a copolymer backbone; a plurality of sidechains covalently linked to said backbone; and a plurality of platinum compounds dissociably linked to said sidechains.
2. The nanoparticle of paragraph 1, wherein said plurality of platinum compounds is selected from Pt(II) compounds, Pt(IV) compounds, and any combinations thereof.
3. The nanoparticle of paragraph 1 or 2, wherein at least one of said plurality of platinum compounds is linked to said sidechain through at least one coordination bond. 4. The nanoparticle of paragraph 3, wherein said coordination bond is between an oxygen of the sidechains and the platinum atom of the platinum compound.
5. The nanoparticle of paragraph 4, wherein said oxygen is a carbonyl oxygen.
6. The nanoparticle of paragraph 4, wherein said oxygen is an amide oxygen.
7. The nanoparticle of any of paragraphs 1-6, wherein said copolymer comprises maleic acid monomers.
8. The nanoparticle of paragraph 7, wherein at least one carboxylic acid of the maleic acid is derivatized to an amide.
9. The nanoparticle of any of paragraphs 1-8, wherein said copolymer is poly(isobutylene-α/Mnaleic acid) (PIMA).
10. The nanoparticle of any of paragraphs 1-9, wherein said copolymer comprises from 2 to 100 monomer units.
11. The nanoparticle of any of paragraphs 1-10, wherein said copolymer comprises from 25 to 50 monomer units.
12. The nanoparticle of any of paragraphs 1-11, wherein said sidechains are selected from the group consisting of polymers, monosaccharides, dicarboxylic acids, and combinations thereof.
13. The nanoparticle of any of paragraphs 1-12, wherein said sidechains are polyethylene glycol (PEG).
14. The nanoparticle of paragraph 13, wherein said PEG sidechains have a molecular weight of from 100 to 5000 Dalton.
15. The nanoparticle of paragraph 13, wherein said PEG sidechains have a molecular weight of from 1000 to 3000 Dalton.
16. The nanoparticle of paragraph 13, wherein said PEG sidechains have a molecular weight of about 2000 Dalton.
17. The nanoparticle of any of paragraphs 1-12, wherein said sidechains are monosaccharides .
18. The nanoparticle of paragraph 17, wherein said monosaccharides are glucosamine.
19. The nanoparticle of any of paragraphs 1-18, wherein said platinum compound is a Pt(II) compound selected from the group consisting of cisplatin, oxaliplatin, carboplatin, paraplatin, sartraplatin, and combinations thereof.
20. The nanoparticle of paragraph 19, wherein said platinum (II) compound is cisplatin.
21. The nanoparticle of paragraph 19, wherein said platinum compound is oxaliplatin. 22. The nanoparticle of any of paragraphs 1-21, wherein the number of sidechains corresponds between 50% and 100% of the number of monomeric units of said polymer backbone.
23. The nanoparticle of any of paragraphs 1-22, wherein the number of said sidechains corresponds to a number greater than 90% of the number of monomeric units of said polymer backbone.
24. The nanoparticle of any of paragraphs 1-23, wherein the number of said platinum compounds corresponds between 10% and 100% of the number of monomeric units of said polymer backbone.
25. The nanoparticle of any of paragraphs 1-24, wherein the number of said platinum compounds corresponds between 25% and 75% of the number of monomeric units of said polymer backbone.
26. The nanoparticle of any of paragraphs 1-25, wherein said sidechains comprise dicarboxylic acids.
27. The nanoparticle of paragraph 26, wherein said dicarboxylic acids are of the formula HOOC-R-COOH, wherein R is a CrC6alkyl, C2-C6alkenyl, or C2-C6alkynyl.
28. The nanoparticle of paragraph 27, wherein said dicarboxylic acid is maleic acid.
29. A biocompatible conjugated polymer nanoparticle comprising: a poly(isobutylene-α/Mnaleic acid) backbone, wherein said backbone contains 25 to
50 monomer units; a plurality of PEG sidechains covalently linked to said backbone, wherein said PEG sidechains have a molecular weight of from 1000 to 3000 Dalton and wherein the number of said PEG sidechains corresponds to between 50% and 100% of the number of monomeric units of said polymer backbone; and a plurality of cisplatin sidegroups dissociably linked to said backbone wherein the number of said cisplatin sidegroups is between 25% and 75% of the number of monomeric units of said polymer backbone.
30. A biocompatible conjugated polymer nanoparticle comprising: a poly(isobutylene-α/Mnaleic acid) backbone, wherein said backbone consist of 40 monomers; a plurality of PEG sidechains covalently linked to said backbone, wherein said PEG sidechains have a molecular weight of 2000 Dalton and wherein the number of said
PEG sidechains is greater than 90% of monomeric units of said polymer backbone; and a plurality of cisplatin sidegroups dissociably linked to said backbone, wherein the number of said cisplatin sidegroups is between 25% and 75% of the number of monomeric units of said polymer backbone.
31. A biocompatible conjugated polymer nanoparticle comprising: a poly(isobutylene-α/Mnaleic acid) backbone, wherein said backbone comprises from
25 to 50 monomers; a plurality of glucosamine sidechains covalently linked to said backbone and wherein the number of said glucosamine sidechains is between 50% and 100% of monomeric units of said polymer backbone; and a plurality of cisplatin sidegroups dissociably linked to said backbone, wherein the number of said cisplatin sidegroups is between 25% and 75% of the number of monomeric units of said polymer backbone.
32. A biocompatible conjugated polymer nanoparticle comprising: a poly(isobutylene-α/Mnaleic acid) backbone, wherein said backbone comprises from
25 to 50 monomers; a plurality of glucosamine sidechains covalently linked to said backbone and wherein the number of said glucosamine sidechains is greater than 90% of monomeric units of said polymer backbone; and a plurality of cisplatin sidegroups dissociably linked to said backbone, wherein the number of said cisplatin sidegroups is between 25% and 75%, inclusive, of the number of monomeric units of said polymer backbone.
33. A carboxylic acid-platinum compound complex conjugated nanoparticle comprising: a carboxylic acid-platinum compound complex; and a plurality of lipid-polymer chains, wherein the carboxylic acid portion of said carboxylic acid-platinum compound complex is covalently bound to said lipid- polymer chains.
34. The nanoparticle of paragraph 33, wherein the carboxylic acid is maleic acid.
35. The nanoparticle of any of paragraphs 33-34, wherein the polymer is PEG.
36. The nanoparticle of any of paragraphs 33-35, wherein the platinum compound is a Pt(II) compound selected from the group consisting of cisplatin, oxaliplatin, carboplatin, paraplatin, sartraplatin, and combinations thereof.
37. The nanoparticle of paragraph 36, wherein the Pt(II) compound is cisplatin.
38. The nanoparticle of any of paragraphs 33-37, wherein the platinum compound loading is from l%-30%. 39. The nanoparticle of any of paragraphs 33-38, wherein the platinum compound loading is from l%-6%.
40. A vesicle, micelle, or liposome compound comprising a plurality of nanoparticles of any of paragraphs 33-39.
41. A dicarbonyl-lipid compound having the structure
Figure imgf000039_0001
42. A vesicle, micelle, liposome or nanoparticle compound comprising a dicarbonyl-lipid compound of paragraph 41 and a platinum compound, wherein the platinum compound is dis sociably linked to the compound of paragraph 41.
43. The nanoparticle of paragraph 42, wherein the platinum compound is selected from Pt(II) compounds, Pt(IV) compounds, and any combinations thereof.
44. The nanoparticle of paragraph 43, wherein said platinum compound is a Pt(II) compound selected from the group consisting of cisplatin, oxaliplatin, carboplatin, paraplatin, sartraplatin, and combinations thereof.
45. The nanoparticle of paragraph 43, wherein said platinum (II) compound is cisplatin.
46. The nanoparticle of paragraph 43, wherein said platinum compound is oxaliplatin.
47. A nanoparticle compound comprising a biocompatible polymer, wherein the polymer comprises at least one monomer having the formula -CH(CO2H)-R-CH(C(O)R')- , wherein R is a bond, Ci-C6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom. Preferably, R is a bond.
48. The nanoparticle of paragraph 47, wherein the polymer comprises from 2 to 100 monomeric units having the formula -CH(CO2H)-R-CH(C(O)R')-.
49. The nanoparticle of any of paragraphs 47-48, wherein the polymer comprises from 25 to 50 monomeric units having the formula -CH(CO2H)-R-CH(C(O)R')-. 50. The nanoparticle of any of paragraphs 47-49, wherein R'
is
Figure imgf000040_0001
Or -NH(CH2CH2O)1nCH3, wherein m is 1-150.
51. The nanoparticle of any of paragraphs 47-50, further comprising a bioactive agent.
52. A pharmaceutical composition comprising: the nanoparticle or compound of paragraphs 1-51; and a pharmaceutically acceptable carrier.
53. A method of treating cancer or metastasis comprising: administering to a subject in need thereof an effective amount of the composition of any of paragraphs 1-52.
54. The method of paragraph 53, wherein said cancer or metastasis is selected from the group consisting of platinum susceptible or resistant tumors.
55. The method of paragraph 54, wherein said cancer or metastasis is selected from the group consisting of breast, head and neck, ovarian, testicular, pancreatic, oral- esophageal, gastrointestinal, liver, gall bladder, lung, melanoma, skin cancer, sarcomas, blood cancers, brain tumors, glioblastomas, tumors of neuroectodermal origin and any combinations thereof.
56. A method of sustain release of a platinum compound at a specific location in a subject comprising: providing at the location a composition of paragraph the composition of any of paragraphs 1-52.
57. The method of paragraph 56, wherein composition is in the form of a gel.
58. The method of any of paragraphs 56-57, wherein the location is a tumor.
59. The method of paragraph 58, wherein the tumor was removed before providing the composition.
[00181] To the extent not already indicated, it will be understood by those of ordinary skill in the art that any one of the various embodiments herein described and illustrated may be further modified to incorporate features shown in any of the other embodiments disclosed herein.
[00182] The following examples illustrate some embodiments and aspects of the invention. It will be apparent to those skilled in the relevant art that various modifications, additions, substitutions, and the like can be performed without altering the spirit or scope of the invention, and such modifications and variations are encompassed within the scope of the invention as defined in the claims which follow. The following examples do not in any way limit the invention.
EXAMPLES Materials and methods
[00183] CellTiter 96 AQueous One Solution Cell Proliferation Assay [3-(4,5- dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt (MTS) assay] reagent was from Promega (Madison, WI). All Polymer solutions were dialyzed in cellulose membrane tubing, types Spectra/Por 4 and Spectra/Por 6 (wet tubing), with mass-average molecular mass cut-off limits of 1000 and 3500 respectively. Operations were performed against several batches of stirred deionized H2O. Commercially supplied (Sigma, Fluka AG, Aldrich Chemie GmbH) chemicals, reagent grade, were used as received. These included N,N-Dimethylformamide (DMF), Poly(isobutylene-alt-maleic anhydride), Glucosamine.HCl, mPEG2000NH2, Diaza(l,3)bicycle[5.4.0]udecane (DBU), Triethyl amine. 1H NMR and 13C NMR were measured at 300 and 400 MHz, respectively, with a Varion-300 or a Brucker-400 spectrometer. IH NMR chemical shifts are reported as δ values in parts per million (ppm) relative to either tetramethylsilane (0.0 ppm) or deuterium oxide (4.80 ppm). Data is reported as follows: chemical shift, multiplicity (s = singlet, d = doublet, t = triplet, q = quartet, m = mutliplet, b = broad), coupling constants (hertz), and integration. Carbon-13 chemical shifts are reported in ppm relative to CDC13 (76.9 ppm) or relative to DMSOd6 (39.5 ppm). 195Pt NMR chemical shifts are reported as δ in ppm relative to Na2PtCl6 (0.0 ppm). In some experiments, 1H NMR and 13C NMR were measured at 500 and 125 MHz, respectively, with a Varion 500 or a Brucker-400 spectrometer.
[00184] Starting materials were azeotropically dried prior to reaction as required, and all air- and/or moisture- sensitive reactions were conducted in flame- and/or oven-dried glassware under an anhydrous nitrogen atmosphere with standard precautions taken to exclude moisture.
Cell Culture and Cell Viability Assay
[00185] The Lewis Lung Carcinoma cell lines (LLC) and Breast Cancer cell line
(4Tl) were purchased from American Type Culture Collection (ATCC, Rockville, MD, USA). Lewis Lung Carcinoma cells were cultured in Dulbecco's Modified Eagle's Medium supplemented with 10% FBS, 50 unit/ml penicillin and 50 unit/ml streptomycin. The 4Tl cells were cultured in RPMI medium supplemented with 10% FBS, 50 unit/ml penicillin and 50 unit/ml streptomycin. Trypsinized cultured LLC and 4Tl cells were washed twice with PBS and seeded into 96-well flat bottomed plates at a density of 2xlO3 cells in 100 μl of medium. Different concentrations of conjugates were tested in triplicate in the same 96-well plate for each experiment. Medium alone was kept as negative control and CDDP as positive control. The plates were then incubated for 48 h in a 5% CO2 atmosphere at 37 0C. The cells were washed and incubated with 100 μl phenol- red free medium (without FBS) containing 20 μl of the CellTiter 96 Aqueous One Solution reagent (Promega, WI, USA). This assay [3- (4,5-dimethylthiazol- 2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt] (MTS) is a colorimetric method for determining the number of viable cells in proliferation or cytotoxicity assays. After 2-h incubation in a 5% CO2 atmosphere at 37 0C, the absorbance in each well was recorded at 490 nm using a VERSA max plate reader (Molecular Devices, Sunnyvale, CA, USA).
[00186] The absorbance reflects the number of surviving cells. Blanks were subtracted from all data and results analyzed using Origin software (OriginLab Corporation, Northampton, USA). The mean of triplicated absorbance data for each tested dose was divided by the mean of untreated control cells. The log of the quotient was used to plot a graph as a function of given dose, i.e. Y = (Tested Absorbance Mean- Background)/(Untreated Absorbance Mean-Background) vs. X = tested dose.
Particle Size Measurement
[00187] High resolution TEM images were obtained on a JEOL 2011 high contrast digital TEM. Samples were prepared on carbon 300 mesh copper grids (Electron Microscopy Sciences) by adding drops of aqueous nanoparticles at different concentrations, and allowed to air-dry. The size distribution of nanoparticles was studied by dynamic light scattering (DLS), which was performed at 25°C on a DLS-system (Malvern NanoZetasizer) equipped with a He Ne laser.
Physicochemical Release Kinetics Studies
[00188] PEVIA-GA-CDDP was suspended in 1 mL of hypoxic-cell lysate from LLC cell line and sealed in a dialysis bag (MWCO ~ 1000 Da). The dialysis bag was incubated in 1 mL of PBS buffer at room temperature with gentle shaking. 10 μL of aliquot was extracted from the incubation medium at predetermined time intervals, treated with 90 μL of 1,2- phenylene diamine solution (1.2 mg in 1 mL DMF) and incubated for 3h at 1000C. The released Pt(IV) was quantified by UV-VIS spectroscopy at characteristic wavelength λ = 704 nm of Pt(IV)- 1,2-phenylene diamine complex. After withdrawing each aliquot the incubation medium was replenished by 10 μL of fresh PBS.
[00189] Alternatively, concentrated PIMA-GA-cisplatin conjugate was resuspended into 100 μL of double distilled water with pH adjusted to 8.5 or 5.5 using IN sodium hydroxide or IN nitric acid and transferred to a dialysis tube (MWCO: 1000 KD, Spectrapor). The dialysis tube was put into a tube containing magnetic pallet and 2 mL solutions of different pH phosphate-buffered saline. Cisplatin release was studied by gently stirring the dialysis bag at 300 rpm using IKA stirrer at 25oC. 10 μL aliquots were taken from the outside solution of dialysis membrane bag at predetermined time intervals and subjected to next UV- Vis active complex formation reaction by adding 100 μL of ortho-phenyldiamine (1.2 mg/ml in DMF) and heating the resulting solution for 3h. 10 μL of fresh solution was added back to outside solution of dialysis membrane bag to maintain same volume. The amount of the drug that was released was evaluated by UV-spectrophotometer (Shimadzu UV 2450) at 706 nm.
FACS analysis for apoptosis
[00190] Cells were grown in 6-well plates incubated in the presence of cisplatin nanoparticle or free cisplatin at 370C for 24 h. After 24 h, the cells were washed with PBS and collected at O0C. The cells were then treated with annexin V-Alexa Fluor 488 conjugate (Molecular Probes, Invitrogen) and incubated in the dark, at room temperature, for 15 min. The cells were then washed with PBS and incubated with propidium iodide (PI) solution (50 g/mL; Sigma) containing RNase (1 mg/mL; Sigma). The cell suspensions were then transferred to FACS tubes and analyzed for Annexin V/PI staining on a BD FACS Calibur instrument. Data were analyzed using a CellQuestPro software (BD Biosciences).
Cellular Uptake Studies
[00191] LLC and 4Tl cells were seeded on glass coverslips in 24-well plates, 50000 cells per well. When cells reached 70% confluency, they were treated with fluorescein isothiocyanate (FITC)-conjudated cisplatin nanoparticles for different durations of 30 min, 2 h, 6 h, 12 h, and 24 h, respectively. For colocalization studies, at indicated time points, the cells were washed with PBS and incubated with Lysotracker Red (Molecular Probes) at 370C for 30 min to allow internalization. The cells were then fixed with 4% paraformaldehyde for 20 min at room temperature, then washed twice with PBS and mounted on glass slides using Prolong Gold Antifade Reagent (Molecular Probes). Images were obtained using a Nikon Eclipse TE2000 fluorescence microscope equipped with green and red filters for FITC and Lysotracker Red, respectively.
In Vivo Murine LLC Lung Cancer and 4Tl Breast Cancer Tumor Model [00192] The LLC Lung Cancer cells and 4Tl Breast Cancer cells (3x105) were implanted subcutaneously in the flanks of 4- week-old C57/ BL6 and BALB/c mice (weighing 20 g, Charles River Laboratories, MA) respectively. The drug therapy was started after the tumors attained volume of 50 mm3. The tumor therapy consisted of administration of cisplatin nanoparticle and free cisplatin or oxaliplatin and free oxaliplatin. The formulations were prepared and validated such that 100 μL of cisplatin nanoparticle and free cisplatin contained 1.25 and 3 mg/kg of cisplatin or 100 μL of oxaliplatin nanoparticle and free oxaliplatin contained 5 and 15 mg/kg of oxaliplatin Administration was by tail vein injection. PBS (100 μL) administered by tail-vein injection was used as a control for drug treatment. The tumor volumes and body weights were monitored on a daily basis. The animals were sacrificed when the average tumor size of the control exceeded 2000 mm3 in the control group. The tumors were harvested immediately following sacrifice and stored in 10% formalin for further analysis. All animal procedures were approved by Harvard institutional IUCAC.
In Vivo Murine Ovarian Cancer Tumor Model
[00193] Ovarian adenocarcinomas were induced in genetically-engineered K- rasLSL/+/Ptenfl/fl mice via intrabursal delivery of adenovirus carrying Cre recombinase, as described previously. Tumor cells were engineered to express luciferase once activated by Adeno-Cre, in order to make tumor imaging feasible before and after drug treatment. Once mice developed medium to large tumors they were placed into one of four treatment groups (controls, cisplatin NPl.25 mg/kg, cisplatin NP-3 mg/kg, and free cisplatin), with all drugs administered intravenously (Lv.).
Tumor Imaging and Efficacy Assessment of Drug Treatment
[00194] Tumor imaging in vivo was performed with the IVIS Lumina II Imaging
System. Quantification of bioluminescence was achieved by using the Living Image Software 3.1 (Caliper Life Sciences). Mice received 150 mg/kg of D-luciferin firefly potassium salt via intraperitoneal (i.p.) injection prior to imaging. Five minutes post-luciferin injection, animals were anesthetized in a 2.5% isoflurane induction chamber. Once anesthesized, mice were placed into the imaging chamber where they were kept under anesthesia by a manifold supplying isoflurane and their body temperature was maintained by a 37°C temperature stage. Bioluminescent signal was collected fifteen minutes after luciferin administration for an exposure time of thirty seconds. Images were taken a day prior to treatment (day 0, baseline), in the middle of the treatment cycle, and one day following the final treatment. Treatment efficacy was quantified by examining the fold increase in bioluminescence of the post- treatment signal as compared to baseline. Statistical analysis of the toxicity data was analyzed using a one-way ANOVA test with the Prism 5™ software.
Biodistribution of Cisplatin
[00195] Cisplatin-nanoparticles and free cisplatin were injected i.v. (dose equivalent to
8 mg/Kg of cisplatin) in mice to study its distribution. After 24 hours of injections, the animals were sacrificed and necropsy was performed to harvest the tumor and kidney. In another study, the animals were dosed repeatedly following the efficacy study protocols, and the animals were sacrificed at the end of the multiple dosing study. The organs were then weighed and dissolved in Cone. HNO3 (approx. 10 mL) by shaking for 24 hours at room temperature and then heating at 1000C for 12 hours. To these mixtures were then added 30% H2O2, the resulting solutions were stirred for 24 hours at room temperature and then heated for another 12 hours to evaporate the liquids. All solid residues were re-dissolved in 1 mL water and then amount of platinum was measured by inductively coupled plasma- spectrometry (ICP).
Histopathology and TUNEL assay (Apoptotic assay)
[00196] The tissues were fixed in 10% formalin, paraffin embedded, sectioned and stained with H&E at the Harvard Medical School Core Facility. Tumor and Kidney paraffin sections were deparaffinized and stained with standard TMR-red fluorescent terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) kit following the manufacturer's protocol (In Situ Cell Death Detection Kit, TMR Red, Roche). Images were obtained using a Nikon Eclipse TE2000 fluorescence microscope equipped with red filter. Toxicity Assessment of Drug Treatment
[00197] Body weights were recorded daily to assess toxicity. In addition, livers and spleens were removed at the end of treatment to record weights and perform extensive pathological examination to assess toxicity of vital organs. Cell apoptosis in vital organs was measured using TUNNEL assay. Statistical analysis of the toxicity data was performed using a two-way ANOVA test with the Prism 5™ software.
Statistical Analysis
[00198] Data were expressed as means + S. D from at least n=3. Statistical analysis was conducted using the GraphPad Prism software (GraphPad, San Diego, CA). The statistical differences were determined by ANOVA followed by Newman Keuls Post Hoc test or Student's t test, p < 0.05 was consideredtoindicate significant differences.
Example 1 - Synthesis of Polymeric Carriers
Poly(isobutylene-alt-maleic acid) PIMA (2)
[00199] Poly(isobutylene-alt-maleic anhydride) 1 (Ig) was dissolved in 5 ml of dry
DMF in 10 mL round bottom flask to which was added double distilled water (ImL) and then resulting reaction mixture was stirred at 800C for 48h. Solvent was removed under vacuum and low molecular weight impurities were removed using dialysis. Aqueous polymer solution was dialyzed for 3 days in cellulose membrane tubing, types Spectra/Por 4 and with mass- average molecular mass cut-off limits of 1000. The colorless solution was then lyophilized to get 732 mg of white colored polymer Poly(isobutylene-alt-maleic acid) PIMA (2). 1H NMR (300 MHz, D2O) δ 3.3-3.5 (m), 2.8 (s), 2.6-2.7 (m), 2.5 (s), 2.2-2.3 (m), 0.8-0.9 (m).
PIMA-EDA O)
[00200] The 10 mL RB flask equipped with magnetic stirrer and dry nitrogen balloon was charged with Poly(isobutylene-alt-maleic anhydride) PIMA 1 (Ig), dry DMF (5mL), Triethyl amine (0.ImL) and excess Ethylendiamine dihydrochloride (Ig). The resulting mixture was stirred at 25°C for 48h. Solvent was removed under vacuum and polymer was purified by removing low molecular weight impurities such as excess Ethylendiamine using dialysis bag of molecular cut off of 3.5KD for 3 days. The polymer solution was then lyophilized to get 0.89 g of PIMA-EDA (3). 1H-NMR (300 MHz, D2O) δ 3.1-3.2 (m), 2.9-3.0 (m), 2.6-2.8 (m), 2.5 (m), 0.8-1.0 (m). PIMA-GA Polymer (4)
[00201] Poly(isobutylene-alt-maleic anhydride) PIMA 1 (0.0064 g, 0.001 mmol) was dissolved in DMF (5 niL) and then was added DBU (0.032 niL, dissolved in 1 niL dry DMF, 0.21 mmol) and the mixture was stirred at 25 0C for Ih. To this solution was added Glucosamine (0.046 g 0.21 mmol) directly. The resulting reaction mixture was allowed to stir at room temperature for 48 h and then quenched by adding double distilled water (ImL). The organic solvent was evaporated under vacuum for 12 hours. The resulting pale yellow solid was purified by dialysis for 3 days using dialysis bag supplied by Pierce (Thermo scientific) of molecular cut off of 3.5KD to colorless solution. Lyophilization gave 104 mg of white colored PIMA-GA (4) polymer. 1H-NMR (300 MHz, CDC13) δ 7.54-7.65 (m, 2 H), 7.33-7.45 (m, 2 H), 7.02-7.19 (m, 14 H), 6.93-6.97 (m, 2 H), 6.83-6.89 (m, 2 H), 6.55 (s, 2 H), 6.15- 6.19 (m, 2 H), 3.90 (s, 2 H), 3.58 (s, 6 H). 1H-NMR (300 MHz, D2O) δ 8.2-8.3 (m), 7.0-7.1 (m), 5.0-5.1 (m), 3.0-3.9 (m), 2.1-2.3 (m), 1.1-1.9 (m), 0.7-1.0 (m). [00202] In another experiment, PIMA (0.045 g) was dissolved in DMF (5 mL) and then was added solution of DBU (0.23 mL) and glucosamine (0.323 g dissolved in 5 mL dry DMF). The resulting reaction mixture was allowed to stir at room temperature for 48 h and then quenched by adding dd water (ImL). The organic solvent was evaporated under vacuum. The resulting pale yellow solid was purified by dialysis for 3 days using dialysis bag of molecular cut off of 3.5KD. Lyophilization gave 104 mg of slightly yellow colored PIMA- GA polymer. 1H-NMR (300 MHz, D2O) δ 8.2-8.3 (m), 7.07.1 (m), 5.0-5.1 (m), 3.0-3.9 (m), 2.1-2.3 (m), 1.1-1.9 (m), 0.7-1.0 (m).
PIMA-PEG Polymer (5)
[00203] The Poly(isobutylene-alt-maleic anhydride) PIMA 1 (3 mg, 0.0005 mmol) and
DBU (0.0023 mL, 0.015 mmol) was dissolved in Dry DMF (10 mL) in 25 mL RB flask under N2 for 1 h and then was added PEG-NH2 (20 mg, 0.01 mmol), the resulting reaction solution was then heated at 80 0C with continuous stirring for 3days. The reaction was allowed to cool to room temperature and then water (1 mL) was added and continue stirring for 1 h. Solvents are removed under vacuum and unreacted PEG-NH2 of MW 2KD was removed from required polymer by dialysis. Dialysis was carried out for 5 days using membrane of molecular cut off of 3.5KD supplied by Pierce (Thermoscientific) to give colorless solution which was then lyophilized to give 19 mg white colored PIMA-PEG (5). 1H-NMR (300 MHz, D2O) δ 3.5-3.7 (m), 3.0-3.1 (m), 2.5-2.8 (m), 0.7-1.0 (m). Example 2 - Synthesis of conjugates
Aquation of CDDP
[00204] CDDP (30mg) and AgNO3 (17mg) was added to 10ml double distilled water.
The resulting solution was stirred in dark at room temperature for 24h. AgCl precipitates were found after reaction. AgCl precipitates are removed from reaction by centrifugation at 10000 rpm for 10 min. The supernatant was further purified by passing through 0.2μm filter.
PIMA-CDDP (6)
[00205] Poly(isobutylene-alt-maleic acid) PIMA 2 (0.006 g, 0.001 mmol) was dissolved in ImI double distilled water containing CDDP (0.00084 g, 0.0028 mmol) in 10 mL round bottom flask to and then resulting reaction mixture was stirred at room temperature (25°C) for 48h. The PIMA-CDDP (6) conjugate was further purified by dialyzing it in cellulose membrane tubing, types Spectra/Por 4 and with mass-average molecular mass cutoff limits of 1000. The resulting turbid solution was then lyophilized to get white colored PIMA-CDDP (6) conjugate. The conjugate was re-suspended for cell culture experiments.
PIMA-EDA-CDDP (7)
[00206] In 10 mL RB flask was weighed PIMA-EDA 3 (0.007 g, 0.001 mmol) polymer to which was added CDDP (0.0084 g, 0.0028 mmol) dissolved in double distilled water (1 mL). The solution was then stirred at room temperature (25°C) for 48h. Dialysis using cellulose membrane with molecular mass cut-off limits of 1000 and lyophilization gave yellowish colored PIMA-EDA-CDDP (7) conjugate.
PIMA-GA-CDDP (8)
[00207] To PIMA-GA 4 (0.0036 g, 0.0003 mmol) weighed in 10 mL RB flask equipped with magnetic stirrer was added ImI double distilled water containing CDDP (0.001 g, 0.0033 mmol) and then the solution was stirred at room temperature (25°C) for 48h. The PIMA-GA-CDDP (8) conjugate formed in solution was further purified by dialysis to remove unattached CDDP with mass-average molecular mass cut-off limits of 1000 for 2-3 hours. Lyophilization of the dialyzed solution resulted in slightly yellow colored PIMA-GA- CDDP (8) conjugate. PIMA-PEG-CDDP (9)
[00208] The brush polymer PIMA-PEG 5 (0.019 g, 0.00007 mmol) was taken in 10 niL RB flask mixed with CDDP (0.0002 g, 0.0007 mmol) dissolved in 0.3 mL double distilled water. After stirring for 3 days at room temperature (25°C) the resulting turbid reaction mixture was dialyzed. The solution containing PIMA-PEG-CDDP (2) conjugate was further purified by dialyzing it in cellulose membrane tubing, types Spectra/Por 4 and with mass-average molecular mass cut-off limits of 1000 for 2-3 hours to remove free CDDP. PIMA-PEG-CDDP (9) conjugate was then lyophilized to get white colored solid. The conjugate was re- suspended in double distilled water for cell culture experiments.
FITC-labeled PIMA-GA-CDDP
[00209] Poly(isobutylene-alt-maleic anhydride) PIMA (0.006 g) was dissolved in
DMF (5 mL) and then was added a solution of DBU (0.0053 mL in DMF) and Glucosamine (0.0075 g dissolved in 5 mL dry DMF) the mixture was stirred at 25°C for Ih. The resulting reaction mixture was allowed to stir at 25°C for 48 h and then to which was added 0.0022 g FITC-EDA (FITC-EDA was synthesized by stirring Fluorescein isothiocyanate in excess ethylene diamine at 25°C for 12 h in DMSO) and continue stirring for another 12 h, reaction mixture was quenched by adding double distlled water (ImL). The organic solvent was evaporated under vacuum. The resulting orange solid was purified by dialysis for 3 days using dialysis bag of molecular cut off of 3.5KD. Lyophilization gave fluorescent orange PIMA-GA-FITC polymer. To this FITC labeled polymer (PIMA-GA-FITC, 0.004 g) was added ImI double distilled water containing cisplatin (0.001 g) and then the solution was stirred at room temperature (25°C) for 48h. The PIMA-GA-FITC-cisplatin conjugate formed in solution was further purified by dialysis to remove unattached cisplatin with mass-average molecular mass cut-off limits of 1000. Lyophilization of the dialyzed solution resulted in orange colored FITC labeled PEVIA-GAFITC-cisplatin conjugate nanoparticles.
PIMA-Oxaliylatin
[00210] Poly(isobutylene-alt-maleic acid) (PIMA) (6mg) was dissolved in ImI double distilled water containing oxaliplatin-OH (lmg) in a round bottom flask to and then resulting reaction mixture was stirred at room temperature (25oC) for 48h. The PIMA-oxaliplatin conjugate was further purified by dialyzing it in cellulose membrane tubing, types Spectra/Por 4 and with mass-average molecular mass cut-off limits of 1000. The resulting turbid solution was then lyophilized to get PIMA-oxaliplatin conjugate. The conjugate was re-suspended for cell culture experiments.
PIMA-GA-Oxalwlatin
[00211] To PIMA-GA (12mg) weighed in 10 niL RB flask equipped with magnetic stirrer was added ImI double distilled water containing oxaliplatinOH (lmg) and then the solution was stirred at room temperature (25°C) for 48h. The PIMA-GA-oxaliplatin conjugate formed in solution was further purified by dialysis to remove unattached oxaliplatin with mass-average molecular mass cut-off limits of 1000. Lyophilization of the dialyed solution resulted in yellow colored PIMA-GA-oxaliplatin conjugate.
Example 3 - NMR analysis of PIMA-GA polymer synthesis using different bases
Synthesis of PIMA-GA using DBU as the base
[00212] Glucosamine hydrochloride (360 mg, 1.66 mmol, 200 equiv) was suspended in 5 mL DMF and treated with DBU (250 μL, 1.66 mmol, 200 equiv) at room temperature for Ih. After Ih glucosamine/DBU (in DMF) solution was added drop wise into poly (isobutylene-alt-maleic anhydride) (50 mg, 0.008 mmol, 1 equiv) solution in 5 mL DMF and the reaction mixture was stirred for 72h at room temperature. The reaction mixture was quenched with 3 mL of dd-water. The PIMA-GA conjugate was purified by dialysis using 2000 MWCO dialysis bag for 72h. The product was lyophilized for 48h to obtain 100 mg cream yellow powder. The product was characterized by 1H NMR spectroscopy (300 MHz). Solubility: product was soluble in water but not soluble in organic solvent e.g. acetone, methanol or acetonitrile. 1H NMR (300 MHz): δ (ppm) = 5.2-5.3 (m, 0.14 H, sugar proton), 5.0-5.1 (m, 0.4 H, sugar proton), 3.6-4.0 (m, 13.07 H, sugar proton), 3.25-3.5 (m, 15.48 H, sugar proton), 3.0-3.2 (m, 6.98 H, sugar proton), 2.5-2.6 (m, 6.97 H, PIMA proton), 1.4-1.7 (m, 19.86 H, PEVIA proton), 0.7-1.2 (m, 23.77 H, PIMA proton). Total sugar protons: total PIMA protons =36.07: 50.6= 0.71. This fits well with the predicted structure if all the residues are derivatized sugar protons and PIMA protons in PIMA-GA conjugate monomer.
Synthesis of PIMA-GA using diisoproylethylamine (DIPEA) as base
[00213] Glucosamine hydrochloride (179 mg, 0.83 mmol, 100 equiv) was suspended in 2 mL DMF and treated with DIPEA (145 μL, 0.83 mmol, 100 equiv) at room temperature for Ih. After Ih poly (isobutylene-alt-maleic anhydride) (50 mg, 0.008 mmol, 1 equiv) (dissolved in 3 mL DMF) was added into the reaction mixture and stirred for 24h at room temperature. The reaction mixture was quenched with 3 mL of dd- water. The PIMA-GA conjugate was purified by dialysis using 1000 MWCO dialysis bag for 24h. The product was lyophilized for 48h to obtain 106 mg white powder. The product was characterized by 1H NMR spectroscopy (300 MHz). Solubility: product was soluble in water but not soluble in organic solvent e.g. acetone, methanol or acetonitrile. 1H NMR (300 MHz): δ (ppm) = 5.2- 5.3 (m, 0.4 H, sugar proton), 4.9-5.1 (m, 2.0 H, sugar proton), 3.4-3.6 (m, 21.86 H, sugar proton), 3.2-3.3 (m, 6.16 H, sugar proton), 2.9-3.1 (m, 3.81 H, sugar proton), 2.4-2.7 (broad, 4.39 H, PIMA proton), 2.1-2.4 (broad, 4.54 H, PIMA proton), 1.7-2.0 (broad, 3.13 H, PIMA proton), 1.3-1.5 (braod, 1.58 H, PIMA proton), 1.1-1.2 (m, 24.12 H, PIMA proton), 0.6-0.9 (m, 27.94 H, PIMA proton). Total sugar protons: total PIMA protons = 39.21: 61.11 = 0.64.
Synthesis of PIMA-GA using trietylamine as base
[00214] Glucosamine hydrochloride (143 mg, 0.66 mmol, 80 equiv) was suspended in
2 mL DMF and treated with triethylamine (100 μL, 0.66 mmol, 80 equiv) at room temperature for Ih. After Ih poly (isobutylene-alt-maleic anhydride) (50 mg, 0.008 mmol, 1 equiv) was added into the reaction mixture and stirred for 24h at room temperature. The reaction mixture was quenched with 3 mL of dd- water. The PIMA-GA conjugate was purified by dialysis using 1000 MWCO dialysis bag for 24h. The product was lyophilized for 48h to obtain 100 mg white powder. The product was characterized by 1H NMR spectroscopy (300 MHz). Solubility: product was soluble in water but not soluble in organic solvent e.g. acetone, methanol or acetonitrile. 1H NMR (300 MHz): δ (ppm) = 5.2-5.3 (m, 0.44 H, sugar proton), 4.9-5.1 (m, 1.51H, sugar proton), 3.7-3.8 (m, 19.01 H, sugar proton), 3.3-3.4 (m, 6.43 H, sugar proton), 3.1-3.2 (m, 11.82, sugar proton), 2.93-2.94 (m, 2.23 H, PIMA proton), 2.6-2.7 (m, 5.84 H, PIMA proton), 2.2-2.5 (broad, 4.91 H, PIMA proton), 1.8-2.1 (broad, 3.83 H, PIMA proton), 1.4-1.6 (broad, 2.52 H, PIMA proton), 1.8-1.2 (m, 18.04 H, PIMA proton), 0.9-1.0 (m, 23.77 H, PIMA proton). Total sugar protons: total PIMA protons = 34.31: 65.7 = 0.52.
Example 4 - Time dependent loading efficiency of PIMA-GA-CDDP.
[00215] Method: PIMA-GA conjugate (50 mg, 0.004 mmol) was dissolved in 1 mL dd-water followed by the addition of (NHi)2Pt(OH)2 (3mL, 0.057 mmol). The reaction was stirred at room temperature for 48h. 200 μL of aliquots were taken out from the reaction mixture after each pre-determined time points (5h, 3 Ih and 48h). The aliquots were filtered through Microcon centrifugal filter device having regenerated cellulose membrane of 3000 MWCO to separate the PIMA-GA-CDDP conjugate. The polymer was washed thoroughly (200 μL x 2) with dd- water to remove any platinum reagent. The platinum content in polymer was determined by the method described before.
[00216] Result: The change in Pt-loading efficiency in PIMA-GA conjugate was determined by the ability of conjugating 1,2-phenylenediamine with Pt giving rise to UV-VIS spectra at wavelength λ = 706 nm. Neither the polymer nor 1,2-phenyldiamine has any characteristic peak at this wavelength. How the Pt-content changes with time in the reaction between PIMA-GA and hydroxy-platin was monitored by UV-VIS spectra. At different predetermined time points (5h, 3 Ih and 48h) 200 μL of aliquots were taken out from the reaction mixture and the Pt-loading in the polymer conjugate was determined. The figure shows that loading of platinum in the polymer conjugate increases with time from 190μg/mg (5h) to 210 μg/mg (3Ih) and reaches maximum 347μg/mg at 48h. This indicates almost 100% of the Pt is complexed with the polymer at this time point as the maximal predicted loading is 37.5% per polymer unit, and we attain 34.7% Pt per polymer.
Example 5 - Rational optimization of the polymer based on structure-activity relationship.
[00217] In order to improve efficacy of the nanoparticles, the inventors derivatized one arm of each monomer of the polymer with biocompatible glucosamine to generate PIMA- glucosamine conjugate (PIMA-GA) (Fig.1 IB). This converted the dicarboxylato bonds with Pt to a monocarboxylato bond and a coordinate bond, which can release Pt more easily given that a coordinate bond is less stable than a monocarboxylato linkage (Fig. 11 B). [00218] Nuclear magnetic resonance (NMR) characterization of the Pt environment revealed that complexation of PIMA-GA and cisplatin in an acidic pH (pH 6.5) generated an isomeric state [PEVIA-GA-Cisplatin (0->Pt)] (8) characterized by the monocarboxylato and a 0->Pt coordination complex as characterized by a single Pt NMR peak at -1611.54 (Fig. HB). Interestingly, complexing the cisplatin with PIMA-GA at an alkaline pH (pH 8.5) favored the formation of an isomeric PIMA-GA-Cisplatin (N->Pt) complex (10), where the Pt is complexed through a monocarboxylato and a more stable N->Pt coordinate bond characterized by a unique peak at -2210 (Fig. 1 IB). Excitingly, the existing of these two pH- dependent states allowed the inventors to further dissect the impact of Pt environment, specifically the leaving groups, on the biological efficacy.
[00219] The complexation of cisplatin to PIMA-glucosamine (PIMA-GA) polymer at a ratio of 15:1 resulted in self assembly into nanoparticles in the desired narrow size bandwidth of 80-150 nm as confirmed by high-resolution transmission electron microscopy (data not shown) and DLS (Fig.12A). Furthermore, the inventors achieved a loading of 175 + 5 μg/mg of polymer (Fig.12B), which is significantly higher than can be achieved using traditional nanoparticle formulations (Avgoustakis K, Beletsi A, Panagi Z, Klepetsanis P, Karydas AG, Ithakissios DS. PLGA-mPEG nanoparticles of cisplatin: in vitro nanoparticle degradation, in vitro drug release and in vivo drug residence in blood properties. I Control Release. 2002 Feb 19;79(l-3):123-35).
Example 6 - Characterizing the uptake and efficacy of nanoparticles in vitro
[00220] Tagging the polymer with fluorescein (Fig. 15) enabled the temporal tracking of uptake of the nanoparticles into the cells, which were co-labeled with a lysotracker-red dye to label the endolysosomal compartments. A rapid uptake of the nanoparticles was observed in the LLC cells within 15 min of treatment with internalization into the endolysosomal compartment as evident by colocalization of the FITC-nanoparticles and the Lysotracker-Red dye (data not shown). In contrast, the uptake into 4Tl cells was delayed, with internalization into the endolysosomal compartment evident only after 2 hours post-incubation. Over a 12 hour period, the fluorescent signals from the lysosomal compartment and the FITC- conjugated dissociate, suggesting a cytosolic distribution of the polymer after processing within the lysosome (data not shown).
[00221] To test the efficacy of the PEVIA-GA-cisplatin nanoparticles in vitro, the inventors performed cell viability assays using Lewis lung carcinoma (LLC) and 4Tl breast cancer cell lines. Cell viability was quantified using a MTS assay at 48 hours post- incubation. Interestingly, the LLC cells (Fig.13A) were more susceptible to cisplatin-nanoparticles than the 4Tl breast cancer cells (Fig.13B). Excitingly, PEVIA-GA-cisplatin (0->Pt) nanoparticles (8) demonstrated significant LLC cell kill with IC50 values (4.25 + 0.16μM) similar (P>0.05) to cisplatin (IC50 = 3.87 + 0.37μM), and superior to carboplatin (IC50 = 14.75 + 0.38μM), which supports the hypothesis that the rate of aquation is critical for efficacy (Fig.13). A similar efficacy was observed when the inventors replaced glucosamine with ethylene diamine, which creates a similar Pt complexation environment as glucosamine (Fig.13A). This was additionally supported by the observation that PIMA-GA-cisplatin (N->Pt) nanoparticles (IC50= 6.36 + 0.19μM) were significantly less active than cisplatin, suggesting that the platinum environment is critical in defining the rate of aquation. To further validate the role of complexation environment, the inventors generated PIMA-GA(20), where only 20 of the 40 monomers comprising a PIMA polymer were derivatized with glucosamine, thereby introducing dicarboxylato bonds and reducing the monocarboxylato plus coordinate bonds that complex Pt to PIMA-GA. As shown in Fig.lόF, the concentration-efficacy curve shifts to the right with PIMA-GA(20)-cisplatin (EC50= 5.85 + 0.13μM) as compared with PIMA-GA- cisplatin (0->Pt) nanoparticles, where all the 40 monomers are derivatized with glucosamine. Empty PIMA-GA polymer had no effect on the cell viability. Table summarizes the EC50 values.
[00222] As shown in Fig. 13 A, while the polymer alone induced cell death at the highest concentrations, complexation of cisplatin significantly shifted the concentration- effect curve to the left, indicating that the PIMA-cisplatin nanoparticle induces cell kill. However, even at a concentration of 50 uM, the PIMA cisplatin failed to induce complete cell kill. In contrast, cisplatin exerts complete cell kill at a concentration greater than 20 uM. This reduction in the efficacy of palatinate when complexed with PIMA can be explained by the dicaroboxylateo linkage between the platinum and the maleci acid monomers, which tightly binds the Pt similar to the linkage that exists in carboplatin, which similarly is less efficacious than cisplatin.
[00223] Labeling the cells for expression of phosphatidylserine on the cell surface, revealed that the cisplatin treatments could induce apoptotic cell death, with LLCs being more susceptible than 4Tl cells (Fig. 14).
Table 1: EC50 values for various complexes
Figure imgf000054_0001
Example 7 - The release of active cisplatin from nanoparticle is pH-dependent [00224] Given that the nanoparticles localized to the lysosomal compartment, the inventors tested the release of Pt from the nanoparticles at pH 5.5, mimicking the acidic pH of the endolysosomal compartment of the tumor (Lin, et al., Eur. J. Cancer, 2004 40(2):291- 297). The inventors also selected pH8.5 as a reference pH in the alkaline range. As shown in Fig. 16, at pH5.5 PIMA-GA-cisplatin (0->Pt) nanoparticles resulted in a sustained but significant release of cisplatin monitored over a 70 hour period. In contrast the release at pH8.5 was significantly lower, indicating a pH-dependent release of Pt. Interestingly, PIMA- GA-cisplatin (N->Pt) released significantly lower amounts of Pt even at pH5.5, consistent with the fact that the N->Pt coordinate bond is stronger than the 0->Pt linkage. As expected, the inventors observed that PEVIA-cisplatin nanoparticles exhibited significantly lower rates of Pt release as compared with both PIMA-GA-cisplatin (N->Pt) and PIMA-GA-cisplatin (O- >Pt) as the Pt is held by more stable dicarboxylato bonds instead of a monocarboxylato and a coordinate bond.
Example 8 - Nanoparticle induces tumor growth delay and regression with reduced nephrotoxicity
[00225] As PIMA-GA-cisplatin (0->Pt) nanoparticles exhibited the desired release rates for platinum and also exhibited in vitro efficacy comparable to cisplatin, the inventors validated the therapeutic efficacy of the nanoparticles in vivo. They randomly sorted mice bearing established Lewis lung carcinoma or 4Tl breast cancer into five groups respectively and treated each group with three doses of (i) PBS (control); (ii) Cisplatin (1.25 mg/kg); (iii) Cisplatin(3mg/kg); (iv) PIMA-GA-Cisplatin (0->Pt) nanoparticles (1.25 mg/kg); (v) PIMA- GA-Cisplatin (0->Pt) nanoparticles (3 mg/kg). The mice injected with PBS formed large tumors by day 16 (day after the last injection), and consequently, were euthanized. The animals in the other groups were also sacrificed at the same time point to evaluate the effect of the treatments on tumor pathology. As shown in Fig. 5, cisplatin induced dose-dependent tumor inhibition, and at a dose equivalent to 1.25 mg/kg of cisplatin, administration of the nanoparticle formulation resulted in greater inhibition of lung carcinoma progression as compared with the free drug. However, at a dose equivalent to 3mg/kg, free cisplatin resulted in a significant reduction in body weight indicating systemic toxicity. In contrast, animals treated with nanoparticles equivalent to 3mg/kg of cisplatin exhibited weight gain, although tumor inhibition was similar in both treatment groups (data not shown). Furthermore, necropsy revealed that treatment with free cisplatin resulted in a significant reduction in the weights of kidney and spleen (Figs. 5D and 5E), indicating nephrotoxicity and hematotoxicity consistent with previous reports. Excitingly, cisplatin nanoparticles had no effect on the weights of the kidneys, and reduced spleen size only at the highest dose (Fig. 5D and 5E). This was further validated by pathological analysis of kidney H&E stained cross-sections, which revealed significant tubular necrosis in the animals treated with free cisplatin as compared with cisplatin nanoparticle. To elucidate the mechanism underlying tumor inhibition, the inventors labeled the tumor cross sections for TUNEL, which revealed a significant induction of apoptosis following treatment with both free cisplatin and PIMA- GAcisplatin(O->Pt) nanoparticles (data not shown). Interestingly, labeling the kidney sections for TUNEL demonstrated significant apoptosis in the animals treated with free cisplatin as opposed to minimal nephrotoxicity in the nanoparticle-treated group (data not shown). Indeed, biodistribution studies using inductively coupled plasma-spectrometry (ICP) revealed that the concentration of Pt in the kidney following administration of the cisplatin nanoparticle is 50% of that attained following administration of free drug (Fig. 5E), which can explain the reduction in nephrotoxicity.
[00226] Treatment with cisplatin (1.25mg/kg) exhibited only a minor tumor growth inhibition as compared with control; in contrast, treatment with nanoparticle-cisplatin at the same dose exerted a dramatic increase in the antitumor efficacy (Fig. 5A). This is consistent with the fact that nanoparticles enable a significantly higher concentration of the active agent to be attained within the tumor as compared to free drug20. At the higher dose, both the free drug and the nanoparticle achieved similar antitumor efficacy (Fig. 5A), which is potentially the theoretical limit of the drug. However, the free drug at this dose resulted in a greater than 20% loss of body weight (Fig. 5B), which is an indicator of non-specific toxicity. Indeed, it induced significant nephrotoxicity as seen by the loss of weight of the kidney (Fig. 5D). Furthermore, although the blood counts were not different between the various treatment groups (Fig. 5C), there was significant loss of weight of the spleen at the highest dose of the free cisplatin (Fig. 5E). In contrast, nanoparticle-cisplatin exhibited no such toxicity even at the highest dose, open up the possibility of dosing at higher levels or for longer time periods, both of which can dramatically impact antitumor outcomes. Furthermore, the ease of manufacturing, the low costs of materials and the increase in therapeutic efficacy and reduction of toxicity can become an example of nanotechnology impacting global health. Without wishing to be bound by theory, the increased therapeutic index can arise from a preferential accumulation of the nanoparticles in the tumors arising from the well-studied EPR effect, and circumventing the kidney as it exceeds the size limit for clearance, which in a previous study was shown to be less than 5 nm (Choi HS, Liu W, Misra P, Tanaka E, Zimmer JP, Itty Ipe B, Bawendi MG, Frangioni JV. Renal clearance of quantum dots. Nat Biotechnol. 2007 ;25: 1165-70).
[00227] Both free cisplatin and PIMA-GA-cisplatin(O->Pt) nanoparticles resulted in similar levels of tumor growth inhibition in the 4Tl breast cancer model (Fig. 17). Interestingly, both 1.25mg/kg and 3mg/kg free cisplatin induced a significant loss of body weight as compared with the cisplatin-nanoparticle treated groups. Consistent with the observations in the lung cancer model, while free cisplatin induced significant apoptosis in the kidney, the nanoparticle-cisplatin treated groups exhibited minimal apoptosis in the kidney but significant levels of apoptosis in the tumor.
[00228] In addition to lung and breast cancer models, the inventors further evaluated the PIMA-GA-cisplatin(O->Pt) nanoparticle in an ovarian cancer model. Epithelial ovarian cancer is the deadliest malignancies of the female reproductive cycle. The discovery of frequent somatic PTEN mutations and loss of heterozygosity at the 10q23 PTEN locus in endometrioid ovarian cancer implicates a key role for PTEN in the etiology of this epithelial ovarian cancer subtype (Obata, K. et al. Frequent PTEN/MMAC1 mutations in endometrioid but not serous or mucinous epithelial ovarian tumors. Cancer Res. 58, 2095-2097 (1998) and Sato, et al., Cancer Res. 2000, 60: 7052-7056 and Sato, et al., Cancer Res. 2000, 60: 7052- 7056). Similarly, K-RAS oncogene is also mutated in endometrioid ovarian cancer, albeit at a lesser frequency (Cuatrecasas, et al., Cancer (1998) 82:1088-1095). In a recent study, the combination of these two mutations in the ovarian surface epithelium was found to induce invasive and widely metastatic endometrioid ovarian adenocarcinomas with complete penetrance, making it a good model for mimicking human tumor progression. In this transgenic model vehicle-treated animals exhibited rapid tumor progression as quantified by luciferase expression. Treatment with the cisplatin nanoparticles resulted in a dose-dependent inhibition of tumor progression, with the lower dose equivalent to 1.25 mg/kg exerting a similar inhibition as a 3mg/kg dose of free cisplatin (Fig. 18). Treatment with the higher dose of cisplatin-nanoparticle (equivalent to 3mg/kg of cisplatin) resulted in greater tumor inhibition without any significant loss of body weight as observed with an equidose of free cisplatin, which is approved for clinical use in ovarian cancer (Fig. 18). Furthermore, TUNEL staining revealed significant apoptosis in the kidney at 3 mg/kg of free cisplatin while the cisplatinnanoparticles at equivalent Pt concentration did not induce apoptosis of the nephrons.
Example 9 - Biodistribution of dsplatin-nanoparticles following multiple dosing
[00229] To study the biodistribution of the cisplatin nanoparticles, the inventors harvested the tumors at the end of the multiple-dosing experiments, where each animal received three doses of free drug or the cisplatin nanoparticle. As shown in Fig. 19, there was a preferential accumulation Pt in both breast and ovarian tumors when administered as a nanoparticle as opposed to when delivered as free cisplatin. Example 10 - Toxicity Assessment of Treatment with PIMA-GA-oxaliplatin
[00230] As seen in Fig. 23B, at a dose of 15mg/kg of free oxaliplatin, all the animals died due to systemic toxicity. In contrast no toxicity was evident even at this dose in the case of oxaliplatin nanoparticle.
Example 11 - Synthetic scheme of lipid-cisplatin conjugate
[00231] In addition to the PMA-GA-cisplatin conjugate, the inventors have also engineered an analog, where maleic acid is conjugated to PEG end of a pegylated lipid (PEG2000-DSPE). The inventors complexed Pt to the maleic acid, resulting in the formation of a platinated lipid derivative where the Pt is at the hydrophilic end and the lipid forms the hydrophobic end. These form micelles in water, and the loading efficiency is 45 μg/mg of lipid derivative. This can be increased by using a lower molecular weight PEG or lipid. See Fig. 10.
Example 12 - Cisplatin-liponanoparticles
Materials and Method
[00232] All reactions were performed under inert conditions unless otherwise indicated. All commercially obtained compounds were used without further purification. DCM, dry DCM, Methanol, Cholesteryl Chloroformate, Cholesterol, Ethylenediamine, Succinic Anhydride, Silver Nitrate, Sodium Sulphate, Pyridine, Cisplatin, L-a- Phosphatidylcholine, Sephadex G-25 and 1,2-Phenylenediamine were bought from Sigma- Aldrich. l,2-Distearoyl-sn-Glycero-3-Phosphoethanolamine-N-[Amino(Polythylene Glycol)2000] and the mini handheld Extruder kit (including 0.2 μm Whatman Nucleopore Track- Etch Membrane, Whatman filter supports and 1.0 mL Hamiltonian syringes) were bought from Avanti Polar Lipids Inc. Anhydrous solvent DMF was supplied by Acros Organics. Phosphotungstic Acid was from Ted Pella, Inc. Analytical thin-layer chromatography (TLC) was performed using precoated silica gel Aluminium sheets 60 F254 bought from EMD Laboratories. Spots on the TLC plates were visualized using alkanine permanganate or 6% Ninhydrin solution in Acetone. IH NMR (300 MHz) spectra were obtained on a Varian Mercury 300 spectrophotometer. The chemical shifts are expressed in parts per million (ppm) using suitable deuterated NMR solvents with reference to TMS at 0 ppm. MTS reagent was supplied by Promega. The cell viability assay and release kinetic data were plotted using GraphPad Prism software. Each sample was done in triplicate. Synthesis of (11 ):
[00233] 1044 μL (15 eq) of ethylene diamine (12) was dissolved in 5.0 niL anhydrous
DCM followed by cooling down to 0-5 0C with ice. 500.0 mg (1.0 eq) of Cholesteryl Chloroformate was dissolved in 5.0 rnL anhydrous DCM and was added to the reaction mixture drop- wise over a period 15 minutes with vigorous stirring and was continued overnight until it comes to rt. The reaction was worked up using water (50 mLX3) and DCM (50 mL), followed by saturated Brine water wash. The organic layer was dried over anhydrous Sodium Sulphate, and evaporated with the help of a rotary evaporator. Light yellow colored clear oily product (13) was separated with 99.1% yield. IH-NMR (300 MHz) d(ppm) = 5.37 (s, IH), 5.06 (S, IH), 4.49 (bs, IH), 3.22-3.20 (m, 2H), 2.82-2.81 (m, 2H), 2.34-2.26 (m, 2H), 2.02-1.83 (m, 6H), 1.54-0.84 (m, 37H)
Synthesis of 15:
[00234] 350 mg (0.74 mmol, 1 eq) of starting material (13) was dissolved in 5.0 mL anhydrous DCM. To it 370.0 mg (3.7 mmols, 5 eq) of Succinic Anhydride (14) and catalytic amount of Pyridine was added. The stirring was continued for Id followed by work up in 0.1 (N) HCl and DCM for several times. The organic layer was dried over Sodium Sulphate and evaporated to get white amorphous solid compound (15). Yield: 95% IH-NMR (300 MHz) d(ppm) = 7.72-7.70 (m, IH), 7.54-7.53 (m, IH), 5.37 (s, IH), 5.07 (s, IH), 4.49 (bs, IH), 4.22-4.19 (m, 2H), 3.36-3.30 (m, 4H), 2.68-2.33 (m, 4H), 2.02-1.83 (m, 6H), 1.54-0.84 (m, 37H).
Synthesis of 16:
[00235] 50 mg (0.166 mmol, 1 eq) of Cisplatin (16) was partially dissolved in 10.0 mL of H2O. To it 28.0 mg (0.166 mmol, 1 eq) Silver Nitrate was added and the resulting reaction mixture was stirred at rt for Id. It looked milky white and Silver Chloride was removed by centrifuging at 25000 Xg for Ih. Synthesis of 7: 200 mg (0.35 mmol, 1.0 eq) of 5 was dissolved in 5.0 mL DMF. To it 20.0 mL of product 6 (cone 5.0mg/mL, 1.0 eq) was added and stirred for Id. The reaction mixture was dried with the help of a lyophilizer. The dried product (17) was used for lipo-nanoparticle synthesis without any further purification.
General Procedure of Synthesizing Lipo-nanoparticles: [00236] 10.0 mg of L-a-Phosphatidylcholine, 5.0 mg of Cholesterol (or Pt(II)- cholesterol conjugate) and 1.0 mg of l,2-Distearoyl-sn-Glycero-3-Phosphoethanolamine-N- [Amino(Polythylene Glycol)2000] were dissolved in 10.0 rnL DCM. It was evaporated into a thin and uniform film with the help of a rotary evaporator. After thorough drying with pump it was hydrated with 1.0 mL H2O for 2 h at 60 0C. The hydrated lipo-nanoparticles looked light yellow to white with little viscous texture. It was passed though Sephadex G-25 column and extruded at 65 0C.
General Method of Pt(II) Quantification in Lipo-nanoparticles
[00237] A measured amount of the extruded lipo-nanoparticle was heated at 100 0C in
1.2 mg/mL concentration of 1,2-Phenylenediamine in DMF for 2h. Pt(II) amount was calculated by UV Spectrophotometry (Shimadzu 2450).
Release Kinetics
[00238] Concentrated drug loaded lipo-nanoparticles were suspended with buffer (or cell lysate) and sealed in a dialysis membrane (MW cutoff 1000, Spectrum Lab). The dialysis bags were incubated in 1.0 mL PBS buffer at room temperature with gentle shaking. A 10 μL portion of the aliquot was collected from the incubation medium at predetermined time intervals, and the released drug was quantified by UV Spectrophotometer (Shimadzu 2450). The results are plotted as percentage release.
Sample Preparation for TEM
[00239] High resolution TEM images were obtained on a Jeol 2011 high contrast digital TEM. For sample preparation, lacy carbon 300 mesh copper grids (Electron microscopy Science) were dipped in the aqueous solution of the lipo-nanoparticle. It was allowed to air dry followed by staining it with 2% aqueous solution of Phosphotungstic acid. The size distribution of lipo-nanoparticles was studied by dynamic light scattering (DLS), which was performed at 260C on a Malvern Zetasizer DLS-system equipped with a He-Ne laser.
Cell Viability Assay
[00240] In a 96 well plate, 2x10 cells were plated. After 4h, cells were treated with different concentrations of free drug or lipo-nanoparticles. Cells without any treatment were kept as control. After 48 h, cell viability was assessed using standard MTS assay according to manufacturer's instructions.
In vivo Efficacy and Toxicity Studies
[00241] BALB/c mice were inoculated s.c. with IXlO5 of 4Tl breast tumor cells inlOOμL PBS on right flank of mice. Treatment with different anticancer agents either free or entrapped in nanop articles was started on day when tumor volume reached 200 mm . Typically the animals received free drug alone or in nanoparticles through i.v route every alternate day for total of three dosages. Once the tumor volume reached 2000 mm3 in control group, mice were sacrificed. Tumor, kidney, spleen, lung, liver were harvested and processed for paraffin embedding and sectioning.
[00242] All patents and publications cited herein are hereby incorporated by reference.

Claims

WHAT IS CLAIMED:
1. A biocompatible conjugated polymer nanoparticle comprising: a copolymer backbone; a plurality of sidechains covalently linked to said backbone; and a plurality of platinum compounds dissociably linked to said sidechains.
2. The nanoparticle of claim 1, wherein said plurality of platinum compounds is selected from Pt(II) compounds, Pt(IV) compounds, and any combinations thereof.
3. The nanoparticle of claim 1 or 2, wherein at least one of said plurality of platinum compounds is linked to said sidechain through at least one coordination bond.
4. The nanoparticle of claim 3, wherein said coordination bond is between an oxygen of the sidechains and the platinum atom of the platinum compound.
5. The nanoparticle of claim 4, wherein said oxygen is a carbonyl oxygen.
6. The nanoparticle of claim 4, wherein said oxygen is an amide oxygen.
7. The nanoparticle of any of claims 1-6, wherein said copolymer comprises maleic acid monomers.
8. The nanoparticle of claim 7, wherein at least one carboxylic acid of the maleic acid is derivatized to an amide.
9. The nanoparticle of any of claims 1-8, wherein said copolymer is poly(isobutylene- α/Mnaleic acid) (PIMA).
10. The nanoparticle of any of claims 1-9, wherein said copolymer comprises from 2 to 100 monomer units.
11. The nanoparticle of any of claims 1-10, wherein said copolymer comprises from 25 to 50 monomer units.
12. The nanoparticle of any of claims 1-11, wherein said sidechains are selected from the group consisting of polymers, monosaccharides, dicarboxylic acids, and combinations thereof.
13. The nanoparticle of any of claims 1-12, wherein said sidechains are polyethylene glycol (PEG).
14. The nanoparticle of claim 13, wherein said PEG sidechains have a molecular weight of from 100 to 5000 Dalton.
15. The nanoparticle of claim 13, wherein said PEG sidechains have a molecular weight of from 1000 to 3000 Dalton.
16. The nanoparticle of claim 13, wherein said PEG sidechains have a molecular weight of about 2000 Dalton.
17. The nanoparticle of any of claims 1-12, wherein said sidechains are monosaccharides.
18. The nanoparticle of claim 17, wherein said monosaccharides are glucosamine.
19. The nanoparticle of any of claims 1-18, wherein said platinum compound is a Pt(II) compound selected from the group consisting of cisplatin, oxaliplatin, carboplatin, paraplatin, sartraplatin, and combinations thereof.
20. The nanoparticle of claim 19, wherein said platinum (II) compound is cisplatin.
21. The nanoparticle of claim 19, wherein said platinum compound is oxaliplatin.
22. The nanoparticle of any of claims 1-21, wherein the number of sidechains corresponds between 50% and 100% of the number of monomeric units of said polymer backbone.
23. The nanoparticle of any of claims 1-22, wherein the number of said sidechains corresponds to a number greater than 90% of the number of monomeric units of said polymer backbone.
24. The nanoparticle of any of claims 1-23, wherein the number of said platinum compounds corresponds between 10% and 100% of the number of monomeric units of said polymer backbone.
25. The nanoparticle of any of claims 1-24, wherein the number of said platinum compounds corresponds between 25% and 75% of the number of monomeric units of said polymer backbone.
26. The nanoparticle of any of claims 1-25, wherein said sidechains comprise dicarboxylic acids.
27. The nanoparticle of claim 26, wherein said dicarboxylic acids are of the formula HOOC-R-COOH, wherein R is a Ci-C6alkyl, C2-C6alkenyl, or C2-C6alkynyl.
28. The nanoparticle of claim 27, wherein said dicarboxylic acid is maleic acid.
29. A biocompatible conjugated polymer nanoparticle comprising: a poly(isobutylene-α/Mnaleic acid) backbone, wherein said backbone contains 25 to 50 monomer units; a plurality of PEG sidechains covalently linked to said backbone, wherein said PEG sidechains have a molecular weight of from 1000 to 3000 Dalton and wherein the number of said PEG sidechains corresponds to between 50% and 100% of the number of monomeric units of said polymer backbone; and a plurality of cisplatin sidegroups dissociably linked to said backbone wherein the number of said cisplatin sidegroups is between 25% and 75% of the number of monomeric units of said polymer backbone.
30. A biocompatible conjugated polymer nanoparticle comprising: a poly(isobutylene-α/Mnaleic acid) backbone, wherein said backbone consist of 40 monomers; a plurality of PEG sidechains covalently linked to said backbone, wherein said PEG sidechains have a molecular weight of 2000 Dalton and wherein the number of said
PEG sidechains is greater than 90% of monomeric units of said polymer backbone; and a plurality of cisplatin sidegroups dissociably linked to said backbone, wherein the number of said cisplatin sidegroups is between 25% and 75% of the number of monomeric units of said polymer backbone.
31. A biocompatible conjugated polymer nanoparticle comprising: a poly(isobutylene-α/Mnaleic acid) backbone, wherein said backbone comprises from
25 to 50 monomers; a plurality of glucosamine sidechains covalently linked to said backbone and wherein the number of said glucosamine sidechains is between 50% and 100% of monomeric units of said polymer backbone; and a plurality of cisplatin sidegroups dissociably linked to said backbone, wherein the number of said cisplatin sidegroups is between 25% and 75% of the number of monomeric units of said polymer backbone.
32. A biocompatible conjugated polymer nanoparticle comprising: a poly(isobutylene-α/Mnaleic acid) backbone, wherein said backbone comprises from
25 to 50 monomers; a plurality of glucosamine sidechains covalently linked to said backbone and wherein the number of said glucosamine sidechains is greater than 90% of monomeric units of said polymer backbone; and a plurality of cisplatin sidegroups dissociably linked to said backbone, wherein the number of said cisplatin sidegroups is between 25% and 75%, inclusive, of the number of monomeric units of said polymer backbone.
33. A carboxylic acid-platinum compound complex conjugated nanoparticle comprising: a carboxylic acid-platinum compound complex; and a plurality of lipid-polymer chains, wherein the carboxylic acid portion of said carboxylic acid-platinum compound complex is covalently bound to said lipid- polymer chains.
34. The nanoparticle of claim 33, wherein the carboxylic acid is maleic acid.
35. The nanoparticle of any of claims 33-34, wherein the polymer is PEG.
36. The nanoparticle of any of claims 33-35, wherein the platinum compound is a Pt(II) compound selected from the group consisting of cisplatin, oxaliplatin, carboplatin, paraplatin, sartraplatin, and combinations thereof.
37. The nanoparticle of claim 36, wherein the Pt(II) compound is cisplatin.
38. The nanoparticle of any of claims 33-37, wherein the platinum compound loading is from l%-30%.
39. The nanoparticle of any of claims 33-38, wherein the platinum compound loading is from l%-6%.
40. A vesicle, micelle, or liposome compound comprising a plurality of nanoparticles of any of claims 33-39.
41. A dicarbonyl-lipid compound having the structure
Figure imgf000065_0001
42. A vesicle, micelle, liposome or nanoparticle compound comprising a dicarbonyl-lipid compound of claim 41 and a platinum compound, wherein the platinum compound is dis sociably linked to the compound of claim 41.
43. The nanoparticle of claim 42, wherein the platinum compound is selected from Pt(II) compounds, Pt(IV) compounds, and any combinations thereof.
44. The nanoparticle of claim 43, wherein said platinum compound is a Pt(II) compound selected from the group consisting of cisplatin, oxaliplatin, carboplatin, paraplatin, sartraplatin, and combinations thereof.
45. The nanoparticle of claim 43, wherein said platinum (II) compound is cisplatin.
46. The nanoparticle of claim 43, wherein said platinum compound is oxaliplatin.
47. A nanoparticle compound comprising a biocompatible polymer, wherein the polymer comprises at least one monomer having the formula -CH(CO2H)-R-CH(C(O)R')- , wherein R is a bond, Ci-C6 alkylene, where the alkylene can comprise one or more double or triple bonds; and R' is a substituted nitrogen atom. Preferably, R is a bond.
48. The nanoparticle of claim 47, wherein the polymer comprises from 2 to 100 monomeric units having the formula -CH(CO2H)-R-CH(C(O)R')-.
49. The nanoparticle of any of claims 47-48, wherein the polymer comprises from 25 to 50 monomeric units having the formula -CH(CO2H)-R-CH(C(O)R')-.
50. The nanoparticle of any of claims 47-49, wherein R' is
Figure imgf000066_0001
or _ NH(CH2CH2O)1nCH3, wherein m is 1-150.
51. The nanoparticle of any of claims 47-50, further comprising a bioactive agent.
52. A pharmaceutical composition comprising: the nanoparticle or compound of claims 1-51; and a pharmaceutically acceptable carrier.
53. A method of treating cancer or metastasis comprising: administering to a subject in need thereof an effective amount of the composition of any of claims 1-52.
54. The method of claim 53, wherein said cancer or metastasis is selected from the group consisting of platinum susceptible or resistant tumors.
55. The method of claim 54, wherein said cancer or metastasis is selected from the group consisting of breast, head and neck, ovarian, testicular, pancreatic, oral-esophageal, gastrointestinal, liver, gall bladder, lung, melanoma, skin cancer, sarcomas, blood cancers, brain tumors, glioblastomas, tumors of neuroectodermal origin and any combinations thereof.
56. A method of sustain release of a platinum compound at a specific location in a subject comprising: providing at the location a composition of claim the composition of any of claims 1-52.
57. The method of claim 56, wherein composition is in the form of a gel.
58. The method of any of claims 56-57, wherein the location is a tumor.
59. The method of claim 58, wherein the tumor had been removed before providing the composition.
PCT/US2010/023217 2009-02-04 2010-02-04 Nanoscale platinum compounds and methods of use thereof WO2010091192A2 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
JP2011549262A JP5844642B2 (en) 2009-02-04 2010-02-04 Nanoscale platinum compounds and methods of use thereof
US13/147,751 US9393227B2 (en) 2009-02-04 2010-02-04 Nanoscale platinum compounds and methods of use thereof
EP10739130.2A EP2393854B1 (en) 2009-02-04 2010-02-04 Nanoscale platinum compounds and methods of use thereof
RU2011134316/04A RU2538199C2 (en) 2009-02-04 2010-02-04 Platinum nanocompounds and methods of applying thereof
BRPI1008869A BRPI1008869A2 (en) 2009-02-04 2010-02-04 nanoparticle, compound, pharmaceutical composition, method for treating cancer or metastasis, and prolonged release method of a platinum compound
CN201080015384.4A CN102388073B (en) 2009-02-04 2010-02-04 Nano level platinic compound and using method thereof
AU2010210593A AU2010210593B2 (en) 2009-02-04 2010-02-04 Nanoscale platinum compounds and methods of use thereof
HK12108633.2A HK1167869A1 (en) 2009-02-04 2012-09-04 Nanoscale platinum compounds and methods of use thereof
US15/182,967 US10512696B2 (en) 2009-02-04 2016-06-15 Nanoscale platinum compounds and methods of use thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US14972509P 2009-02-04 2009-02-04
US61/149,725 2009-02-04
US24000709P 2009-09-04 2009-09-04
US61/240,007 2009-09-04

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/147,751 A-371-Of-International US9393227B2 (en) 2009-02-04 2010-02-04 Nanoscale platinum compounds and methods of use thereof
US15/182,967 Division US10512696B2 (en) 2009-02-04 2016-06-15 Nanoscale platinum compounds and methods of use thereof

Publications (2)

Publication Number Publication Date
WO2010091192A2 true WO2010091192A2 (en) 2010-08-12
WO2010091192A3 WO2010091192A3 (en) 2010-12-02

Family

ID=42542651

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/023217 WO2010091192A2 (en) 2009-02-04 2010-02-04 Nanoscale platinum compounds and methods of use thereof

Country Status (9)

Country Link
US (2) US9393227B2 (en)
EP (1) EP2393854B1 (en)
JP (2) JP5844642B2 (en)
CN (3) CN105295058B (en)
AU (1) AU2010210593B2 (en)
BR (1) BRPI1008869A2 (en)
HK (1) HK1167869A1 (en)
RU (1) RU2538199C2 (en)
WO (1) WO2010091192A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102525928A (en) * 2012-02-20 2012-07-04 中国药科大学 Cis-platinum long-cycle liposome and preparation method thereof
JP2013535589A (en) * 2010-08-16 2013-09-12 ニルスタル エス.アー. Cosmetic spun fiber, method for obtaining it and use thereof
WO2014201376A2 (en) 2013-06-14 2014-12-18 Invictus Oncology Pvt. Ltd. Lipid-based platinum compounds and nanoparticles
EP2958553A4 (en) * 2013-02-19 2016-10-12 Nat Health Research Institutes Caged platinum nanoclusters for anticancer chemotherapeutics
WO2016185402A1 (en) 2015-05-19 2016-11-24 Invictus Oncology Pvt. Ltd. Process for preparing supramolecular platinum-based compounds
US10426753B2 (en) 2014-04-03 2019-10-01 Invictus Oncology Pvt. Ltd. Supramolecular combinatorial therapeutics
US11497715B2 (en) 2013-03-15 2022-11-15 Cureport, Inc. Methods and devices for preparation of lipid nanoparticles
RU2795256C2 (en) * 2015-05-19 2023-05-02 Акамара Терапьютикс, Инк. Method for obtaining supramolecular platinum compounds
EP3509603B1 (en) * 2016-09-07 2024-01-03 Alyssum Therapeutics, Inc. Immune memory induction by platinum based compounds

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2010210593B2 (en) * 2009-02-04 2015-09-03 The Brigham And Women's Hospital, Inc. Nanoscale platinum compounds and methods of use thereof
EP2884944B1 (en) 2012-08-15 2020-10-07 MiMedx Group, Inc. Reinforced placental tissue grafts and methods of making and using the same
US9943551B2 (en) 2012-08-15 2018-04-17 Mimedx Group, Inc. Tissue grafts composed of micronized placental tissue and methods of making and using the same
US8946163B2 (en) 2012-11-19 2015-02-03 Mimedx Group, Inc. Cross-linked collagen comprising metallic anticancer agents
US9155799B2 (en) 2012-11-19 2015-10-13 Mimedx Group, Inc. Cross-linked collagen with at least one bound antimicrobial agent for in vivo release of the agent
US8940684B2 (en) 2012-11-19 2015-01-27 Mimedx Group, Inc. Cross-linked collagen comprising an antifungal agent
US10111910B2 (en) 2013-01-18 2018-10-30 Mimedx Group, Inc. Methods for treating cardiac conditions
US10206977B1 (en) 2013-01-18 2019-02-19 Mimedx Group, Inc. Isolated placental stem cell recruiting factors
US10029030B2 (en) 2013-03-15 2018-07-24 Mimedx Group, Inc. Molded placental tissue compositions and methods of making and using the same
US10335433B2 (en) 2013-04-10 2019-07-02 Mimedx Group, Inc. NDGA polymers and metal complexes thereof
US9446142B2 (en) * 2013-05-28 2016-09-20 Mimedx Group, Inc. Polymer chelator conjugates
US10449220B2 (en) 2013-08-30 2019-10-22 Mimedx Group, Inc. Micronized placental compositions comprising a chelator
JP7464348B2 (en) 2014-01-17 2024-04-09 ミメディクス グループ インコーポレイテッド Methods for inducing angiogenesis
JP7099822B2 (en) 2014-08-28 2022-07-12 ミメディクス グループ インコーポレイテッド Collagen-enhanced tissue graft
WO2017151912A1 (en) * 2016-03-02 2017-09-08 The Brigham And Women's Hospital, Inc. A reporter platform for real time monitoring of drug efficacy
CN109134874B (en) * 2018-07-29 2021-04-09 桂林理工大学 Two-dimensional gadolinium-based compound with magnetic refrigeration effect and preparation method thereof
CN110455761A (en) * 2019-08-19 2019-11-15 齐鲁工业大学 A kind of fluorescence probe and its application based on conjugated polymer nanoparticle and gold nanorods
CN111715409B (en) * 2020-07-01 2021-07-23 中南大学 Combined lead inhibitor of micro-fine particle galena and application thereof
CN112175013B (en) * 2020-09-23 2021-11-23 华南理工大学 Photoactivated cellulose nano drug-loaded material and preparation method thereof
CN113648423B (en) * 2021-07-22 2022-11-22 南方医科大学 Amphiphilic conjugate anti-tumor nano-drug, preparation method thereof, nano-assembly and application
CN114163481B (en) * 2021-12-06 2023-06-23 郑州大学 Platinum-containing drug nano vesicle and preparation method and application thereof

Family Cites Families (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4680338A (en) * 1985-10-17 1987-07-14 Immunomedics, Inc. Bifunctional linker
DE3630497A1 (en) 1986-09-08 1988-03-10 Behringwerke Ag CIS-PLATINUM COMPLEXES, METHOD FOR THE PRODUCTION THEREOF AND PHARMACEUTICAL AGENTS CONTAINING THESE COMPOUNDS
EP0307827A3 (en) * 1987-09-15 1989-12-27 Kuraray Co., Ltd. Novel macromolecular complexes, process for producing same and medicinal use of such complexes
US4931553A (en) 1988-05-11 1990-06-05 Gill Devinder S Platinum-polymer complexes and their use as antitumor agents
RU2033998C1 (en) * 1990-01-29 1995-04-30 Институт нефтехимического синтеза им.А.В.Топчиева АН СССР Complex compound of cis-diamminoplatinum (ii) with copolymer of maleic acid sodium salt and furan showing immunostimulating and antimalignant activities at broad range of therapeutic doses and low nephrotoxicity
US5871710A (en) 1992-09-04 1999-02-16 The General Hospital Corporation Graft co-polymer adducts of platinum (II) compounds
US5547982A (en) 1995-02-27 1996-08-20 Johnson Matthey, Inc. Anti-tumor platinum complexes
EP1880736A1 (en) * 1999-04-23 2008-01-23 Alza Corporation Releasable linkage and composition containing same
US6365197B1 (en) 2000-09-21 2002-04-02 Council Of Scientific And Industrial Research Antibacterial composition comprising Oenostacin from Oenothera biennis
US20030129224A1 (en) 2001-11-13 2003-07-10 Paul Tardi Lipid carrier compositions and methods for improved drug retention
GB0129121D0 (en) * 2001-12-05 2002-01-23 Ic Vec Ltd Compound
WO2004054499A2 (en) * 2002-08-02 2004-07-01 Transave, Inc. Platinum aggregates and process for producing the same
JP2006519262A (en) 2003-02-28 2006-08-24 アルザ・コーポレーシヨン Liposome compositions for reducing liposome-induced complement activation
CN100384894C (en) * 2003-03-24 2008-04-30 三共株式会社 Higher molecular modifier and medicinal composition
JP2006521300A (en) * 2003-03-31 2006-09-21 プリヴァ−ラケマ,エー.エス. Pharmaceutical composition containing platinum complex as active substance and method for producing the same
US7332527B2 (en) 2003-05-16 2008-02-19 Board Of Regents Of The University Of Nebraska Cross-linked ionic core micelles
US7846412B2 (en) * 2003-12-22 2010-12-07 Emory University Bioconjugated nanostructures, methods of fabrication thereof, and methods of use thereof
EP1722762A2 (en) * 2004-03-02 2006-11-22 Massachusetts Institute of Technology Nanocell drug delivery system
CN101001609A (en) * 2004-08-11 2007-07-18 柯尼卡美能达医疗印刷器材株式会社 Method of manufacturing pharmaceutical preparations containing liposomes
JP2008509205A (en) * 2004-08-13 2008-03-27 アイシー・ベック・リミテッド Vector containing polymer-modified siRNA liposomes
GB0418172D0 (en) * 2004-08-13 2004-09-15 Ic Vec Ltd Vector
CN101056620A (en) * 2004-11-08 2007-10-17 特兰萨夫公司 Methods of treating cancer with lipid-based platinum compound formulations administered intraperitoneally
WO2006091790A1 (en) 2005-02-23 2006-08-31 Xenoport, Inc. Platinum-containing compounds exhibiting cytostatic activity, synthesis and methods of use
CN1911450A (en) * 2006-08-21 2007-02-14 沈阳药科大学 Live target adjuvant containing D-galactose and sterol or aliphatic alcohol and its preparation
CN100496609C (en) * 2006-12-30 2009-06-10 上海艾力斯医药科技有限公司 Stable liposome composition
WO2008112565A2 (en) 2007-03-09 2008-09-18 Anthony Manganaro Method and composition for treating cancer
AU2010210593B2 (en) * 2009-02-04 2015-09-03 The Brigham And Women's Hospital, Inc. Nanoscale platinum compounds and methods of use thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP2393854A4 *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2013535589A (en) * 2010-08-16 2013-09-12 ニルスタル エス.アー. Cosmetic spun fiber, method for obtaining it and use thereof
CN102525928A (en) * 2012-02-20 2012-07-04 中国药科大学 Cis-platinum long-cycle liposome and preparation method thereof
EP2958553A4 (en) * 2013-02-19 2016-10-12 Nat Health Research Institutes Caged platinum nanoclusters for anticancer chemotherapeutics
US11497715B2 (en) 2013-03-15 2022-11-15 Cureport, Inc. Methods and devices for preparation of lipid nanoparticles
WO2014201376A2 (en) 2013-06-14 2014-12-18 Invictus Oncology Pvt. Ltd. Lipid-based platinum compounds and nanoparticles
US10730899B2 (en) 2013-06-14 2020-08-04 Akamara Therapeutics, Inc. Lipid-based platinum compounds and nanoparticles
US10426753B2 (en) 2014-04-03 2019-10-01 Invictus Oncology Pvt. Ltd. Supramolecular combinatorial therapeutics
WO2016185402A1 (en) 2015-05-19 2016-11-24 Invictus Oncology Pvt. Ltd. Process for preparing supramolecular platinum-based compounds
US11358978B2 (en) 2015-05-19 2022-06-14 Akamara Therapeutics, Inc. Process for preparing supramolecular platinum-based compounds
RU2795256C2 (en) * 2015-05-19 2023-05-02 Акамара Терапьютикс, Инк. Method for obtaining supramolecular platinum compounds
EP3509603B1 (en) * 2016-09-07 2024-01-03 Alyssum Therapeutics, Inc. Immune memory induction by platinum based compounds

Also Published As

Publication number Publication date
EP2393854A2 (en) 2011-12-14
AU2010210593A8 (en) 2015-09-03
AU2010210593A1 (en) 2011-08-25
RU2011134316A (en) 2013-03-10
RU2538199C2 (en) 2015-01-10
AU2010210593B2 (en) 2015-09-03
US20120189571A1 (en) 2012-07-26
US20160367682A1 (en) 2016-12-22
EP2393854B1 (en) 2019-12-04
CN102388073A (en) 2012-03-21
CN105295058B (en) 2018-09-21
WO2010091192A3 (en) 2010-12-02
CN109293927A (en) 2019-02-01
JP2016006068A (en) 2016-01-14
JP2012516898A (en) 2012-07-26
US9393227B2 (en) 2016-07-19
CN102388073B (en) 2015-11-25
BRPI1008869A2 (en) 2016-03-15
JP5844642B2 (en) 2016-01-20
HK1167869A1 (en) 2012-12-14
CN105295058A (en) 2016-02-03
EP2393854A4 (en) 2015-12-02
US10512696B2 (en) 2019-12-24

Similar Documents

Publication Publication Date Title
US10512696B2 (en) Nanoscale platinum compounds and methods of use thereof
CN106265510B (en) The multistage target polymer micella and preparation method thereof of pH trigger-type drug release in a kind of tumour cell
US8603532B2 (en) Nanostructures for drug delivery
CA2864950C (en) Functional pla-peg copolymers, the nanoparticles thereof, their preparation and use for targeted drug delivery and imaging
US10786465B2 (en) Polymer/copolymer nanoparticles conjugated to gambogic acid
Sunoqrot et al. Facile synthesis and surface modification of bioinspired nanoparticles from quercetin for drug delivery
CN113365613A (en) Conjugates and nanoparticles of hyaluronic acid and epigallocatechin-3-O-gallate and uses thereof
JP4991563B2 (en) Dosage form in which hydrophobic anticancer agent is encapsulated inside bile acid-chitosan complex and method for producing the same
AU2016326747A1 (en) Drug formulation based on particulates comprising polysaccharide-vitamin conjugate
Tabasi et al. Metal–polymer-coordinated complexes as potential nanovehicles for drug delivery
Wang et al. A reduction-degradable polymer prodrug for cisplatin delivery: preparation, in vitro and in vivo evaluation
WO2009145594A2 (en) Drug delivery carrier
CN102051016A (en) Degradable amphiphilic triblock copolymer micelle and preparation method and application of degradable amphiphilic triblock copolymer micelle
AU2015215843B2 (en) Nanoscale platinum compounds and methods of use thereof
Paraskar et al. Nanoplatinates for Breast Cancer
Paraskar Nanoplatinates for Breast Cancer PRINCIPAL INVESTIGATOR
TWI459975B (en) Pharmaceutical composition with a comb polymeric micelle, production method and use of the composition
US11969438B1 (en) Polyols and polyol-based hydrogels with anti-cancer activity
KR101935497B1 (en) Anti-cancer drug Effective for Multidrug resistant cancer cells
US20130259944A1 (en) Methods and compositions for treating cancer with platinum particles
Bhattacharyya et al. Synthesis of 1, 2, 3-triazole linked 5 fluorouracil-carbon dots-folate conjugates for target specific anticancer activity and cell imaging applications
WO2002043770A2 (en) Homing peptide multimers, their preparation and uses

Legal Events

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

Ref document number: 201080015384.4

Country of ref document: CN

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

Ref document number: 10739130

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2011549262

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 6198/DELNP/2011

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2010210593

Country of ref document: AU

Date of ref document: 20100204

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2010739130

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2011134316

Country of ref document: RU

WWE Wipo information: entry into national phase

Ref document number: 13147751

Country of ref document: US

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: PI1008869

Country of ref document: BR

ENP Entry into the national phase

Ref document number: PI1008869

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20110804