WO2012019049A1 - Devices, systems, and methods for excavating cancer cells - Google Patents

Devices, systems, and methods for excavating cancer cells Download PDF

Info

Publication number
WO2012019049A1
WO2012019049A1 PCT/US2011/046653 US2011046653W WO2012019049A1 WO 2012019049 A1 WO2012019049 A1 WO 2012019049A1 US 2011046653 W US2011046653 W US 2011046653W WO 2012019049 A1 WO2012019049 A1 WO 2012019049A1
Authority
WO
WIPO (PCT)
Prior art keywords
tumor
migration
nanofibers
tumor cells
substrate
Prior art date
Application number
PCT/US2011/046653
Other languages
French (fr)
Inventor
Ravi V. Bellamkonda
Anjana Jain
Original Assignee
Georgia Tech Research Corporation
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 Georgia Tech Research Corporation filed Critical Georgia Tech Research Corporation
Priority to AU2011285639A priority Critical patent/AU2011285639B2/en
Priority to JP2013523347A priority patent/JP5894989B2/en
Priority to CA2807483A priority patent/CA2807483C/en
Priority to EP11815346.9A priority patent/EP2600792B1/en
Priority to US13/814,009 priority patent/US11273250B2/en
Publication of WO2012019049A1 publication Critical patent/WO2012019049A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0087Galenical forms not covered by A61K9/02 - A61K9/7023
    • A61K9/0092Hollow drug-filled fibres, tubes of the core-shell type, coated fibres, coated rods, microtubules or nanotubes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M5/00Devices for bringing media into the body in a subcutaneous, intra-vascular or intramuscular way; Accessories therefor, e.g. filling or cleaning devices, arm-rests
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/05Phenols
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4355Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having oxygen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/443Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with oxygen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/34Macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyesters, polyamino acids, polysiloxanes, polyphosphazines, copolymers of polyalkylene glycol or poloxamers
    • 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
    • A61K47/593Polyesters, e.g. PLGA or polylactide-co-glycolide
    • 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
    • A61K47/62Medicinal 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 a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/6435Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the peptide or protein in the drug conjugate being a connective tissue peptide, e.g. collagen, fibronectin or gelatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/70Web, sheet or filament bases ; Films; Fibres of the matrix type containing drug
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/04Macromolecular materials
    • A61L31/06Macromolecular materials obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/08Materials for coatings
    • A61L31/10Macromolecular materials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/14Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L31/16Biologically active materials, e.g. therapeutic substances
    • 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
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/416Anti-neoplastic or anti-proliferative or anti-restenosis or anti-angiogenic agents, e.g. paclitaxel, sirolimus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2400/00Materials characterised by their function or physical properties
    • A61L2400/12Nanosized materials, e.g. nanofibres, nanoparticles, nanowires, nanotubes; Nanostructured surfaces

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Vascular Medicine (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Surgery (AREA)
  • Biomedical Technology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nanotechnology (AREA)
  • Inorganic Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Anesthesiology (AREA)
  • Hematology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Polymers & Plastics (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Materials For Medical Uses (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Prostheses (AREA)

Abstract

Methods, devices, and systems are provided for guiding tumor movement, particularly in vivo for treatment of patients. The method may include implanting into a tissue site where tumor cells are present a device having one or more surface structures or substrates, such as aligned nanofibers, which provide physical guidance cues for directing the migration of the tumor cells from the first tissue location to a selected second location, for tumor cell extraction or death. The devices and systems may include a cytotoxic agent for contacting tumor cells migrated via the substrate. All or a portion of the at least one substrate may include one or more biochemical cues, such as a coating of laminin or another protein, which may be provided in a concentration gradient to facilitate uni -directional tumor cell migration.

Description

DEVICES, SYSTEMS, AND METHODS FOR EXCAVATING CANCER CELLS
Cross-Reference to Related Applications
Priority is claims to U.S. Provisional Application No. 61/370,630, filed August 4, 20.10. The application is incorporated herein by reference.
Background
The present application relates to implantable medical devices, systems, and.
methods for treatment, of cancer, in particular, the present, application relates to systems and methods for directing cancer eel! migration in order to remove, relocate, or manage the growth of tumors, including tumors that would otherwise be inoperable or lead to a recurrence of the tumor.
Medul!oblastomas are highly invasive tumors of the cerebellum and the most common childhood malignant brain tumor, constituting 20-40% of all pediatric brain tumors. Treatment of these invasive intracranial brain tumors in children provides significant challenges thai are further complicated by the confined space and need to preserve as much non-cancerous, "normal" tissue as possible to avoid long-term cognitive dysfunction. In such cases, surgery is complicated and chemotherapy is prone to -major side effects because cycototoxic drugs cannot differentially kill invading tumor cells surrounded by normal cells.
Malignant gliomas also are among the most aggressive and least successfully treated types of cancer, with few patients surviving longer than a year following diagnosis. This bleak prognosis is largely attributed to the uniquely invasive ability of gliomas cells to detach from the tumor mass, infiltrate normal brain tissue, evade immunodetection and resist normally cytotoxic therapies. The invasion of these tumors prevents complete surgical removal and contributes to recurrence and a rapid, lethal outcome.
Studies have shown that the invasion of malignant gliomas predominantly occurs along major elongated structures, such as white matter fiber tracts and blood vessels, which act as '"highways"' for the spread of these tumors. It is generally believed that the myelin along the white matter fiber tracts aids with the adhesion and migration of the glioma cells. In addition, other proteins in the basement membrane have been implicated in association with migration along the blood vessels. Accordingly, there remains a need to develop an alternative treatment for meduUobiastomas and other malignancies, such as malignant gliomas.
Brief Description of the Drawings
FIG. 1 is a schematic showing different embodiments of an implantable system thai includes a substrate with a film of a plurality of aligned nanofibers.
FIG. 1A are scanning electron micrograph images of uniaxiaSly aligned nanofibers, with the magnified nanofi bers having a scale bar = 1 μm.
FIGS. 2A and 2B are cross-sectional views of a schematic of one embodiment of a tubular construct having a plurality of aligned nanofibers. FIG. 2A illustrates a tubular conduit with an aligned nanofiber film conduit while FIG. 2B illustrates a tubular conduit having a spiral aligned nanofiber film.
FIG. 3 is a perspective view of a schematic of an implantable system having a tubular conduit with a plurality of aligned nanofibers and a cytotoxic polymeric sink according to an embodiment.
FIG. 4 is a schematic illustration of a method for treating a patient according to an embodiment.
FIGS. 5A-5.D are images of tumor ceils seeded on a smooth film (A/B) or an aligned nanofiber film (C/D) taken 2 hours after seeding (A/C) or 10 days after seeding
(B/D).
FIG. 6 is a graph illustrating the tumor cell .migration of cells seeded on aligned nanofibers as compared to a smooth film.
FIGS. 7.4-7F are bright field and .fluorescent images of tumor cells cultured with a cyciopamrne conjugated collagen hydrogel 'sink' (A-C) and an apoptotic 'sink' (D-F). FIGS. 7B and 7E show live tumor cells stained with catcein. FIGS. 7C and 7F show dead tumor cells stained with ethidium homodimer.
FIG. 8 is a sectioned image of a rat having a single scaffold implanted or three scaffolds implanted ill ustrating the ability of the scaffolds to promote the directional migration of the tumor cells through the scaffolds.
Summary
One embodiment of the present description includes an implantable system for promoting tumor ceil migration for cell reraoval or death. The systera comprises at least one substrate having a surface configured to prov ide cues for directing tumor ce!i migration a!ong the substrate surface, the at least one substrate comprising a plurality of aligned nanofibers. Desirably, the system further comprises at least one cytotoxic agent for contacting tumor ceils migrated via the at least one substrate.
In another aspect, an implantable device is provided for promoting tumor cell migration for cell removal or death. The device comprises at least one film having a surface configured to provide cues for directing tumor ceil migration along the substrate surface, wherein the surface comprises a coating material gradient to effect uni-direclional or bi-direction growth of cells across the surface.
In still another aspect, a method is provided for guiding tumor movement in vivo. The method comprises implanting into a tissue site where tumor ceils are present a device having one or .more surface structures which provide physical guidance cues for directing the migration of the tumor cells from the first tissue location to a selected second location.
In another aspect a method is provided for treating a patient comprising implanting at a tissue site in the patient a dev ice comprising a substrate having a surface configured to provide cues for directing tumor cell migration along the substrate surface, the substrate comprising a plurality of aligned nanofibers. The method further comprises subsequently killing or removing tumor cells thai have migrated along the substrate surface.
Detailed Description
Embodiments of the present description address the above-described needs by providing implantable devices and systems that offer mi gratory tumors an alternate, preferential pathway for migration - a pathway that ultimately leads to their death or removal. In particular, innovative implantable devices and systems are provided that advantageously exploit the properties and mechanics of cell motility and migration characteristic of metastasis to manage growth of malignant tumors, particularly in vasive malignant brain tumors, lire implantable structures beneficially can be used to guide tumor extraction and tumor movement from one location (such as an inoperable location) to a secondary location (such as an operable location or cytotoxic sink).
In an embodiment, the implantable device for promoting tumor cell migration comprises a substrate having a surface configured to provide cues for directing tumor cell migration along the substrate surface. The implantable device desirably is sized and shaped so thai it may be imp! anted at or near malignant tumors using minimally invasive techniques, In particular embodiments, the implantable devices provide an attractive alternative for migrating tumors ceils; promote unidirectional migration of tumors; and provide a 'sink" to either collect, capture and/or kill the migrating tumor cells. In particular, the topographical or biochemical cues promote migration of the tumor cells into the implantable device while minimizing or eliminating diffusion of any such cues, thereby maintaining the stability of the primary tumor location and mitigating or avoiding any unintended contributions to tumor migration.
Embodiments of the present description generally comprise a substrate having a surface configured to provide physical and/or chemical cues for directing (guiding) tumor eel! migration along the substrate surface, in various embodiments, the physical guidance cues incl ude one or more of substrate topography features, such as grooves, and films comprised of arrays of nanofiber or niierofiber. such as aligned nanofibers.
In addihon to guiding such tumor movement by physical topographical guidance cues, the methods and devices described herein optionally may be used in combination with other guidance means, such as electric fields, chemoattractants, and cell seeding that may serve to modulate or enhance the tumor cell movement or extraction. Such other guidance means may be known in the art, although not applied in the context of tumor cell extraction or movement in vivo.
In a particular embodiment, the substrate comprises a plurality of aligned nanofibers. The plurality of aligned nanofibers may be in the form of a nanofiber film, a tubular construct, or any other suitable three-dimensional configuration. (See FIG. I ). In particular embodiments, the plurality of aligned nanofibers are in the form of a single nanofiber film disposed in a tubular construct. (See FIGS. 1, 2A, ami 2B). Such tubular constructs may, for example, provide structural support for the nanofiber films.
Alternatively, the tubular construct can also contain two or more nanofiber films. The two or more films may be stacked on top of each other, optionally with a spacer material therebetween, to provide multiple surfaces on which the tumor cells may migrate. (See FIG. 1). In still other embodiments, the nanofiber films can be intricately designed such that one or more of the films can be inserted into the tumor and then converge at points outside the tumor where the cells migrate, and may be subsequently be removed or killed. As used herein, the terms "nanofiber" refers to a fiber, strand, fibril, or threadlike structure having a diameter from about 40 nm to about 1500 nm. As used herein, the term "nanofilament" is synonymous with "nanofiber." in an embodiment, the nanofibers have a diameter from about 200 nm to about 1000 am, from about 400 nm to about 1000 nm, from about 500 to about 800 nm, or from about 600 to about 800 nm.
In one embodiment in which the substrate surface comprises grooves thai provide the physical cues for tumor ceil migration. For example, the grooves may he dimensioned to be approximately the same width and approximately the depth, or half the depth, as the diameter the nanofibers.
As used herein, the term "aligned nanofibers" refers to nanofibers having a uniaxial orientation. As used herem, the term "uniaxial orientation" refers to a collection of nanofibers where greater than 50% of the nanofibers are oriented within 40° of an axis, i.e., .±20º of the axis. Importantly, the nanofibers are oriented in the structure over several millimeters in length, e.g., between 2 and 100 mm. in an embodiment, at least 60%, at least 75%, or at least 85%, of the nanofibers are within 20 degrees of the uniaxial orientation. Such uniaxiaily aligned nanofibers are illustrated in FIG. 1A.
As used herein, the term "implantable device" means that the device is suitable for use in vivo, i.e., by implantation into a patient in need of treatment, for example treatment of a malignant tumor, such as a medulloblastoma or metastatic glioma. In an embodiment, the device is used in the treatment of other types of malignances. The implantation site may be in the brain of the patient.
The nanofibers may be formed from at least one polymer using methods known in the art. The nanofibers may be composed of synthetic or natural polymers or a mixture of synthetic and natural polymers. The polymer may be biodegradable or nonbiodegradable, or may include a mixture of biodegradable and non-biodegradable polymers. In particular embodiments, the nanofibers desirably comprise a biodegradable synthetic po!ymer. For example, in an embodiment, the polymer is a biocompatible, thermoplastic polymer such as a polyester or poly amide suitable for use in in vivo applications in humans.
Representative examples of synthetic polymers include poly (hydroxy acids) such as poly (lactic acid), poly (g!y colic acid), and poiy( lactic acid-co-glyco!ic acid), poly (lactide), polyiglyeolide), poly (lactide-co-giycolide), polyanhydrides, poSyorthoesters, poly amides, polyalkylenes such as polyethylene and polypropylene, polyalkylene glycols such as poly(ethylene glycol), polyalkylene oxides such as poly(ethylene oxide), polyvinyl alcohols, polyvinyl ethers, polyvinyl esters, polyvinylpyrrolidone, poly (vinyl alcohols). polyCbutyric acid), poly(vaieric acid), and poly(lactide-cocaprolactone), copolymers and blends thereof. As used herein, "derivatives" include polymers having substitutions, additions of chemical groups, for example, aikyl, alkylene, hydroxylations, oxidations, and other modifications routinely made by those skilled in the art. Exemplary biodegradable polymers include polymers of hydroxy acids such as lactic acid and gly colic acid, and copolymers with polyethylene glycol (PEG), polyanhydrides, poly(ortho)esters, poly(butyric acid), poly(valeric acid), poly(laclide-co-caprolactone), blends and copolymers thereof. Desirably, the biodegradable polymer nanofibers include a po!yicaproiactone), a poly(lactic-co-gly colic acid), a poly (aery lonitrile),or a combination thereof.
Representative examples of suitable natural polymers include proteins such as albumin, collagen, gelatin, Matrigel, fibrin, polypeptide or self-assembling peptide-based hydrogels. and prolamines, for example, zein, arid polysaccharides such as alginate, agarose, cellulose and polyliydroxyalkanoates, for example, poly hydroxybuty rale.
The structure of the implantable device may include a film of uniaxially oriented nanofibers or a plurality of such films in a stack. (FIG. 1). In one embodiment, each Sim layer is about 10 pm thick. Thicker or thinner layers may also be used; however, the thickness typically is selected to be one capable of handling and manipulation to stack or otherwise assemble the implantable device. For example, the film thickness may enable manual handling, such as to facilitate separation from a mandrel or other (temporary) substrate on which the nanofibers are electrospun. In embodiments comprising a plurality of stacked nanofiber films, each layer may be oriented such that the nanofiber orientation in the stack is essentially the same (e.g., such that the axial direction of all layers is pointing in substantially the same direction) or such that the nanofiber orientation of each layer in the stack is offset (e.g., such that the axial direction of each layer is substantially perpendicular).
Optionally, the stacked structure includes a spacer between some or all of the layers of uniaxially oriented nanofiber films. The spacer may be water soluble or water insoluble, porous or non-porous, preferably is biocompatible, and may be
bioerodible/hiodegradabie. The spacer may have a thickness between about 25 and about 800 μτη. In an embodiment, each spacer layer in the stack has a thickness of about 50 to about 250 pm. in m embodiment,, the spacer includes a hydrogel such as a thermo- reversible (i.e., temperature responsive) hydrogel. in one embodiment, the structure consists of alternating layers of oriented nanofibers and layers of a hydrogel or other spacer. The hydrogel, for instance, may he an agarose hydrogel or other hydrogel known in the art. In oilier embodiments, the spacer material may be another gel or gel-like material, such as polyethylene glycol, agarose, alginate, polyvinyl alcohol, collagen, Matrigei, chitosan, gelatin, or combination thereof,
in an alternative embodiment the uniaxially aligned nanofibers are provided in the structure in a form other than a plurality of layers. For example, the aligned nanofiber layers may be evenly spaced throughout the three-dimensional structure or a three- dimensional structure may be formed by rolling one layer, i.e., a film, of aligned nanofibers in on itself to form a tubular conduit (e.g., having an inner lumen, illustrated in FIG. 2A) or tubular scaffold (illustrated m FIG- 2B). The aligned nanofiber layers also may be engineered in any other suitabie configuration to facilitate surgical implantation or minimally invasive implantation of the device at a location proximate to the tumor.
The nanofibers structure optionally may be disposed in a secondary structure for containing, positioning, or securing the uniaxially oriented nanofiber structure, and/or for further directing or limiting tumor ceil growth. The secondary structure may also aid in shielding healthy tissues from contact with the migrating tumor cells.
in one embodiment, the secondary structure may be a tubular conduit, in which the nanofiber film(s) can be contained. This structure desirably also is made of a
biocompatible polymer suitable for use in vivo. The polymer may be biodegradable or non-biodegradable, or a mixture thereof. In one embodiment, the secondary structure comprises a polysulfone, polycaprolactone, polyurethatie. or PLGA. in a particular embodiment, the secondary structure is a tubular conduit made of polysulfone, polycaprolactone, polyurethane, PLGA, or a combination thereof. The secondary structure may be substantially .flexible or rigid, depending upon its particular performance needs.
The nanofibers described herein may be made by essentially any technique 'known in the art. Desirably, the nanofibers are made using an electrospinning technique using essentially any biocompatible polymer that is amenable to electrospinning. The eiectrospinniog equipment may include a rotating drum or other adaptation at the collector end to generate fibers oriented in the millimeter range.
In certain embodiments, the implantable devices provided herein further comprise biochemical cues to provide cues for directional migration of the tumor cells. For example, in embodiments the biochemical cue may comprise a coating of the plurality of uniaxially aligned nanofibers or nanofiber film with one or more bioactive agents capable of promoting directional migration of the tumor cells. Such coatings may be applied to the substrate using methods known to those skilled in the art. including, for example, nano- iiikjet printing.
As used herein, the term "bioactive agent" refers to a molecule thai exerts an effect on a ceil or tissue. Representative examples of types of bioactive agents incl ude therapeutics, vitamins, electrolytes, amino acids, peptides, polypeptides, proteins, carbohydrates, lipids, polysaccharides, nucleic acids, nucleotides, polynucleotides, glycoproteins, lipoproteins, glycolipids, glyeosaminoglyeans, proteoglycans, growth factors, differentiation factors, hormones, neurotransmitters, prostaglandins,
immunoglobulins, cytokines, and antigens. Various combination of these molecules can be used. Examples of cytokines include macrophage derived chemokines, macrophage inflammatory proteins, inter!eukins, tumor necrosis factors. Examples of proteins include fibrous proteins (e.g., collagen, eiastin) and adhesion proteins (e.g.. actin, fibrin, fibrinogen, fibronectin, vitronectin, laminin, cadherins. se.lecti.ns, intracellular adhesion molecules, and integrins). In various cases, the bioactive agent may be selected from fibronectin, laminin, thrombospondin, tenascin C, leptin, leukemia inhibitory factors. ROD peptides, antiTNFs, endostatin, angioslatin, thronibospondin, osteogenic protein- L bone morphogenic proteins, osteonectin, somaiomedin-like peptide, osteocalcin, interferons, and interleukins.
In particular embodiments, the bioactive agent comprises proteins, small molecules or biopolymers which have migration-promoting effects. Non-limiting examples of such bioactive agents include myelin or basement membrane proteins, such as laminin, collagen, and Mairigei. in embodiments, the bioactive agent, comprises different proteins and molecules that are attractive to various tumor cells, in other aspects, the bioactive agent comprises charged elements such as polyiysine or biopolymers. such as chitosan, capable of promoting migration of tumor ceils away from the tumor in a directed manner. in embodiments, the above-described biochemical cues may be applied to the plurality of nanofibers uniformly or in a gradient. Such gradients may be increasing or decreasing in the concentration or mass of the bioactive agent per gi ven area. (See FIG. 4). in certain embodiments, the gradient aids in promoting one-directionai migration of the tumor cells. In another embodiment, the gradient promotes bi-directional migration of cells. For instance, the gradient may comprise a higher concentration in one direction and a lower concentration in another direction to promote migration of two different cell types in opposite directions. Such bi-directional promoting gradients could limit tumor cell migration in one direction along the plurality of nanofibers away from the tumor site, while promoting migration of other ceil types to the tumor site. Not wishing to be bound by any theory, it is believed that by migration of non-tumor cells to the tumor site, such as ceils that elicit immune system sensiti vity in tumors, could function to direct a natural immune response to the tumors. In certain embodiments, the immune-promoting cells could be genetically altered to remove the immune privilege status of the tumor.
Although the foregoing embodiments describe the preferred use of nanofibers, those skilled in the art should appreciate that the embodiments of the implantable devices provided herein aiso may be prepared using other types of materials engineered to promote directional migration of the tumor ceils. Thus, in particular embodiments non-nanofiber based substrates may be used to induce migration of tumor cells in a desired direction to a desired location. For example, gradients of bioactive agents can be created in other materials, such as hydrogels and polymeric films, along with other directional cues that may or may not be based on topographical guidance of cells from the tumor to a region where the ceils may be killed or removed.
The implantable devices desirably further comprise a cytotoxic agent. The cytotoxic agent may be tethered or conjugated directly to the plurality of nanofibers or a polymeric "sink\ (See FIG, 3).
As used herein, "xycoioxic agenC means an apoptosis-inducing drug capable of inducing programmed cell death of tumor cells, in embodiments, the cytotoxic agent can be specific to a mutated signaling pathway that caused the tumor growth, in other embodiments, the cytotoxic agent can be a drug that is cytotoxic to any ceils but winch the conjugation of the prevents healthy normal cells from being affected. In various examples, the cytotoxic agent may include cyclopamine, honokiol, furegrelate, doxorubicin, or a combination thereof, in one embodiment, the cytotoxic agent comprises cyciopamine, which inhibits the over-expressed Sonic Hedgehog pathway (i.e.. a type of mutated signaling pathway). Other cytotoxic agents are also envisioned.
As used herein, the polymeric "sink' describes a portion of the implantable device designed to receive tumor cells at a targeted secondary location remote from the primary tumor. As illustrated in FIG. 3. ihe polymeric sink desirably is disposed at, adjacent, or about a second end of the plurality of nanofibers distal to a first end of ihe plurality of nanofibers, the first end of the plurality of nanofibers being positioned into or substantially adjacent to at least a portion of the tumor. For example, a first end of the plurality of nanofibers may be disposed into or substantially adjacent to a tumor that is disposed m an inoperable area by using a caiheter without risking the life of the patient. The plurality of nanofibers directs tumor migration to the polymeric "sink', which is placed in a more accessible region for removal or surgical resection. Non-limiting examples of materials suitable for use as the polymeric sink include various polymeric and biopolymeric thin films, including PAN-MA, PVA, PMMA, chitosan, laminin, collagen, etc., or hydrogels, such as agarose, Matrigel, Neuroge!, collagen, chitosan, or a composite of various hydrogels. in particular embodiments, the polymeric sink is disposed in a pouch made of a biocompatible material (e.g., thai will not induce an immune response). Non-limiting examples of the pouch material include Teflon. The sink may be in communication with one or a plurality of implanted devices. One example of this is illustrated in FIG. 4.
Hie implantable devices provided herein desirably are quite durable and are capable of maintaining their integrity and topography upon implantation into a patient in need. Desirably, the implantable devices are sized and shaped such that they may be implanted using minimally invasive techniques. For example, in particular embodiments, a large-gauge needle, catheter, or trochar can be used to deploy the implantable device within the tumor with minimal disruption to the surrounding tissue. Alternatively, the implantable device may be implanted in an open surgical procedure.
The tumor ceils that migrate to a secondary location from the primary tumor site by means of the implantable device may be dealt with using various methodologies. For example, in embodiments surgical resection can be used to remove the tumor cells from the secondary location which is more accessible than the primary tumor site. In other embodiments, a cytotoxic agent can be used in ihe polymeric sink to kill the tumor cells without requiring removal of the tumor cells. Thus, embodiments of the present description provide a novel and innovative method for treating malignancies, particularly those malignancies that are inoperable or inaccessi ble to deli very of cytotoxic agents.
The present description may be further understood with reference to tire following non-limiting examples.
Examples
In Vitro Experiments
Fabrication and Characterization of Aligned Nanofiber Films for Tumor Cell Migration Slow degrading nanofibers films were fabricated from a poly(caprolactone) (PCL) polymer using a electrospinniiig processes known to those skilled in the art. 'The nanofiber films were coated with the extraceliular matrix protein, laminin, to promote ceil migration. Larmnin has been shown to be in higher concentration at the periphery of the tumor core, suggesting that the protein may have a significant role in tumor cell migration.
To determine the effect of the topographical cues provided by the aligned nanofibers on tumor cell migration, an experiment was conducted to compare tumor ceil migration on aligned nanofibers films to smooth films. The results, illustrated in FIGS. 5- 6, demonstrate that the aligned nanofiber films promoted significantly higher tumor cell migration as compared to the smooth films as evidenced by the distance of migration. (FIG. 5: tumor ceils imaged 2 hours alter seeding on a smooth film (A), tumor cells imaged 10 days after seeding on a smooth film (B), tumor cells imaged 2 hours after seecing on aligned nanofibers (C), and tumor cells imaged 10 days after seeding on aligned nanofibers (D); FICi, 6. qualitative analysis of tumor cell migration on the aligned nanofibers as compared to smooth film).
Fabrication and Characterization of Apopiotic Hydrogel Sink
The smoothened inhibitor, cyclopamine. was evaluated to determine the effective drug concentration required to induce apoptosis in tumor cells. The viability of the tumors cells was measured at different concentrations of cyclopamine. Ilea! thy cells (e.g., neurons and glia) were not affected by exposure to the drug. However, the results suggested that the collagen hydrogel scaffold should be conjugated with 30 uM of cyclopamine.
A nanofiber film having a cytotoxic sink was prepared by conjugating the cyclopamine to the backbone of a collagen hydrogel. A crosslinker N,N'- carbonyldiimidazole was used to link the hydroxy] group on the cyciopamine with an amine group on the collagen. To verify whether ihe cyciopamine was conjugated to the collagen hydrogel. C13 NMR was performed. Three conditions, cyciopamine, cyciopamine conjugated to collagen, and cyciopamine and collagen scaffold without the crosslinker were analyzed using C 13 NMR. A third condition was included to determine if indeed the cyciopamine was tethered to the hydrogel rather than entrapped by the collagen. Cyciopamine has four carbons that are involved in double bonds, which appear between 150 and 120 ppnv The four peaks for the four carbons in the double bonds in cyciopamine were present in the cyciopamine only spectra and in cyciopamine tethered to collagen; however, ihe peaks were absent when cyciopamine was mixed with the collagen hydrogel.
Tumor cells were cultured within the cyciopamine conjugated collagen hydrogel sink to determine whether the bioactivity of cyciopamine was diminished and i f the ceils underwent apoptosis. As illustrated in FIG. 7, bright field and floureseent images of the cells clearly underwent apoptosis when they were cultured in the apoptoiic sink (D-F) as compared to a cyciopamine conjugated collagen hydrogel sink (A-C). (bright field images (A/D), live tumor cells stained with calcein (B/E). dead tumor cells stained with ethidium horaodimer (C/F)). Notably, the number of dead cells in the cyciopamine conjugated collagen hydrogels was higher than the number of dead cells in the collagen only hydrogels.
Based on the foregoing in vitro experiments, ii was determined that the tumor cells did indeed migrate further with topographical cues and that an apoptosis inducing sink may be effective to kill the tumor cells. Accordingly, preliminary in vivo experiments were conducted to evaluate the effectiveness of embodiments of the implantable scaffolds described herein at promoting directed tumor cell migration.
In Vivo Experiments
24 Rowett Nude Rats were inoculated with U87mg -GFP cells, which is a human glioblastoma cell line. Seven days after the animals were inoculated with the tumor cells, 5 mm scaffolds were implanted into the brain near the tumor site. Hie tumors were inoculated 2 mm deep from the surface of the brain. The conduits were implanted 1 .5 mm from the surface of the brain. The scaffolds comprised a conduit, made of 10% polycaprolactone (PCX) and polyurethane. Within the conduit PCL aligned nanofibers were inserted. Either a single scaffold or three scaffolds were implanted into the rats. The animals were perfused when they displayed symptoms from the tumors. From Day 16 to Day 18 afier inoculation, the rats were perfused and the brains were dissected out with the scaffold. The scaffolds and brains were sectioned in a transverse orientation obtaining 50 μ,ιτι sections. The sections were imaged after they had been sectioned. The tumor cells were visible due to their GFP expression. As illustrated in FIG. 8, the scaffolds were effectiv e at promoting the directional migration of the tumor cells through the scaffolds.
Publications cited herein and the materials for which they are cited are specifically incorporated by reference. Modifications and variations of the methods and devices described herein will he obvious to those skilled in the art from the foregoing detailed description. Such modifications and variations are intended to come within the scope of the appended claims.

Claims

Claims
1. An implantable system for promoting tumor ceil migration for cell removal or death, the system comprising:
at least one substrate having a surface configured, to provide cues for directing tumor cell migration along the substrate surface, the at least one substrate comprising a plurality of aligned na.nofibe.rs; and
at least one cytotoxic agent for contacting tumor cells migrated via the at least one substrate.
2. The system of claim 1, wherein the plurality of aligned nanofibers form a tubular
construct,
3. The system of claim 1 , wherein the plurality of aligned nanofibers are di sposed in a
tubular construct,
4. The system of claim 3, wherein the tubular construct comprises an annular tube formed from a po!ycaproiactone, a polyurethane, or a combination thereof.
5. The system of claim L wherein the at ieast one cytotoxic agent is tethered or conjugated to at least a portion of the plurality of aligned nanofibers.
6. The system of claim 1, wherein the at least one cytotoxic agent is tethered or conjugated to a polymeric sink material.
7. The system of claim 6, wherein the polymeric sink material comprises a hydrogel .
8. The system of claim L wherein the cytotoxic agent comprises eyclopamine, honokioi, furegreiate, doxorubicin, or a combination thereof,
9. The system of claim 15 wherein all or a portion of the at least one substrate comprises one or more biochemical cues to facilitate uni -directional tumor cell migration,
10. The system of claim L wherein the plurality of nanoiibers comprises a coating selected from the group consisting of extracellular matrix proteins, growth factors, cytokines, peptides, and combinations thereof,
1 1 . The system of claim 10, wherein the coating is disposed uniformly along the length of the plurality of aligned nanofibers.
12. The system of claim 10, wherein the coating is disposed in a concentration gradient along the length of the plurality of aligned nanoiibers from a first end of the plurality of aligned nanofibers to a second end of the plurality of aligned nanofibers, the second end being distal to the first end.
13. The system of claim 12, wherein the concentration gradient is effective to promote unidirectional migration of tumor ceils.
14. The system of claim 1 2, wherein the concentration gradient is effective to promote bidirectional migration of non-tumor cells.
15. The system of claim 1 , wherein the nanoiibers comprise a synthetic polymer.
16. The system of any one of claims 9 to 15, wherein the nanofiber films comprise a coating of myelin or a basement membrane protein.
1 7. The system of claim 1 6, wherein the nanofiber films comprise a !aminin coating.
18. The system of claim i . wherein the nanofibers have a diameter in the range of about 400 nm to about 800 urn .
19. The system of claim 1 , wherein the plurality of nanofibers are in the form of a nanofiber film having a. thickness in the range of 5 microns to 20 microns.
20. The system of any one of claim 1 to 19, wherein the plurality of aligned nanofibers are synthetic polymeric fibers having a diameter from 600 n.m to 800 nm, which are coated with an extracellular matrix protein having a concentration gradient along the length of the fibers,
21 . The system of claim 20, wherein the cytotoxic agent comprises collagen hydroge!
eovalently coupled to an apoptotic triggering agent.
22. The system of claim 21 , wherein the hydroge! is at least partially encased in a cell- imperm cable pouch.
23. An implantable device for promoting tumor ceil migration for ceM removal or death, the device comprising:
at least one film having a surface configured to provide cues for directing tumor ceil migration along the substrate surface, wherein the surface comprises a coating material gradient to effect uni -directional or bi -direct) on growth of ce!is across the surface.
24. The device of claim 23, further comprising a cytotoxic agent positioned for contacting tumor cells migrated via the surface.
30. A. method for guiding tumor movement in vivo, comprising:
implanting into a tissue site where tumor cells are present a device having one or more surface structures which provide physical guidance cues for directing the migration of the tumor ceils from the first tissue location to a selected second location.
31 The method of claim 30, wherein the surface structure comprises nanofibers or grooves The method of claim 30 or 31 , further comprising applying another guidance means at the tissue implantation site, wherein the guidance means comprises an electric field, one or more biochemical cue, or ceil seeding, and works in combination with the physical guidance cues of the surface structure to direct migration of the tumor cells to the selected second location.
The method of claim 30 or 31 , wherein the surface structure comprises a plurality of aligned nanofibers having a diameter from 400 am to 800 nm, which are coated with an extracellular matrix protein.
The method of any one of claims 30 to 33, wherein the tumor cells comprise
Medulloblastoma ceils or malignant glioma cells
A method for treating a patient comprising:
implanting at a tissue site in the patient a device comprising a substrate having a surface configured to provide cues for directing tumor cell migration along the substrate siiiface, the substrate comprising a plurality of aligned nanofibers; and subsequently killing or removing tumor cells that have migrated along the substrate surface.
The method of claim 35, wherein the tissue site comprises a tumor or a void of a resected tumor.
The method of claim 35, further comprising contacting tumor cells thai have migrated along at least a portion of the substrate with a cytotoxic agent.
'The method of claim 37, wherein the cytotoxic agent is part of the implanted device.
The method of claim any one of claims 35 to 38, wherein tumor cells are relocated from a first region to a second region from a primary tumor via the implantable device before the tumor cells are killed or removed.
PCT/US2011/046653 2010-08-04 2011-08-04 Devices, systems, and methods for excavating cancer cells WO2012019049A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
AU2011285639A AU2011285639B2 (en) 2010-08-04 2011-08-04 Devices, systems, and methods for excavating cancer cells
JP2013523347A JP5894989B2 (en) 2010-08-04 2011-08-04 Apparatus, system, and method for extracting cancer cells
CA2807483A CA2807483C (en) 2010-08-04 2011-08-04 Devices, systems, and methods for excavating cancer cells
EP11815346.9A EP2600792B1 (en) 2010-08-04 2011-08-04 Devices, systems, and methods for excavating cancer cells
US13/814,009 US11273250B2 (en) 2010-08-04 2011-08-04 Devices, systems, and methods for excavating cancer cells

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US37063010P 2010-08-04 2010-08-04
US61/370,630 2010-08-04

Publications (1)

Publication Number Publication Date
WO2012019049A1 true WO2012019049A1 (en) 2012-02-09

Family

ID=45559831

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/046653 WO2012019049A1 (en) 2010-08-04 2011-08-04 Devices, systems, and methods for excavating cancer cells

Country Status (6)

Country Link
US (1) US11273250B2 (en)
EP (2) EP2600792B1 (en)
JP (1) JP5894989B2 (en)
AU (1) AU2011285639B2 (en)
CA (1) CA2807483C (en)
WO (1) WO2012019049A1 (en)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013134352A1 (en) * 2012-03-06 2013-09-12 The Uab Research Foundation Aligned nanofibrous structures for axonal regeneration after spinal cord injury or surgery
EP2714073A1 (en) * 2011-06-03 2014-04-09 President and Fellows of Harvard College In situ antigen-generating cancer vaccine
WO2014083019A2 (en) * 2012-11-27 2014-06-05 Sergas Agent, product and use
US9132210B2 (en) 2005-12-13 2015-09-15 President And Fellows Of Harvard College Scaffolds for cell transplantation
US9297005B2 (en) 2009-04-13 2016-03-29 President And Fellows Of Harvard College Harnessing cell dynamics to engineer materials
US9370558B2 (en) 2008-02-13 2016-06-21 President And Fellows Of Harvard College Controlled delivery of TLR agonists in structural polymeric devices
US9381235B2 (en) 2009-07-31 2016-07-05 President And Fellows Of Harvard College Programming of cells for tolerogenic therapies
US9603894B2 (en) 2010-11-08 2017-03-28 President And Fellows Of Harvard College Materials presenting notch signaling molecules to control cell behavior
US9610328B2 (en) 2010-03-05 2017-04-04 President And Fellows Of Harvard College Enhancement of skeletal muscle stem cell engraftment by dual delivery of VEGF and IGF-1
US9675561B2 (en) 2011-04-28 2017-06-13 President And Fellows Of Harvard College Injectable cryogel vaccine devices and methods of use thereof
US9693954B2 (en) 2010-06-25 2017-07-04 President And Fellows Of Harvard College Co-delivery of stimulatory and inhibitory factors to create temporally stable and spatially restricted zones
US9770535B2 (en) 2007-06-21 2017-09-26 President And Fellows Of Harvard College Scaffolds for cell collection or elimination
US9821045B2 (en) 2008-02-13 2017-11-21 President And Fellows Of Harvard College Controlled delivery of TLR3 agonists in structural polymeric devices
EP3201314A4 (en) * 2014-09-26 2018-03-21 Rensselaer Polytechnic Institute Nanocomposite scaffold for the in vitro isolation of cells
US9937249B2 (en) 2012-04-16 2018-04-10 President And Fellows Of Harvard College Mesoporous silica compositions for modulating immune responses
US10045947B2 (en) 2011-04-28 2018-08-14 President And Fellows Of Harvard College Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration
US10111935B2 (en) 2014-04-30 2018-10-30 Fundaciön Pedro Barrié De La Maza, Condo De Fenosa Agent for capturing tumor cells and methods of use thereof
US10647959B2 (en) 2011-04-27 2020-05-12 President And Fellows Of Harvard College Cell-friendly inverse opal hydrogels for cell encapsulation, drug and protein delivery, and functional nanoparticle encapsulation
US10682400B2 (en) 2014-04-30 2020-06-16 President And Fellows Of Harvard College Combination vaccine devices and methods of killing cancer cells
US11150242B2 (en) 2015-04-10 2021-10-19 President And Fellows Of Harvard College Immune cell trapping devices and methods for making and using the same
US11202759B2 (en) 2010-10-06 2021-12-21 President And Fellows Of Harvard College Injectable, pore-forming hydrogels for materials-based cell therapies
US11555177B2 (en) 2016-07-13 2023-01-17 President And Fellows Of Harvard College Antigen-presenting cell-mimetic scaffolds and methods for making and using the same
US11752238B2 (en) 2016-02-06 2023-09-12 President And Fellows Of Harvard College Recapitulating the hematopoietic niche to reconstitute immunity
US11786457B2 (en) 2015-01-30 2023-10-17 President And Fellows Of Harvard College Peritumoral and intratumoral materials for cancer therapy

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107206121B (en) * 2014-11-13 2020-09-08 新加坡国立大学 Tissue scaffold devices and methods for making the same
US10493233B1 (en) * 2018-06-05 2019-12-03 Duke University Bi-directional access to tumors
CN115252589A (en) * 2020-09-27 2022-11-01 成都金瑞基业生物科技有限公司 Medical application of honokiol
CN115282341B (en) * 2022-06-27 2023-05-23 华南理工大学 Porous metal material with polypeptide bidirectional gradient distribution surface and preparation method and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030017141A1 (en) * 1999-04-08 2003-01-23 Poznansky Mark C. Purposeful movement of human migratory cells away from an agent source
WO2008137659A1 (en) * 2007-05-04 2008-11-13 University Of Virginia Patent Foundation Compositions and methods for making and using laminin nanofibers
US20100129418A1 (en) * 2008-11-18 2010-05-27 Lowell Jeffry Lawrence Method of inducing negative chemotaxis using an ellagitannin or gallotannin

Family Cites Families (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2121286B (en) 1982-06-02 1985-11-06 Ethicon Inc Improvements in synthetic vascular grafts, and methods of manufacturing such grafts
US5217492A (en) 1982-09-29 1993-06-08 Bio-Metric Systems, Inc. Biomolecule attachment to hydrophobic surfaces
CA1233953A (en) 1984-03-30 1988-03-15 Robert D. Ainsworth Orthopedic device and method of making the same
US5053453A (en) 1988-11-01 1991-10-01 Baxter International Inc. Thromboresistant materials and methods for making same
USRE36370E (en) 1992-01-13 1999-11-02 Li; Shu-Tung Resorbable vascular wound dressings
US5916585A (en) 1996-06-03 1999-06-29 Gore Enterprise Holdings, Inc. Materials and method for the immobilization of bioactive species onto biodegradable polymers
US6347930B1 (en) 1997-09-11 2002-02-19 Hospal R & D Int. Device and method for manufacturing a segmented tubular capsule containing a biologically active medium
US6165217A (en) 1997-10-02 2000-12-26 Gore Enterprise Holdings, Inc. Self-cohering, continuous filament non-woven webs
US6303136B1 (en) 1998-04-13 2001-10-16 Neurotech S.A. Cells or tissue attached to a non-degradable filamentous matrix encapsulated by a semi-permeable membrane
JP4341049B2 (en) 1998-06-05 2009-10-07 オルガノジェネシス インク. Tubular graft prosthesis made by biotechnology
US20020081732A1 (en) 2000-10-18 2002-06-27 Bowlin Gary L. Electroprocessing in drug delivery and cell encapsulation
US7615373B2 (en) 1999-02-25 2009-11-10 Virginia Commonwealth University Intellectual Property Foundation Electroprocessed collagen and tissue engineering
US6716225B2 (en) 2001-08-02 2004-04-06 Collagen Matrix, Inc. Implant devices for nerve repair
US6893431B2 (en) 2001-10-15 2005-05-17 Scimed Life Systems, Inc. Medical device for delivering patches
US7622299B2 (en) 2002-02-22 2009-11-24 University Of Washington Bioengineered tissue substitutes
US20030175410A1 (en) 2002-03-18 2003-09-18 Campbell Phil G. Method and apparatus for preparing biomimetic scaffold
US8337893B2 (en) 2002-07-10 2012-12-25 Florida Research Foundation, Inc, University Of Sol-gel derived bioactive glass polymer composite
US20050038498A1 (en) 2003-04-17 2005-02-17 Nanosys, Inc. Medical device applications of nanostructured surfaces
US7163920B2 (en) 2003-09-30 2007-01-16 Ethicon, Inc. Peptide with osteogenic activity
US7704740B2 (en) 2003-11-05 2010-04-27 Michigan State University Nanofibrillar structure and applications including cell and tissue culture
US20060085063A1 (en) 2004-10-15 2006-04-20 Shastri V P Nano- and micro-scale engineering of polymeric scaffolds for vascular tissue engineering
WO2006096791A2 (en) 2005-03-07 2006-09-14 Georgia Tech Research Corporation Nanofilament scaffold for tissue regeneration
US7531503B2 (en) 2005-03-11 2009-05-12 Wake Forest University Health Sciences Cell scaffold matrices with incorporated therapeutic agents
CN101410508B (en) 2006-01-27 2013-07-03 加利福尼亚大学董事会 Biomimetic scaffolds
US20080220042A1 (en) 2006-01-27 2008-09-11 The Regents Of The University Of California Biomolecule-linked biomimetic scaffolds
WO2008122044A2 (en) 2007-04-02 2008-10-09 Georgia Tech Research Corporation Implantable device for communicating with biological tissue
US9770535B2 (en) * 2007-06-21 2017-09-26 President And Fellows Of Harvard College Scaffolds for cell collection or elimination
US20090043380A1 (en) * 2007-08-09 2009-02-12 Specialized Vascular Technologies, Inc. Coatings for promoting endothelization of medical devices
US20090082856A1 (en) * 2007-09-21 2009-03-26 Boston Scientific Scimed, Inc. Medical devices having nanofiber-textured surfaces
US8940331B2 (en) * 2008-11-22 2015-01-27 The Board Of Trustees Of The Leland Stanford Junior University Hydrogels, methods of making hydrogels, methods of using hydrogels, and methods of isolating, trapping, attracting, and/or killing cancer cells
JP2012527217A (en) 2009-04-24 2012-11-08 ジ・オハイオ・ステート・ユニバーシティ Bidirectional microenvironment system
US20110038936A1 (en) 2009-08-17 2011-02-17 Kimberly Ann Griswold System and method for electrospun drug loaded biodegradable chemotherapy applications
US9572909B2 (en) 2009-09-14 2017-02-21 Virginia Commonwealth University Electrospun nerve guides for nerve regeneration designed to modulate nerve architecture

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030017141A1 (en) * 1999-04-08 2003-01-23 Poznansky Mark C. Purposeful movement of human migratory cells away from an agent source
WO2008137659A1 (en) * 2007-05-04 2008-11-13 University Of Virginia Patent Foundation Compositions and methods for making and using laminin nanofibers
US20100129418A1 (en) * 2008-11-18 2010-05-27 Lowell Jeffry Lawrence Method of inducing negative chemotaxis using an ellagitannin or gallotannin

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
JOHNSON ET AL.: "Quantitative Analysis of Complex Glioma Cell Migration on Electrospun Polycaprolactone Using Time-Lapse Microscopy.", TISSUE ENGINEERING: PART C., vol. 15, 2009, pages 531 - 541, XP055083103 *
LI ET AL.: "Nanofiber Scaffolds with Gradations in Mineral Content for Mimicking the Tendon-to-Bone Insertion Site.", NANO LETTERS, vol. 9, 2009, pages 2763 - 2768, XP055083107 *
SCHWALL ET AL.: "Micro- and Nanoscale Hydrogel Systems for Drug Delivery and Tissue Engineering.", MATERIALS., vol. 2, 2009, pages 577 - 612, XP055083106 *
See also references of EP2600792A4 *
XIE ET AL.: "Electrospun Micro- and Nanofibers for Sustained Delivery of Paclitaxel to Treat C6 Glioma in Vitro.", PHARMACEUTICAL RESEARCH., vol. 23, 2006, pages 1817 - 1826, XP019405186 *

Cited By (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9446107B2 (en) 2005-12-13 2016-09-20 President And Fellows Of Harvard College Scaffolds for cell transplantation
US11096997B2 (en) 2005-12-13 2021-08-24 President And Fellows Of Harvard College Scaffolds for cell transplantation
US10149897B2 (en) 2005-12-13 2018-12-11 President And Fellows Of Harvard College Scaffolds for cell transplantation
US10137184B2 (en) 2005-12-13 2018-11-27 President And Fellows Of Harvard College Scaffolds for cell transplantation
US9132210B2 (en) 2005-12-13 2015-09-15 President And Fellows Of Harvard College Scaffolds for cell transplantation
US10695468B2 (en) 2007-06-21 2020-06-30 President And Fellows Of Harvard College Scaffolds for cell collection or elimination
US9770535B2 (en) 2007-06-21 2017-09-26 President And Fellows Of Harvard College Scaffolds for cell collection or elimination
US10568949B2 (en) 2008-02-13 2020-02-25 President And Fellows Of Harvard College Method of eliciting an anti-tumor immune response with controlled delivery of TLR agonists in porous polymerlc devices
US9821045B2 (en) 2008-02-13 2017-11-21 President And Fellows Of Harvard College Controlled delivery of TLR3 agonists in structural polymeric devices
US10258677B2 (en) 2008-02-13 2019-04-16 President And Fellows Of Harvard College Continuous cell programming devices
US9370558B2 (en) 2008-02-13 2016-06-21 President And Fellows Of Harvard College Controlled delivery of TLR agonists in structural polymeric devices
US10328133B2 (en) 2008-02-13 2019-06-25 President And Fellows Of Harvard College Continuous cell programming devices
US9297005B2 (en) 2009-04-13 2016-03-29 President And Fellows Of Harvard College Harnessing cell dynamics to engineer materials
US10080789B2 (en) 2009-07-31 2018-09-25 President And Fellows Of Harvard College Programming of cells for tolerogenic therapies
US9381235B2 (en) 2009-07-31 2016-07-05 President And Fellows Of Harvard College Programming of cells for tolerogenic therapies
US9610328B2 (en) 2010-03-05 2017-04-04 President And Fellows Of Harvard College Enhancement of skeletal muscle stem cell engraftment by dual delivery of VEGF and IGF-1
US9693954B2 (en) 2010-06-25 2017-07-04 President And Fellows Of Harvard College Co-delivery of stimulatory and inhibitory factors to create temporally stable and spatially restricted zones
US11202759B2 (en) 2010-10-06 2021-12-21 President And Fellows Of Harvard College Injectable, pore-forming hydrogels for materials-based cell therapies
US9603894B2 (en) 2010-11-08 2017-03-28 President And Fellows Of Harvard College Materials presenting notch signaling molecules to control cell behavior
US10647959B2 (en) 2011-04-27 2020-05-12 President And Fellows Of Harvard College Cell-friendly inverse opal hydrogels for cell encapsulation, drug and protein delivery, and functional nanoparticle encapsulation
US9675561B2 (en) 2011-04-28 2017-06-13 President And Fellows Of Harvard College Injectable cryogel vaccine devices and methods of use thereof
US10045947B2 (en) 2011-04-28 2018-08-14 President And Fellows Of Harvard College Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration
EP2714073A1 (en) * 2011-06-03 2014-04-09 President and Fellows of Harvard College In situ antigen-generating cancer vaccine
US10406216B2 (en) 2011-06-03 2019-09-10 President And Fellows Of Harvard College In situ antigen-generating cancer vaccine
EP2714073A4 (en) * 2011-06-03 2014-12-31 Harvard College In situ antigen-generating cancer vaccine
US9486512B2 (en) 2011-06-03 2016-11-08 President And Fellows Of Harvard College In situ antigen-generating cancer vaccine
WO2013134352A1 (en) * 2012-03-06 2013-09-12 The Uab Research Foundation Aligned nanofibrous structures for axonal regeneration after spinal cord injury or surgery
US9937249B2 (en) 2012-04-16 2018-04-10 President And Fellows Of Harvard College Mesoporous silica compositions for modulating immune responses
US11278604B2 (en) 2012-04-16 2022-03-22 President And Fellows Of Harvard College Mesoporous silica compositions comprising inflammatory cytokines comprising inflammatory cytokines for modulating immune responses
CN105120891A (en) * 2012-11-27 2015-12-02 塞加斯 Agent for modulating the dissemination of cancer cells, product comprising said agent, and use of said agent in the treatment and/or prevention of cancer
WO2014083019A3 (en) * 2012-11-27 2014-07-24 Sergas Agent for modulating the dissemination of cancer cells, product comprising said agent, and use of said agent in the treatment and/or prevention of cancer
WO2014083019A2 (en) * 2012-11-27 2014-06-05 Sergas Agent, product and use
US10022400B2 (en) 2012-11-27 2018-07-17 Fundacion Ramon Dominguez Methods of capturing tumor cells
US10682400B2 (en) 2014-04-30 2020-06-16 President And Fellows Of Harvard College Combination vaccine devices and methods of killing cancer cells
US10111935B2 (en) 2014-04-30 2018-10-30 Fundaciön Pedro Barrié De La Maza, Condo De Fenosa Agent for capturing tumor cells and methods of use thereof
US10392594B2 (en) 2014-09-26 2019-08-27 Renssealer Polytechnic Institute Nanocomposite scaffold for the in vitro isolation of cells
EP3201314A4 (en) * 2014-09-26 2018-03-21 Rensselaer Polytechnic Institute Nanocomposite scaffold for the in vitro isolation of cells
US11786457B2 (en) 2015-01-30 2023-10-17 President And Fellows Of Harvard College Peritumoral and intratumoral materials for cancer therapy
US11150242B2 (en) 2015-04-10 2021-10-19 President And Fellows Of Harvard College Immune cell trapping devices and methods for making and using the same
US11752238B2 (en) 2016-02-06 2023-09-12 President And Fellows Of Harvard College Recapitulating the hematopoietic niche to reconstitute immunity
US11555177B2 (en) 2016-07-13 2023-01-17 President And Fellows Of Harvard College Antigen-presenting cell-mimetic scaffolds and methods for making and using the same

Also Published As

Publication number Publication date
US20130172846A1 (en) 2013-07-04
US11273250B2 (en) 2022-03-15
AU2011285639A1 (en) 2013-02-21
EP2600792B1 (en) 2017-11-01
CA2807483C (en) 2019-02-26
EP2600792A4 (en) 2014-04-16
JP2013538079A (en) 2013-10-10
EP2600792A1 (en) 2013-06-12
EP3298986A1 (en) 2018-03-28
JP5894989B2 (en) 2016-03-30
AU2011285639B2 (en) 2014-12-18
CA2807483A1 (en) 2012-02-09

Similar Documents

Publication Publication Date Title
US11273250B2 (en) Devices, systems, and methods for excavating cancer cells
Muylaert et al. Early in-situ cellularization of a supramolecular vascular graft is modified by synthetic stromal cell-derived factor-1α derived peptides
Jiang et al. CO2-expanded nanofiber scaffolds maintain activity of encapsulated bioactive materials and promote cellular infiltration and positive host response
Dongargaonkar et al. Electrospun blends of gelatin and gelatin–dendrimer conjugates as a wound-dressing and drug-delivery platform
US8691543B2 (en) Nanofibrous scaffold comprising immobilized cells
ES2847893T3 (en) Biomedical patches with fibers arranged in space
CA2188948A1 (en) Device for delivery of substances and methods of use thereof
Chopra et al. Nanomaterials: a promising therapeutic approach for cardiovascular diseases
Barnum et al. 3D‐printed hydrogel‐filled microneedle arrays
JP2009524507A (en) Biomimetic scaffold
Zhu et al. Anti-neoplastic cytotoxicity of SN-38-loaded PCL/Gelatin electrospun composite nanofiber scaffolds against human glioblastoma cells in vitro
Qi et al. Electrospun fibers of acid-labile biodegradable polymers containing ortho ester groups for controlled release of paracetamol
JP2011156355A (en) Differential loading of drug-eluting medical device
Sa’adon et al. Drug-loaded poly-vinyl alcohol electrospun nanofibers for transdermal drug delivery: Review on factors affecting the drug release
Nune et al. Self-assembling peptide nanostructures on aligned poly (lactide-co-glycolide) nanofibers for the functional regeneration of sciatic nerve
US20220118160A1 (en) An implantable device and a method for implanting said device in a subject
Mancipe et al. Electrospinning: New strategies for the treatment of skin melanoma
Biazar Application of polymeric nanofibers in medical designs, part IV: drug and biological materials delivery
Zhu et al. In vitro and in vivo evaluation of Rapamycin-eluting nanofibers coated on cardia stents
JP7044288B2 (en) Medical sheet
EP3589243A1 (en) An intravascular cell therapy device
US20230218507A1 (en) Microneedle patch for in-situ seeding of cells
Schmitt Influence of neuroimplant materials, drugs and drug-material combinations on healthy cells of the brain
van Almen PhD et al. Early in-situ cellularization of a supramolecular vascular graft is modified by synthetic stromal cell-derived factor-1α derived peptides
CZ2015117A3 (en) Process or preparing flat textile fabric of biologically degradable and biologically compatible nanofibers, especially for skin wound covers and apparatus for making the same

Legal Events

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

Ref document number: 11815346

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2013523347

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2807483

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2011815346

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2011815346

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2011285639

Country of ref document: AU

Date of ref document: 20110804

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 13814009

Country of ref document: US