WO2014141289A1 - Photo - chemo composition on the basis of microcapsules with a core -shell structure - Google Patents

Photo - chemo composition on the basis of microcapsules with a core -shell structure Download PDF

Info

Publication number
WO2014141289A1
WO2014141289A1 PCT/IN2013/000144 IN2013000144W WO2014141289A1 WO 2014141289 A1 WO2014141289 A1 WO 2014141289A1 IN 2013000144 W IN2013000144 W IN 2013000144W WO 2014141289 A1 WO2014141289 A1 WO 2014141289A1
Authority
WO
WIPO (PCT)
Prior art keywords
core
shell
chemo
polymer
nanomedicine
Prior art date
Application number
PCT/IN2013/000144
Other languages
French (fr)
Inventor
Koyakutty Manzoor
Ramachandran Ranjith
M. Giridharan L.
V Nair Shantikumar
Original Assignee
Amrita Vishwa Vidyapeetham University
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 Amrita Vishwa Vidyapeetham University filed Critical Amrita Vishwa Vidyapeetham University
Priority to PCT/IN2013/000144 priority Critical patent/WO2014141289A1/en
Publication of WO2014141289A1 publication Critical patent/WO2014141289A1/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
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5146Organic macromolecular compounds; Dendrimers obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyamines, polyanhydrides
    • A61K9/5153Polyesters, e.g. poly(lactide-co-glycolide)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/0057Photodynamic therapy with a photosensitizer, i.e. agent able to produce reactive oxygen species upon exposure to light or radiation, e.g. UV or visible light; photocleavage of nucleic acids with an agent
    • A61K41/0071PDT with porphyrins having exactly 20 ring atoms, i.e. based on the non-expanded tetrapyrrolic ring system, e.g. bacteriochlorin, chlorin-e6, or phthalocyanines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5161Polysaccharides, e.g. alginate, chitosan, cellulose derivatives; Cyclodextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5169Proteins, e.g. albumin, gelatin

Definitions

  • the present invention is related to core-shel l photo-chemo nanomedicine intended for combinatorial delivery of a photosensitizer drug and chemo drug against disease like cancer. More particularly, the present invention provides a method for synthesizing stable, well dispersed, unagglomerated core-shell nanoparticles of total size ⁇ l 000nm, loaded with a photosensitizer drug and chemo-drug at least one each in the core and shell. More specifically, the invention is related to i) polymer-core/protein-shell and ii) polymer-core/polymer-shell nanoparticles, each core or shell are loaded with at least one photosensitizer drug or chemo-drug, separately. These nanomedicines are used for the treatment of cancer type diseases using combinatorial photodynamic therapy (PDT) and chemotherapy.
  • PDT combinatorial photodynamic therapy
  • Cancer one of world's deadliest diseases and leading cause of death, is operated by several altered molecular pathways. Regardless of the improvements in diagnostic tools and sophisticated treatment approaches, it's highly heterogeneous genotypes and patient to patient variations make the cure very difficult.
  • Conventional therapies such as chemotherapy and radiation therapy mainly focus on killing cancer cells by causing DNA damage by direct ionization, intercalation or reactive oxygen stress.
  • New molecular targeted therapies cause inhibition of aberrant protein kinase or enzymatic activ ity that supports cancer survival.
  • none of these methods could provide significant curative effect for cancer.
  • cancer is operated by multiple molecular mechanisms involving complex protein kinase networks, DNA replication, mutations, drug resistance, and existence of cancer stem cells having privileged advantages of enhanced ROS scavenging capacity and preferentially activated DNA repair mechanism.
  • Photodynamic therapy is a promising method of treating cancer type disease using photosensitive chemicals that release reactive oxygen species upon excitation with light, in the presence of oxygen.
  • This highly reactive ROS species kil l malignant cel ls by apoptosis and/or necrosis along with shut down of tumour microvasculature and stimulation of the host immune system.
  • the neoplastic conditions mainly treated by PDT inc ludes inoperable esophageal tumors, head and neck cancers, micro-invasive endo-bronchial non-smal l cell lung carcinoma, and cancers in, bile duct, gall bladder, gullet, lung, mouth, skin etc.
  • PDT is also being investigated in preclinical and clinical studies for breast, prostate and ovarian cancers.
  • ROS being is the main mediator of PDT assisted cancer cell death, its inhibition by cancer stem cells due to its enhanced ROS scavenging ability and DNA repair capacity pose a major challenge for PDT to cure cancer completely. Abrogation of such drug-resistant mechanisms could have major therapeutic implications in PDT.
  • AM RITA VISWAVIDYAPEETHAM UNIVERSITY represented by its Director, Amrita Centre for Nanosciences, Dr. Shantikumar Nair
  • One of the possible methods to enhance the therapeutic efficacy of PDT is to inhibit the molecular mechanism responsible for ROS resistance or DNA repair mechanisms of cancer cells using other chemical drugs such as small molecule inhibitors that can down regulate the respective intracellular pathways.
  • small molecule inhibitors that can down regulate the respective intracellular pathways.
  • simultaneous use of small molecule drugs that inhibits the ROS scavenging activity in cancer along with photosensitizes can improve the effectiveness of PDT.
  • cancer cells in advanced stages of the disease frequently exhibit multiple genetic alterations leading to ineffectiveness of conventional single agent chemotherapy.
  • Development of drug resistance is another major obstacle to the success of anticancer chemotherapy.
  • Tumor cells utilize multiple mechanisms to reduce the accumulation of the anticancer agents at its intracellular site of action or develop pint mutations in the drug binding domain of the kinase.
  • Nanoparticle mediated drug delivery improves the conventional cancer therapy by aiding the delivering of appropriate drug combinations in optimum therapeutic dosages on the tumour site and also by minimising the risk systemic toxicities. They also aids targeted drug delivery by conjugating drug loaded nanoparticles with tumour specific bio-markers like antibodies, peptide and other ligands.
  • AMRITA VISWAVIDYAPEETHAM UNIVERSITY represented by its Director, Amrita Centre for Nanosciences, Dr. Shantikumar Nair
  • Simple nanoparticle aided drug del ivery systems possess architectural limitations to carry multiple therapeutic agents in optimum concentrations and also to give the desired drug release.
  • the core and shell can be made up of d ifferent materials such as polymers or proteins according to the nature of the drug, its sequence of del ivery and the release kinetics needed.
  • a conventional cytotoxic stress inducing agent like PS or DNA damaging agents or other ROS agents can be used along with its drug resistance inhibitors like DNA damage inhibitors (PARP inhibitor, Chk 1 & Chk2 inhibitors and like), ROS scavenging inhibitors, cytoskeletal disturbing agents (for example dasatinib) etc.
  • PARP inhibitor DNA damage inhibitor
  • Chk 1 & Chk2 inhibitors and like ROS scavenging inhibitors
  • cytoskeletal disturbing agents for example dasatinib
  • Biodegradable or biocompatible polymeric and protein nanoparticle were reported to be used for delivering single or multiple therapeutics inc luding chemo-drugs, pro-drugs, contrast agents for treating and diagnosing various diseases and disorders; especially cancer and related manifestations. Majority of such nanoparticle mediated delivery of therapeutics also possess provision for attaching a specific ligand to aid the targeted drug delivery.
  • chemotherapeutic or imaging agent is described in patents US 006/165440, US 200801 81 852, US 20090226393. US 20 1 1 0020457. US 2007/0009441. US 2008/0253969, etc.
  • Polymeric micelle having a structure of core and shell were reported to be used for drug delivery applications, were disclosed in patents EP 0552802, US 6080396, US 54495 13 etc.
  • US 8021652 disclose the preparation and use of biodegradable branched polylactide derivatives forming polymeric micelles for delivery of poorly water soluble drugs.
  • US patents 201 1 / 100271 72 and 201 l / 10229556 discloses use of lipid coated polymeric particles for drugs ' & radiopharmaceutical agents and adjuvant molecules respectively.
  • Amphiphil ic block co polymer and polymeric composition comprising the same for drug delivery applications are disc losed in US 73 1 1901 & US 2008/01 52616.
  • the preparation of nanoparticles with micellar structure comprising an amphiphilic block copolymer with a hydrophobic agent encapsulated within the micellar structure, and a functionalized corona (US patent no: 2007/0253899), drug-loaded micelle comprising a triblock copolymer, wherein said micelle has a drug-loaded inner core, a crosslinked outer core, and a hydrophilic shell, wherein the multiblock copolymer (US patent no: 201 0/0159020), drug-loaded poly(alkyl-cyanoacrylate) nanoparticles (U S patent no: 2008/01 82776) etc are reported.
  • PCL polycapro lactone
  • PEG Polyethylene glycol
  • the drug is loaded in the polymer core made of cellulose or collagen or lactose or alginate by milling process.
  • the outer polymer coat is not carrying any therapeutic or imaging agents.
  • Polymer dendrimers having a core- shell nature were disclosed for their use in delivering multiple therapeutic agents, photosensitizers, contrast agents or biologically active agents (DNA, RNA etc.,) in U S 2005/028 1 777 & US 2009/0012033
  • nanoparticle having a core-shell nature or structure like multi-layered dendrimers, polymeric micelles and polymer or lipid coated nanoparticles were disclosed in the above mentioned prior arts, none of these arts have complied with a definitive structure of a nano-construct comprising of a distinct polymeric core and a distinct polymer /protein shell loaded with a photosensitizer and chemo-drug in combination to aid a combinatorial treatment using PDT and chemotherapy togother. Contrary to the earlier inventions, we have synthesized a polymer-core and polymer/protein shell nanomedicine, each having a cross linked/solid structure with a distinct interphase.
  • polymeric nanopartices were used in few of the prior arts, polymer-protein core shell systems are not reported. Furthermore, in our method, these protein molecules are cross- linked for better encapsulation of drug molecule.
  • the cross-linking methods are selected in such a way that the chemical stability of drug payloads remains unchanged. Accordingly, there exist no prior art on the preparation of polvmer-core/protein shell or polymer-core/polymer-shell nanomedicine encapsulating photosensitizer and cheino drugs separately or com bined in core- shell nanoparticle aiding the seq uential or simultaneous delivery of these drugs at the site of; action, in a targeted manner.
  • a multifunctional polymer/protein and polymer/polymer core-shell photo-chemo nanomedicine that can provide simultaneous or sequential delivery of two different types of therapeutic agents such as a photosensitizer drug in combination with an anti-neoplastic agent to cancer and related diseases in a targeted fashion.
  • the nanomedicine is based on a polymeric nano-core and a shell made up of either another polymer or protein.
  • the core and the shell will be loaded with drugs in the following possible formats: a) core with a photosensitizer and shell with anti-neoplastic chemodrugs, b) core with antineoplastic chemodrugs and shell with a photosensitizer.
  • AMR1TA V1SWAVIDYAPEETHAM UNIVERSITY represented by its Director, Amrita Centre for Nanosciences, Dr. Shantikumar Nair
  • the core and shel l are made up of bio-compatible po lymers or prote ins suited for the dug loading and preferred controlled release. This unique nanomed ic ine is intended for the combinatorial cancer treatment using photodynamic therapy and chemotherapy.
  • nanoparticle refers to primary inventive nanoparticles formed by protein or polymer, measuring size about 1 - 1000 nm, preferably 1 - l OOnm, most preferably around 1 -50 nm in size showing " multifunctional ' property of delivering mu ltiple therapeutic agents such as photosensitizer, chemo-drugs, small molecule inhibitors etc ., in different combinations of at least one photosensitizer and one or more chemo-drugs together.
  • core-shell nanoparticle refers to a heterogeneous nanoconstruct formed with a central nanoparticle core and an outer nano-shell and both have a distinct interphase.
  • therapeutics refers to photosensitizers, chemo-drugs, small molecule inhibitors, pro-drugs, etc that have a therapeutic effect against a d iseases, espec ially cancer and related clinical manifestationss.
  • targeting ligand refers to biomo lecu les that can spec ifical ly identify and target another molecule like an antigen or receptor on the surface of cell-membrane of diseased cells such as that of cancer / tumor.
  • Targeting ligand include antibodies, peptides, aptamers, vitamins like folic acid, sugar molecules like mannose ligands, carbohydrates etc.
  • nanomedicine refers to nanoparticles loaded with therapeutics.
  • photo-chemo nanomedicine refers to a nanoparticle loaded with at least one photosensitizer and chemo-drug in a single core-shell nanomedicine system.
  • FIGURE CAPTIONS
  • Figure 1 Schematic showing preparation of polymer-protein core-shell nanomedic ine construct made of PLGA-mTHPC core & albumin-dasatinib shell
  • Figure 2 A) Atomic force microscopic image and B) DLS analysis of PLGA-mTHPC nanocore , showing particle size of ⁇ 80nm.
  • Figure 3 FT1R spectrum of PLGA-mTHPC core, albumin-dasatinib shell and the complete nanomedicine construct.
  • Figure 4 Figure A represents the comparison of florescence property of bare mTHPC and nano-core encapsulated mTHPC. B) is the singlet oxygen generation by the nano-core by SOSGR assay .
  • AMRITA VISWAVIDYAPEETHA UNIVERSITY represented by its Director, Amrita Centre for Nanosciences, Dr. Shantikumar Nair Figure 5 : Cellular uptake stud ies of nanomedicine construct by U 87MG human glioma cells.
  • Figure 6 In vitro cell scratch assay for migration inhibition analysis.
  • A) Microscopic images of in vitro scratch assay showing effective inhibition of cell migration on the nanomedicine treated cells.
  • B) and C) are the quantitative representation of migration inhibition in terms of no of cell migrated and percentage scratch healed respectively.
  • D is western blot analysis showing p-Src down regulation by the nano-shell encapsulated dasatinib.
  • Figure 7 Confocal microscopic images showing actin cytoskeleton disruption in the core-shell nanomedicine treated glioma cells
  • Figure 8 Effect of nanomedicine treatment on the morphology and attachment ability of U87MG cells.
  • Figure A is SEM images showing significant reduction in philopodial extensions in the treated cells in comparison with normal cells.
  • B is confocal imaging of actin (red) and vincuiin (green) showing reduction in focal adhesion point formation by nanomed icine treatment.
  • Figure 10 Analysis of cytotoxicity by the nanomedicine construct.
  • Graph A showing absence of dark toxicity of nanomedicine.
  • Figure B show ing l ight induced cytotoxicity by the nanomed icine construct and nano-core.
  • Figure C is early stage apoptosis induced by nanomedicine mediated combination therapy.
  • the main feature of the inventive nanomedicine is the core-shell structure where the core is formed by a biocompatible and biodegradable polymer loaded with either a photosensitizer drug or a chemo-drug and the shell formed over the core, either by another polymer or a protein and loaded with either a photosensitizer or a chemo-drug.
  • the' core-shell structure will always have a photosensitizer and chemo-drugs in combination, for example, in case of a metastatic cancer, aberrant kinase associated with migration of cancer cells can be inhibited by a chemo-drug loaded in the shell and photosenzitizer drug in the core.
  • This facilitate application of PDT assisted ROS stress after curtailing the migratory capacity of metastaic cells.
  • An advantage of this method is that, molecular pathways responsible for migration and metastasis can be inhibited by relatively less concentration of small-molecules and complete cell death can be achieved by PDT.
  • enhanced PDT mediated cytotoxicity can be achieved by delivering an antioxidant inhibitor molecules along with photo-sensitizer using a core-shel l nanomedicine. It is well known that radiation resistant cancer cells have evolved various cellular anti-oxidant pathways to counteract radiation/PDT or chemotherapy mediated ROS stress. In add ition, a small populations of cancer stem cells, are the main causes of cancer relapse and drug resistance as they exhibit elevated activity of such antioxidant pathways and over expression of ROS scavengers.
  • a combinatorial cancer treatment using a photo-drug along with antioxidant inhibitors like diethyl-dithiocarbamate, methoxyestradiol, 1-buthionine sulfoximine, 3-amino- l ,2,4-triazole, etc may potentiate the PDT mediated cytotoxicity.
  • another radiation or chemo resistance mechan ism of cancer or cancer stem cells are preferential activation of DNA repair mechanism.
  • a small molecule inhibitor of enzymes responsible for activated DNA repair damage can be loaded on to the shell of the core-shell nanomed ic ine together with a photorned icine at the core.
  • the DNA damage caused by ROS due to PDT cannot be effectively repaired by the cancer cel ls, th us ac hieving better toxic ity effects even inn radiation resistance or cancer stem cells.
  • Smal l mo lec u le inh i bi tors for suc DNA damage repair molecu les include PARP inhibitors. Clik inhibitors, etc .
  • the polymer for making nano-core and nano-shell is selected from the group of biodegradable polymers such as, but not limited to poly glycolic acid (PGA), poly(lactic-co-glycolic acid) (PLGA), glycolide/trimethylene carbonate copolymers (PGA/TMC); poly-lactides (PLA), poly-L-lactide (PLLA), Poly-DL-Iactide (PDLLA), L- lactide/DL-lactide copolymers; lactide/tetramethyl-glycol ide copolymers, poly-caprolactone (PCL), poly-valerolacton(PVL).
  • biodegradable polymers such as, but not limited to poly glycolic acid (PGA), poly(lactic-co-glycolic acid) (PLGA), glycolide/trimethylene carbonate copolymers (PGA/TMC); poly-lactides (PLA), poly-L-lactide (PLLA), Poly-DL-Iactide (PDLLA), L- lac
  • PHB poly-hydroxy butyrate
  • PVA poly vinyl alcohol
  • PVP poly-hydroxy valerate
  • PVP polyvinylpyrrolidone
  • PEI Polyethyleneimine
  • lactide/trimethylene carbonate copolymers chitosan, carboxymethyl chitosan, chitin, pollulan, etc., or blends thereof.
  • the protein used for nano-shell is chosen from the group of protein such as human serum albumin, or protamine, transferrin, lactoferrin, fibrinogen, gelatin, mucin, soy protein, apoferritin, ferritin, lectin, lactoferrin, gluten, whey protein, prolamines such as gliadin, hordein, secalin, zein, avenin, or combinations thereof.
  • protein such as human serum albumin, or protamine, transferrin, lactoferrin, fibrinogen, gelatin, mucin, soy protein, apoferritin, ferritin, lectin, lactoferrin, gluten, whey protein, prolamines such as gliadin, hordein, secalin, zein, avenin, or combinations thereof.
  • the photozensitizer loaded on either nanomediciene core or shell is chosen from, and not limited by chlorine e6 (Ce6), meso-tetra(3- hydroxyphenyI)chlorin (m-THPC), methylene blue, benzoporphyrin derivative monoacid ring A ' (BPD or verteporfin), photofrin, Rose bengal, metal phthalocyanine, hypericin, toluidine blue O, pyropheophorbide-a hexyl ether (HPPH), Indium pyropheophorbide, padoporfin, padeliporfin, and combinations thereof.
  • chlorine e6 Ce6
  • m-THPC meso-tetra(3- hydroxyphenyI)chlorin
  • m-THPC meso-tetra(3- hydroxyphenyI)chlorin
  • m-THPC meso-tetra(3- hydroxyphenyI)chlorin
  • the protein shell is formed by
  • r f j n g to the schematic given in Fig. 1 , for the preparation of polymer-core/protein-shell j T
  • a VISWAVIDYAPEETHAM UNIVERSITY represented by its Direc tor, A in ri til Centre for s C j el ices, Dr. Shantikumar Nair
  • the precipitated particles are purified by washing and dried by lyophilization.
  • a suitable protein for example albumin
  • the desired drug-2 for example a chemodrug, Dasatinib
  • the lyophilized polymer core nanoparticles are suspended in the protein-drug-2 mixture and the method of coaservation is applied to form the shell.
  • the core-shell nanoparticle is bio-conjugated with specific ligands for example folic acid, aid targeted delivery.
  • the nanomeidicine core showed an average size of ⁇ 80 nm and an overall size of - 120 nm as shown in the figure 2.
  • Figure 2 A and 2B denotes the AFM image and DLS measurement of ⁇ 80nm sized PLGA-mTHPC core, where 2C and 2D denotes the SEM image and DLS measurements of ⁇ 120nm sized nanomedicne construct.
  • the contrast difference between the nano core and shell clearly visualized in the TEM image (Fig 2C inset) establishes our claim of core-shell nanomedicine.
  • the nanomedicine construct showed an overall size of ⁇ 120 nm as depicted in figure.
  • the nanomedicne core, shell and the entire nanomedicine construct shows distinct FTIR pattern as depicted in figure 3, shows the successful incorporation of the specific therapeutics in the nanomedicine construct.
  • the IR peaks at 3430cm- land 1647cm- l for PLGA-mTHPC possibly represent hydroxy! groups and primary/secondary amine groups of mTHPC respectively. Presence of aromatic amines in the porphyrin structure of mTHPC was evident from IR peak at 1288cm-l . This peak may also represent the presence of carboxylic and ester linkages in the PLGA matrix. Also, the presence of ketone groups in the PLGA matrix was confirmed by IR peak at 1 121 cm-l .
  • mTHPC restores its native intact structural chemistry even after encapsulation within PLGA, despite electrospray invokes high voltage to produce PLGA-mTHPC nanoparticles.
  • IR peaks of core-shell nanomedicine showed additive IR peaks of PLGA-mTHPC and BSA-dasatinib.
  • the nanomedicne core show enhanced flurescence when compared with the same concentrations of photosensitizer at ⁇ dress ⁇ 652nm in aqueous medium.
  • the figure 4A shows the 38 fold increase in florescence intensity by the PLGA-mTHPC nano-core in comparison with same concentrations of bare photosensitizer.
  • Figure 4B depicts the singlet oxygen generation from the nano-core under laser irradiation measured by SOSGR assay , providing proof of its capability to induce enormous ROS stress to the diseased cells.
  • PLGA-mTHPC released singlet oxygen at least 25 times greater than free mTHPC of equimolar concentration in aqueous medium.
  • a steady state increase in the release of singlet oxygen from PLGA-mTHPC for 50 minutes was found when singlet oxygen is detected at emission ⁇ dress ⁇ 525nm during light irradiation.
  • the PLGA-mTl lPC/albumin-Dasatinib nanomedicine shows a time and concentration dependent cell uptake pattern.
  • Fig. 5A refers to the uptake studies of the nanomedicine by FACS analyzing, showing an effective particle uptake by U87MG glioma cells after 6hr incubation.
  • Refrering to fig. 5B depicts a quantitative data showing time and concentration dependent pattern of nanomedicine uptake as a measure of mTHPC florescence.
  • the mTHPC florescence from the nanomedicine treated cells visualized under florescent microscope, depicted iii figure 5C.
  • Fig 6A depicts the microscopic images of effective inhibition of cell migration.
  • the nanomedicine treated cel l showed effective inhibition in migration even after 1 8 hrs. while the untreated cells migrated and healed the scratch area.
  • the quantitative measure of the same is depicted in figure 6B and 6C by means of total no. of cells migrated and the percentage scratch healing respectively.
  • the p-Src level down regulation by nanomedicine-dasatinib, that intern inhibit cell migration is proved by western blot analysis depicted in fig.
  • FIG. 6D The nanomedicine treatment induced changes in cytoskeleton arrangement investigated is depicted in figure 7.
  • the actin staining-confocal images of U87MG cell showed loss of proper cytoskeleton in the drug treated group. Loss of philopodial extension (fig 8A) and focal adhesion points (8B) were also seen in the nanomedicine treated cells.
  • the nanomedicine induced intracellular ROS was visualized by DCFH staining and live cell D1C imaging.
  • the nanomedicine treated cells showed enhanced production of intracellular ROS compared to bare photosensitizer and showed morphological changes associated with apoptosis (fig. 8).
  • the photo-chemo nanomedicine treated ceils showed enhanced cytotoxicity compared to cells treated w ith nanoparticles having photodrug alone (fig 9).
  • the dasatinib in the nano shell enhanced the PDT mediated cytotoxicity.
  • the confocal images of the cells, one hour past the light irradiation shows the early stage apoptosis; Annexin V (F1TC conjugated) bound to the externalized phosphatidyl serine as a result of apoptosis.
  • the design of the nanomedicine is in such a way to simultaneously carry two different drugs and deliver it specifically to the tumor cells in desired fashion.
  • the targeting is achieved by a specific biomarker ligand conjugated to the nanomedicine construct.
  • the core and shell are made by different polymers or a protein and polymer aiding the optimum release kinetics.
  • Example I Polymer core /protein shell photo-chemo-nanomedicine.
  • PLGA Poly(lactic- co-glycolic ac id (50: 50)] nanocore and small molecule inhibitor dasatinib entrapped protein shell.
  • PLGA solution containing ! mM mTHPC was prepared by dissolv ing it in 1 .5 wt % PLGA solution in acetone and was stirred for 1 hr in dark prior to electrospray.
  • This solution was then electrosptrayed at a rate of 1.5 ml/hr to a grounded glass beaker containing de-ionized water premixed with 0.001 v/v Tweeen 20® under constant stirring (550 - 600 rpm). The tip to target distance was maintained as 7.5 throughout the experiment.
  • the electrospray was carried out under ambient temperature, pressure and 55 ⁇ 5% humidity, by applying a potential of 1 .4kV/cm using a high voltage supply.
  • AM RITA VISWAVIDYAPEETHAM UNIVERSITY represented by its Director, Amrita Centre for Nanosciences, Dr. Shantikumar Nair
  • the particles were acquired by lyophilizing the electrosprayed solution.
  • the A lbumin-dasatinib solution was prepared by mixing fresh dasatinib-DMSO stock in 5- 10 wt % BSA solution in de- ionized water to get a final dasatinib concentration of 1 0 ⁇ ⁇ .
  • Lyophilized PLGA-mTHPC particles were mixed with the BSA-dasatinib solution and stirred for 2 hours prior to co-acrervation.
  • BSA- dasatinib nanoshell was prepared over PLGA-mTHPC nanoparticles by ethanol co-acervation, in that 1 :2 volume of absolute ethanol was added drop wise ( ⁇ ⁇ /min) under stirring (800- 1 000 rpm) till the solution became turbid, to aid optimum albumin-dasatinib co-acervate coating over the PLGA- mTHPC NPs.
  • the system is then added with 35 ⁇ ⁇ of 4mg/ml l -Ethy l-3-[3-dimethylaminopropyl] carbodiimide hydrochloride (EDC) and stirred for 2 hours to link and harden the coacervates formed.
  • EDC dimethylaminopropyl
  • Example 2 Polymer core /polymer shell photo-chemo-nanomedicine
  • This solution was then electrosptrayed at a rate of 1 ml/hr to a grounded glass beaker kept at a distance of 7cm that containing de-ionized water with 0.00 1 v/v Tweeen 20® as surfactant and was maintained in constant stirring (600 rpm).
  • the electrospray was carried out under ambient temperature, pressure and 60 ⁇ 5% humidity, by apply ing a potent ial of 1 .3 kV/cm, using a high voltage supply.
  • the partic les were acquired by lyophilizing the electrosprayed solution. T he lyophilized particles are then mixed with 0.5% chitosan solution containing 5 mM sorafenib.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Physics & Mathematics (AREA)
  • Nanotechnology (AREA)
  • Optics & Photonics (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

The present invention is related to a core-shell photo-chemo nanomedicine aiding sequential or simultaneous delivery of a photosensitizer drug and at least one chemotherapeutic drug for combinatorial photodynamic therapy and chemotherapy of cancer. The nanomedicine core is made up of a biodegradable or biocompatible polymer and the shell is either by polymer or a protein aiding the following two configurations; (i) polymer core-polymer shell and (ii) polymer core-protein shell. The core-shell nanomedicine is made in such a way that, while the core is loaded with photosensitizer, the shell will be loaded with a chemo-drug or vice-versa. Along with improving drug solubility, stability, bioavailability and therapeutic efficiency, the core-shell nanomedicine platform aids sequential or simultaneous delivery of photosensitizer and chemo drugs to cancer type diseases and thereby facilitates the application of photodynamic reactive oxygen stress together with or followed by chemotherapy. In addition, this core-shell nanomedicine can be targeted specifically to the disease site by conjugating with targeting molecules such as antibodies, peptides, small molecules, vitamins, proteins, etc. This core-shell photo-chemo nanomedicine system is capable of excreting photodynamic reactive oxygen stress together with or followed by chemotherapy.

Description

FIELD OF INVENTION
The present invention is related to core-shel l photo-chemo nanomedicine intended for combinatorial delivery of a photosensitizer drug and chemo drug against disease like cancer. More particularly, the present invention provides a method for synthesizing stable, well dispersed, unagglomerated core-shell nanoparticles of total size < l 000nm, loaded with a photosensitizer drug and chemo-drug at least one each in the core and shell. More specifically, the invention is related to i) polymer-core/protein-shell and ii) polymer-core/polymer-shell nanoparticles, each core or shell are loaded with at least one photosensitizer drug or chemo-drug, separately. These nanomedicines are used for the treatment of cancer type diseases using combinatorial photodynamic therapy (PDT) and chemotherapy.
BACKGROUND
Cancer, one of world's deadliest diseases and leading cause of death, is operated by several altered molecular pathways. Regardless of the improvements in diagnostic tools and sophisticated treatment approaches, it's highly heterogeneous genotypes and patient to patient variations make the cure very difficult. Conventional therapies such as chemotherapy and radiation therapy mainly focus on killing cancer cells by causing DNA damage by direct ionization, intercalation or reactive oxygen stress. New molecular targeted therapies cause inhibition of aberrant protein kinase or enzymatic activ ity that supports cancer survival. However, none of these methods could provide significant curative effect for cancer. One of the main reasons is that cancer is operated by multiple molecular mechanisms involving complex protein kinase networks, DNA replication, mutations, drug resistance, and existence of cancer stem cells having privileged advantages of enhanced ROS scavenging capacity and preferentially activated DNA repair mechanism.
Photodynamic therapy (PDT) is a promising method of treating cancer type disease using photosensitive chemicals that release reactive oxygen species upon excitation with light, in the presence of oxygen. This highly reactive ROS species kil l malignant cel ls by apoptosis and/or necrosis along with shut down of tumour microvasculature and stimulation of the host immune system. The neoplastic conditions mainly treated by PDT inc ludes inoperable esophageal tumors, head and neck cancers, micro-invasive endo-bronchial non-smal l cell lung carcinoma, and cancers in, bile duct, gall bladder, gullet, lung, mouth, skin etc. PDT is also being investigated in preclinical and clinical studies for breast, prostate and ovarian cancers.
Even though PDT possess high potential for cancer treatment, the effectiveness of this therapy is limited by the problems associated with non-specific light toxicity, delivery of PS drugs, and the ability of cancer stem cells to survive PDT by scavenging ROS very effectively. ROS, being is the main mediator of PDT assisted cancer cell death, its inhibition by cancer stem cells due to its enhanced ROS scavenging ability and DNA repair capacity pose a major challenge for PDT to cure cancer completely. Abrogation of such drug-resistant mechanisms could have major therapeutic implications in PDT.
AM RITA VISWAVIDYAPEETHAM UNIVERSITY represented by its Director, Amrita Centre for Nanosciences, Dr. Shantikumar Nair One of the possible methods to enhance the therapeutic efficacy of PDT is to inhibit the molecular mechanism responsible for ROS resistance or DNA repair mechanisms of cancer cells using other chemical drugs such as small molecule inhibitors that can down regulate the respective intracellular pathways. For example, simultaneous use of small molecule drugs that inhibits the ROS scavenging activity in cancer along with photosensitizes can improve the effectiveness of PDT. Alternatively, inhibition of DNA repair process using small molecule drugs followed by application of ROS stress by PDT, or inhibition of metastasis / migratory capacity of cancer followed by application of ROS stress by PD, etc, are promising combinatorial approach to achieve better curative effects.
In addition, studies have reported the important role of cancer micro environment in attaining drug resistant and uncontrolled proliferation of cancer. It is understood that destroying cancer micro environment is critical in achieving better therapeutic effects. One possibility is to use molecules that disrupt the cytoskeletal arrangement and cancer cell-niche interactions by inhibiting the integrin binding capac ity, in combination with application of ROS stress by PDT may yield better curative effects. Conceivably, combinations of agents with complementary mechanisms of act ion could be more effective in dealing with cancer than single agents or single therapy.
In another aspect, cancer cells in advanced stages of the disease frequently exhibit multiple genetic alterations leading to ineffectiveness of conventional single agent chemotherapy. Development of drug resistance is another major obstacle to the success of anticancer chemotherapy. Tumor cells utilize multiple mechanisms to reduce the accumulation of the anticancer agents at its intracellular site of action or develop pint mutations in the drug binding domain of the kinase. In order to overcome these challenges, it is proposed to apply multiple stress mechanisms against cancer simultaneously. For example, application of DNA intercalating agents together with ROS stress by photodynamic therapy, or silencing of oncogene using small interfering RNA together with application of PDT , etc are promising options.
However, all the above methods demands simultaneous delivery of chemical drugs together with photosensitive drugs to cancer. The success of such combinatorial approaches lies heavily of our ability to deliver appropriate drug combinations at appropriate concentrations, specifically at the tumour sites. Overcoming the challenges faced by such a strategy includes attaining optimum drug concentrations of drugs at the tumour sites in appropriate sequence with least systemic toxicities.
The emerging field of nano-drug delivery offer great promise for targeted delivery of drugs to tumor type diseases. Nanoparticle mediated drug delivery improves the conventional cancer therapy by aiding the delivering of appropriate drug combinations in optimum therapeutic dosages on the tumour site and also by minimising the risk systemic toxicities. They also aids targeted drug delivery by conjugating drug loaded nanoparticles with tumour specific bio-markers like antibodies, peptide and other ligands.
AMRITA VISWAVIDYAPEETHAM UNIVERSITY represented by its Director, Amrita Centre for Nanosciences, Dr. Shantikumar Nair Although there are number of nanoparticle based drug del ivery systems reported so far, most of them deal with single agent delivery. Simple nanoparticle aided drug del ivery systems possess architectural limitations to carry multiple therapeutic agents in optimum concentrations and also to give the desired drug release. There are also risks regarding possible cross-talk between the drugs and photo-degradation of chemodrugs by the photosensizers. So a heterogeneous drug del ivery system like core-shell nanoparticles were two drug molecules can be incorporated in each layer without cross-interactions are needed. The core and shell can be made up of d ifferent materials such as polymers or proteins according to the nature of the drug, its sequence of del ivery and the release kinetics needed. In such a platform a conventional cytotoxic stress inducing agent like PS or DNA damaging agents or other ROS agents can be used along with its drug resistance inhibitors like DNA damage inhibitors (PARP inhibitor, Chk 1 & Chk2 inhibitors and like), ROS scavenging inhibitors, cytoskeletal disturbing agents (for example dasatinib) etc.
PRIOR ART
Biodegradable or biocompatible polymeric and protein nanoparticle were reported to be used for delivering single or multiple therapeutics inc luding chemo-drugs, pro-drugs, contrast agents for treating and diagnosing various diseases and disorders; especially cancer and related manifestations. Majority of such nanoparticle mediated delivery of therapeutics also possess provision for attaching a specific ligand to aid the targeted drug delivery. In prior art use of nanoparticles with and without a targeting ligand for the delivery of chemotherapeutic or imaging agent is described in patents US 006/165440, US 200801 81 852, US 20090226393. US 20 1 1 0020457. US 2007/0009441. US 2008/0253969, etc. Use of polymeric nanoparticles for photosensitizer delivery is de liberated in patents U S 201 1 /1 0238001 , U S 201 1 /0022 129 etc. U S 20 1 1 0165258 disclosed the use of polymer aggregates having both hydrophtlic and hydrophobic segments for the delivery of drugs and diagnostic agents. Simultaneous loading of pro-drug along with its activating enzyme in nanoparticles for two step targeted tumour therapy has disclosed in U S patent 201 1 /1 021 7363.
Polymeric micelle having a structure of core and shell were reported to be used for drug delivery applications, were disclosed in patents EP 0552802, US 6080396, US 54495 13 etc. US 201 1 10091 534, US 2010/01 59019, US 7638558 disclosed multi-block polymeric micelles for drug delivery applications. US 8021652 disclose the preparation and use of biodegradable branched polylactide derivatives forming polymeric micelles for delivery of poorly water soluble drugs. US patents 201 1 / 100271 72 and 201 l / 10229556discloses use of lipid coated polymeric particles for drugs ' & radiopharmaceutical agents and adjuvant molecules respectively. Amphiphil ic block co polymer and polymeric composition comprising the same for drug delivery applications are disc losed in US 73 1 1901 & US 2008/01 52616. In the prior art, the preparation of nanoparticles with micellar structure comprising an amphiphilic block copolymer with a hydrophobic agent encapsulated within the micellar structure, and a functionalized corona (US patent no: 2007/0253899), drug-loaded micelle comprising a triblock copolymer, wherein said micelle has a drug-loaded inner core, a crosslinked outer core, and a hydrophilic shell, wherein the multiblock copolymer (US patent no: 201 0/0159020), drug-loaded poly(alkyl-cyanoacrylate) nanoparticles (U S patent no: 2008/01 82776) etc are reported.
AM RITA VISWAVIDYAPEETHAM UNIVERSITY represented by its Director, Ani rita Centre for Nanoseiences, Dr. Shantikumar Nair Conversely patent US 2009/0214633 discloses the use of particles prepared by nano- encapsulation having lipid core and polymeric shell for protein drug delivery applications. US 2008/0248126 discloses use of multi-layered polymeric or oligomeric nanoparticles prepared by ring opening polymerisation for drug delivery applications, in that the drug is either loaded in the core or in shell. In a similar art disclosed in US patent 2010/0203 149, nanoparticle having drug loaded polymer core surrounded by water soluble polymeric chains for its potential use in cytoplasmic drug delivery to cancer cells.
Use of polycapro lactone (PCL) or Polyethylene glycol (PEG) coated polymeric nanoparticles for anticancer drug delivery was disclosed in US patent 201 1 /0052709. In this, the drug is loaded in the polymer core made of cellulose or collagen or lactose or alginate by milling process. The outer polymer coat is not carrying any therapeutic or imaging agents. Polymer dendrimers having a core- shell nature were disclosed for their use in delivering multiple therapeutic agents, photosensitizers, contrast agents or biologically active agents (DNA, RNA etc.,) in U S 2005/028 1 777 & US 2009/0012033
Even though nanoparticle having a core-shell nature or structure like multi-layered dendrimers, polymeric micelles and polymer or lipid coated nanoparticles were disclosed in the above mentioned prior arts, none of these arts have complied with a definitive structure of a nano-construct comprising of a distinct polymeric core and a distinct polymer /protein shell loaded with a photosensitizer and chemo-drug in combination to aid a combinatorial treatment using PDT and chemotherapy togother. Contrary to the earlier inventions, we have synthesized a polymer-core and polymer/protein shell nanomedicine, each having a cross linked/solid structure with a distinct interphase.
Even though the polymeric nanopartices were used in few of the prior arts, polymer-protein core shell systems are not reported. Furthermore, in our method, these protein molecules are cross- linked for better encapsulation of drug molecule. The cross-linking methods are selected in such a way that the chemical stability of drug payloads remains unchanged. Accordingly, there exist no prior art on the preparation of polvmer-core/protein shell or polymer-core/polymer-shell nanomedicine encapsulating photosensitizer and cheino drugs separately or com bined in core- shell nanoparticle aiding the seq uential or simultaneous delivery of these drugs at the site of; action, in a targeted manner.
SUMMARY OF THE INVENTION
In the present invention, a multifunctional polymer/protein and polymer/polymer core-shell photo-chemo nanomedicine that can provide simultaneous or sequential delivery of two different types of therapeutic agents such as a photosensitizer drug in combination with an anti-neoplastic agent to cancer and related diseases in a targeted fashion. Specifically, the nanomedicine is based on a polymeric nano-core and a shell made up of either another polymer or protein. The core and the shell will be loaded with drugs in the following possible formats: a) core with a photosensitizer and shell with anti-neoplastic chemodrugs, b) core with antineoplastic chemodrugs and shell with a photosensitizer.
AMR1TA V1SWAVIDYAPEETHAM UNIVERSITY represented by its Director, Amrita Centre for Nanosciences, Dr. Shantikumar Nair The core and shel l are made up of bio-compatible po lymers or prote ins suited for the dug loading and preferred controlled release. This unique nanomed ic ine is intended for the combinatorial cancer treatment using photodynamic therapy and chemotherapy.
DEFEN1TIONS
The term "nanoparticle" as used herein refers to primary inventive nanoparticles formed by protein or polymer, measuring size about 1 - 1000 nm, preferably 1 - l OOnm, most preferably around 1 -50 nm in size showing " multifunctional ' property of delivering mu ltiple therapeutic agents such as photosensitizer, chemo-drugs, small molecule inhibitors etc ., in different combinations of at least one photosensitizer and one or more chemo-drugs together.
The term "core-shell nanoparticle" as used herein refers to a heterogeneous nanoconstruct formed with a central nanoparticle core and an outer nano-shell and both have a distinct interphase.
The term "therapeutics" as used herein refers to photosensitizers, chemo-drugs, small molecule inhibitors, pro-drugs, etc that have a therapeutic effect against a d iseases, espec ially cancer and related clinical manifestationss.
The term "targeting ligand" as used herein refers to biomo lecu les that can spec ifical ly identify and target another molecule like an antigen or receptor on the surface of cell-membrane of diseased cells such as that of cancer / tumor. Targeting ligand include antibodies, peptides, aptamers, vitamins like folic acid, sugar molecules like mannose ligands, carbohydrates etc.
The term "nanomedicine' as used herein refers to nanoparticles loaded with therapeutics.
The term "photo-chemo nanomedicine, as used herein refers to a nanoparticle loaded with at least one photosensitizer and chemo-drug in a single core-shell nanomedicine system.
FIGURE CAPTIONS:
Figure 1 : Schematic showing preparation of polymer-protein core-shell nanomedic ine construct made of PLGA-mTHPC core & albumin-dasatinib shell
Figure 2: A) Atomic force microscopic image and B) DLS analysis of PLGA-mTHPC nanocore , showing particle size of ~ 80nm. C) SEM image of PLGA-mTHPC : BSA-dasatinib core-shell nanomedicine, Inset is TEM image of core-shell nanomed ic ine show ing the difference in contrast between the two D) DLS analysis of core-shell nanomed ic ine show ing an overall size of ~ 120nm.
Figure 3 : FT1R spectrum of PLGA-mTHPC core, albumin-dasatinib shell and the complete nanomedicine construct.
Figure 4: Figure A represents the comparison of florescence property of bare mTHPC and nano-core encapsulated mTHPC. B) is the singlet oxygen generation by the nano-core by SOSGR assay .
AMRITA VISWAVIDYAPEETHA UNIVERSITY represented by its Director, Amrita Centre for Nanosciences, Dr. Shantikumar Nair Figure 5 : Cellular uptake stud ies of nanomedicine construct by U 87MG human glioma cells. A) Dot plot showing nanomedicine uptake determined FACS analysis. B) Concentration and time dependent pattern of nanomedicine uptake exhibited by U 87MG cells C) Florescence image showing mTHPC florescence from nano-core inside the cells after 6hr incubation
Figure 6: In vitro cell scratch assay for migration inhibition analysis. A) Microscopic images of in vitro scratch assay showing effective inhibition of cell migration on the nanomedicine treated cells. B) and C) are the quantitative representation of migration inhibition in terms of no of cell migrated and percentage scratch healed respectively. D is western blot analysis showing p-Src down regulation by the nano-shell encapsulated dasatinib.
Figure 7: Confocal microscopic images showing actin cytoskeleton disruption in the core-shell nanomedicine treated glioma cells
Figure 8 : Effect of nanomedicine treatment on the morphology and attachment ability of U87MG cells. Figure A is SEM images showing significant reduction in philopodial extensions in the treated cells in comparison with normal cells. B is confocal imaging of actin (red) and vincuiin (green) showing reduction in focal adhesion point formation by nanomed icine treatment.
Figure 9: Confocal-DIC imaging of ROS generation; DCFHDA florescence (green) corresponds to extend of ROS generation.
Figure 10: Analysis of cytotoxicity by the nanomedicine construct. Graph A showing absence of dark toxicity of nanomedicine. Figure B show ing l ight induced cytotoxicity by the nanomed icine construct and nano-core. Figure C is early stage apoptosis induced by nanomedicine mediated combination therapy.
DETAILED DESCRIPTIONS OF THE INVENTIONS
The main feature of the inventive nanomedicine is the core-shell structure where the core is formed by a biocompatible and biodegradable polymer loaded with either a photosensitizer drug or a chemo-drug and the shell formed over the core, either by another polymer or a protein and loaded with either a photosensitizer or a chemo-drug.
As the above nanomedicine is intended to combine photodynamic therapy with chemotherapy, the' core-shell structure will always have a photosensitizer and chemo-drugs in combination, for example, in case of a metastatic cancer, aberrant kinase associated with migration of cancer cells can be inhibited by a chemo-drug loaded in the shell and photosenzitizer drug in the core. This facilitate application of PDT assisted ROS stress after curtailing the migratory capacity of metastaic cells. An advantage of this method is that, molecular pathways responsible for migration and metastasis can be inhibited by relatively less concentration of small-molecules and complete cell death can be achieved by PDT.
AM RITA VISWAV1DYAPEETHAM UNIVERSITY represented by its Director, Amrita Centre for Nanoseiences, Dr. Shantikumar Nair In another embodiment, enhanced PDT mediated cytotoxicity can be achieved by delivering an antioxidant inhibitor molecules along with photo-sensitizer using a core-shel l nanomedicine. It is well known that radiation resistant cancer cells have evolved various cellular anti-oxidant pathways to counteract radiation/PDT or chemotherapy mediated ROS stress. In add ition, a small populations of cancer stem cells, are the main causes of cancer relapse and drug resistance as they exhibit elevated activity of such antioxidant pathways and over expression of ROS scavengers. A combinatorial cancer treatment using a photo-drug along with antioxidant inhibitors like diethyl-dithiocarbamate, methoxyestradiol, 1-buthionine sulfoximine, 3-amino- l ,2,4-triazole, etc may potentiate the PDT mediated cytotoxicity.
I n yet another embodiment, another radiation or chemo resistance mechan ism of cancer or cancer stem cells are preferential activation of DNA repair mechanism. In such case, a small molecule inhibitor of enzymes responsible for activated DNA repair damage can be loaded on to the shell of the core-shell nanomed ic ine together with a photorned icine at the core. In such case, the DNA damage caused by ROS due to PDT cannot be effectively repaired by the cancer cel ls, th us ac hieving better toxic ity effects even inn radiation resistance or cancer stem cells. Smal l mo lec u le inh i bi tors for suc DNA damage repair molecu les include PARP inhibitors. Clik inhibitors, etc .
In a preferred embodiment of the said core-shell nanomedicine, the polymer for making nano-core and nano-shell is selected from the group of biodegradable polymers such as, but not limited to poly glycolic acid (PGA), poly(lactic-co-glycolic acid) (PLGA), glycolide/trimethylene carbonate copolymers (PGA/TMC); poly-lactides (PLA), poly-L-lactide (PLLA), Poly-DL-Iactide (PDLLA), L- lactide/DL-lactide copolymers; lactide/tetramethyl-glycol ide copolymers, poly-caprolactone (PCL), poly-valerolacton(PVL). poly-hydroxy butyrate (PHB), poly vinyl alcohol (PVA) poly-hydroxy valerate(PHV), polyvinylpyrrolidone (PVP), Polyethyleneimine (PEI) and lactide/trimethylene carbonate copolymers, chitosan, carboxymethyl chitosan, chitin, pollulan, etc., or blends thereof.
In a preferred embodiment of the said core-shell nanomedicine, the protein used for nano-shell is chosen from the group of protein such as human serum albumin, or protamine, transferrin, lactoferrin, fibrinogen, gelatin, mucin, soy protein, apoferritin, ferritin, lectin, lactoferrin, gluten, whey protein, prolamines such as gliadin, hordein, secalin, zein, avenin, or combinations thereof.
In a preferred embodiment of said core-shell nanomedicine, the photozensitizer loaded on either nanomediciene core or shell is chosen from, and not limited by chlorine e6 (Ce6), meso-tetra(3- hydroxyphenyI)chlorin (m-THPC), methylene blue, benzoporphyrin derivative monoacid ring A ' (BPD or verteporfin), photofrin, Rose bengal, metal phthalocyanine, hypericin, toluidine blue O, pyropheophorbide-a hexyl ether (HPPH), Indium pyropheophorbide, padoporfin, padeliporfin, and combinations thereof.
AM RITA VISWAVIDYAPEETHAM UNIVERSITY represented by its Director, Amrita Centre for Nanosciences, Dr. Shantikumar Nair
Figure imgf000010_0001
preferred embodiment of said core-shell nanomedicine, the protein shell is formed by
Figure imgf000010_0002
rfjng to the schematic given in Fig. 1 , for the preparation of polymer-core/protein-shell
Figure imgf000010_0003
j TA VISWAVIDYAPEETHAM UNIVERSITY represented by its Direc tor, A in ri til Centre for sCjelices, Dr. Shantikumar Nair The precipitated particles are purified by washing and dried by lyophilization. In the next step-2, a suitable protein, for example albumin, is treated with the desired drug-2, for example a chemodrug, Dasatinib, for electrostatic binding. In the third step, the lyophilized polymer core nanoparticles are suspended in the protein-drug-2 mixture and the method of coaservation is applied to form the shell. In the step 4, the core-shell nanoparticle is bio-conjugated with specific ligands for example folic acid, aid targeted delivery.
In relation to the above method of preparing embodiment, the nanomeidicine core showed an average size of ~80 nm and an overall size of - 120 nm as shown in the figure 2. Figure 2 A and 2B denotes the AFM image and DLS measurement of ~80nm sized PLGA-mTHPC core, where 2C and 2D denotes the SEM image and DLS measurements of ~120nm sized nanomedicne construct. The contrast difference between the nano core and shell clearly visualized in the TEM image (Fig 2C inset) establishes our claim of core-shell nanomedicine. The nanomedicine construct showed an overall size of ~ 120 nm as depicted in figure.
In yet another aspect of the above mentioned embodiment, the nanomedicne core, shell and the entire nanomedicine construct shows distinct FTIR pattern as depicted in figure 3, shows the successful incorporation of the specific therapeutics in the nanomedicine construct. The IR peaks at 3430cm- land 1647cm- l for PLGA-mTHPC possibly represent hydroxy! groups and primary/secondary amine groups of mTHPC respectively. Presence of aromatic amines in the porphyrin structure of mTHPC was evident from IR peak at 1288cm-l . This peak may also represent the presence of carboxylic and ester linkages in the PLGA matrix. Also, the presence of ketone groups in the PLGA matrix was confirmed by IR peak at 1 121 cm-l . All these indicated that mTHPC restores its native intact structural chemistry even after encapsulation within PLGA, despite electrospray invokes high voltage to produce PLGA-mTHPC nanoparticles. IR peaks of core-shell nanomedicine showed additive IR peaks of PLGA-mTHPC and BSA-dasatinib.
In yet another aspect of the above embodiment, the nanomedicne core show enhanced flurescence when compared with the same concentrations of photosensitizer at λ„ωχ 652nm in aqueous medium. The figure 4A shows the 38 fold increase in florescence intensity by the PLGA-mTHPC nano-core in comparison with same concentrations of bare photosensitizer. Figure 4B depicts the singlet oxygen generation from the nano-core under laser irradiation measured by SOSGR assay , providing proof of its capability to induce enormous ROS stress to the diseased cells. PLGA-mTHPC released singlet oxygen at least 25 times greater than free mTHPC of equimolar concentration in aqueous medium. A steady state increase in the release of singlet oxygen from PLGA-mTHPC for 50 minutes was found when singlet oxygen is detected at emission λ„ωχ 525nm during light irradiation.
In yet another aspect of the same embodiment, the PLGA-mTl lPC/albumin-Dasatinib nanomedicine shows a time and concentration dependent cell uptake pattern. Fig. 5A refers to the uptake studies of the nanomedicine by FACS analyzing, showing an effective particle uptake by U87MG glioma cells after 6hr incubation. Refrering to fig. 5B depicts a quantitative data showing time and concentration dependent pattern of nanomedicine uptake as a measure of mTHPC florescence. The mTHPC florescence from the nanomedicine treated cells, visualized under florescent microscope, depicted iii figure 5C.
AM RITA VISWAVIDYAPEETHAM UNIVERSITY represented by its Director, Amrita Centre for Nanosciences, Dr. Shantikumar Nair In yet another aspect of the above embodiment, the cell migration inhibition properties of the core- shell nanomedicine is depicted in the figure 6. Fig 6A depicts the microscopic images of effective inhibition of cell migration.. The nanomedicine treated cel l showed effective inhibition in migration even after 1 8 hrs. while the untreated cells migrated and healed the scratch area. The quantitative measure of the same is depicted in figure 6B and 6C by means of total no. of cells migrated and the percentage scratch healing respectively. The p-Src level down regulation by nanomedicine-dasatinib, that intern inhibit cell migration is proved by western blot analysis depicted in fig. 6D. The nanomedicine treatment induced changes in cytoskeleton arrangement investigated is depicted in figure 7. The actin staining-confocal images of U87MG cell showed loss of proper cytoskeleton in the drug treated group. Loss of philopodial extension (fig 8A) and focal adhesion points (8B) were also seen in the nanomedicine treated cells.
In yet another aspect of the above embodiment, the nanomedicine induced intracellular ROS was visualized by DCFH staining and live cell D1C imaging. The nanomedicine treated cells showed enhanced production of intracellular ROS compared to bare photosensitizer and showed morphological changes associated with apoptosis (fig. 8). The photo-chemo nanomedicine treated ceils showed enhanced cytotoxicity compared to cells treated w ith nanoparticles having photodrug alone (fig 9). The dasatinib in the nano shell enhanced the PDT mediated cytotoxicity. The confocal images of the cells, one hour past the light irradiation (fig. 9B) shows the early stage apoptosis; Annexin V (F1TC conjugated) bound to the externalized phosphatidyl serine as a result of apoptosis.
The authors hav e invented a core-shell nanomedicine hav ing a clear interface for delivering a photosensitizer drug and a chemo-drug simultaneously/sequentially for combinatorial cancer treatment to aid enhanced cytotoxicity to cancer cells. The design of the nanomedicine is in such a way to simultaneously carry two different drugs and deliver it specifically to the tumor cells in desired fashion. The targeting is achieved by a specific biomarker ligand conjugated to the nanomedicine construct. The core and shell are made by different polymers or a protein and polymer aiding the optimum release kinetics.
Examples
Example I: Polymer core /protein shell photo-chemo-nanomedicine.
(PLGA-mTHPC core/Albumin-Dasatinib shell)
In this example preparation of a combinatorial polymer-protein core-shell nanomedicine with photsensitizer meta-tetra (hydroxyphenyl) chlorine (mTHPC) loaded polymeric [PLGA: Poly(lactic- co-glycolic ac id (50: 50)] nanocore and small molecule inhibitor dasatinib entrapped protein shell is presented. PLGA solution containing ! mM mTHPC was prepared by dissolv ing it in 1 .5 wt % PLGA solution in acetone and was stirred for 1 hr in dark prior to electrospray. This solution was then electrosptrayed at a rate of 1.5 ml/hr to a grounded glass beaker containing de-ionized water premixed with 0.001 v/v Tweeen 20® under constant stirring (550 - 600 rpm). The tip to target distance was maintained as 7.5 throughout the experiment. The electrospray was carried out under ambient temperature, pressure and 55±5% humidity, by applying a potential of 1 .4kV/cm using a high voltage supply.
AM RITA VISWAVIDYAPEETHAM UNIVERSITY represented by its Director, Amrita Centre for Nanosciences, Dr. Shantikumar Nair The particles were acquired by lyophilizing the electrosprayed solution. The A lbumin-dasatinib solution was prepared by mixing fresh dasatinib-DMSO stock in 5- 10 wt % BSA solution in de- ionized water to get a final dasatinib concentration of 1 0 μ Μ . Lyophilized PLGA-mTHPC particles were mixed with the BSA-dasatinib solution and stirred for 2 hours prior to co-acrervation. BSA- dasatinib nanoshell was prepared over PLGA-mTHPC nanoparticles by ethanol co-acervation, in that 1 :2 volume of absolute ethanol was added drop wise ( Ι ΟΟμΙ/min) under stirring (800- 1 000 rpm) till the solution became turbid, to aid optimum albumin-dasatinib co-acervate coating over the PLGA- mTHPC NPs. The system is then added with 35μ Ι of 4mg/ml l -Ethy l-3-[3-dimethylaminopropyl] carbodiimide hydrochloride (EDC) and stirred for 2 hours to link and harden the coacervates formed. The core-shell nanomedicine constructs were collected by lyophilizing the above solution.
Example 2: Polymer core /polymer shell photo-chemo-nanomedicine
(PCL- tTHPC core/ Chitosan-Sorufenib shell)
In this example preparation of a combinatorial polymer-polymer core-shell nanomedicine with photsensitizer mTHPC loaded Poiycaprolactone (PCL) nanocore and dasatin ib encapsulated chitosan shell is presented. I mM mTHPC containing polymer solution was prepared by disso lving the required 4mg/ml stock in 1 .0 wt % PCL solution in chloroform and was stirred for 1 hr in dark prior to electrospray. This solution was then electrosptrayed at a rate of 1 ml/hr to a grounded glass beaker kept at a distance of 7cm that containing de-ionized water with 0.00 1 v/v Tweeen 20® as surfactant and was maintained in constant stirring (600 rpm). The electrospray was carried out under ambient temperature, pressure and 60±5% humidity, by apply ing a potent ial of 1 .3 kV/cm, using a high voltage supply. The partic les were acquired by lyophilizing the electrosprayed solution. T he lyophilized particles are then mixed with 0.5% chitosan solution containing 5 mM sorafenib. 0.25 wt% Tween 80 w as added to his solution to prevent particle aggregation and the system was we l l m ixed by stirring for 30 min. Chitosan-sorafenib nanoshell was prepared over PCL-mTHPC nanopatic les by ionic gelation process. Aqueous tripolyphosphate (TPP: 0.25% w/v) solution w as added dropwise into the above solution and stirred under room temperature. The core shell nanoparticles were obtained by centrifuging the suspention at 12,000 rpm for 30 min
AMRITA VISWAVIDYAPEETHAM UNIVERSITY represented by its Director, Amrita Centre for Nanosciences, Dr. Shantikumar Nair REFERENCES CITED
1 . Araujo, J., & Logothetis, C. (2010). Cancer treatment reviews, 36(6), 492-500.
2. Aziz, F., Telara, S., Moseley, H., Goodman, C, Manthri, P., & Eljamel, M. S. (2009). Photodiagnosis and photodynamic therapy, 6(3-4), 227-30.
3. Bao, S., Wu, Q., McLendon, R. E., Hao, Y., Shi, Q., Hjelmeland, A. B„ Dewhirst, M. W ., et at. (2006).
Nature, 444(7120), 756-60.
4. Brunton, V. G., & Frame, M. C. (2008). Current opinion in pharmacology, 8(4), 427-32.
5. Diehn, M., Cho, R. W., Lobo, N. A., Kalisky, T., Dorie, M. J ., ulp, A. N., Qian, D., et al. (2009).
Nature, 458(7239), 780-783.
6. Jiang, F., Cho, K.-K., Mikkelse, T., Tong, L., Lew, Y . S., Hochbaum, N., Shargorodsky, J., et al.
(2002). Journal of neuro-oncology, 56( 1 ), 51 -8.
7. K stle, M., Grimm, S., Nagel, R., Breusing, N., & Grune, T. (201 1 ). Free radical biology & medicine, 50(2), 305- 12.
8. Khdair, A., Chen, D., Patil, Y., Ma, L., Dou, Q. P, Shekhar, M. P. V., & Panyam, J. (2010). Journal of controlled release, 141 (2), 137-44.
9. Khdair, A., Handa, H., Mao, G., & Panyam, J. (2009). European journal of pharmaceutics and biopharmaceutics, 71 (2), 2 14-22.
10. Peng, C.-L., Lai, P.-S., Lin, F.-H., Yueh-Ksiu Wu, S„ & Shieh, .-J . (2009). Biomateria!s, 30(21 ), 3614-25.
1 1 . Raguz, S., & Yague, E. (2008). British journal of cancer, 99(3), 387- 1 .
US patent documents
165440 12/2000 Esenaliev et al
181852 6/2008 Yu et al
0226393 9/2009 Wang et al
0020457 1/201 1 Panyam et al
0009441 1/2007.. Erathodiyil et al
0253969 10/2008 Yu et al
10238001 9/201 1 Chen et al
0022 129 1 /201 1 Robert et al
0165258 7/2 1 1.. Kabanov et al
■10217363 9/201 1 Jun then
6080396 6/2000
5449513 9/1 95 . . Yokoyama et al
100 1534 4/201 1 Kurt et al
015901 6/2010 Yang et al
7638558 12/2009 Kurt et al
8021652 9/201 1 Seo et al.
10027172 2/201 1
10229556 9/201 1
731 1901 12/2007 , , Seo et al.
0152616 6/2008 Seo et al
0253899 1 1/2007 Ai et al
0159020 6/2010
0182776 7/2008 Lee et al
0214633 8/2009 Yuk et al
0248126 10/2008 Cheng et al
0203 149 7/2010 Radosz et al
0052709 3/201 1 Lillard et al
0281777 12/2005 Albrechet
0012033 1/2009 Cordell et al
European Patent(s)
EP 0552802 7/1993 Nair et al

Claims

Claims:
1 . We c laim a core-shell photo-chemo nano medic ine system having a polymer core with average size of < 500nm and a shell made up of another polymer or protein with average size of < 500nm, and the core and the shell are loaded with a photosensitizer drug and chemo- drug, separately with distinct interface.
2. The composition of claim 1 , wherein the polymer used is a single or blended cross linked polymer and the protein used is in its native or denatured and cross linked form.
3. The composition of claim 1 , wherein the polymer core is loaded with at least one photosensitizer or chemo-drug
4. The composition of claim 1 , wherein the polymer/protein shell is loaded with at least one chemo- or a photosensitizer-drug
5. The composition of claim 1. wherein the polymer used is preferably a natural or synthetic biodegradable polymer at least one from the group, but not l imited to poly glycolic acid (PGA), poly(lactic-co-glycolic acid) (PLGA). glyeol ide/trimethylene carbonate copolymers (PGA/TMC); poly-lactides (PLA), poly-L-lactide (PLLA), Poly-DL-Iactide ( PDLLA), L- lactide/DL-lactide copolymers; lactide/tetramethyl-glycolide copolymers, poly-caprolactone (PCL), poly-valerolacton(PVL), poly-hydroxy butyrate (PHB), poly vinyl alcohol (PVA) poly-hydroxy valerate(PHV), polyvinylpyrrol idone (PV P), Polyethyleneimine (PEI) and lactide/trimethylene carbonate copolymers, chitosan. carboxymethyl chitosan. chitin. pollulan, etc., or blends thereof.
6. The composition of claim 1 , wherein the protein is chosen from, but not l imited to the group of human serum albumin, or protamine, transferrin, lactoferrin, fibrinogen, gelatin, mucin, soy protein, apoferritin, ferritin, lectin, lactoferrin, gluten, whey protein, prolamines such as gliadin, hordein, secalin, zein, avenin, or combinations thereof.
7. The composition of claim 1 , wherein the chemo drug loaded to the polymer core or polymer/protein shell is chosen from, but not limited to the group of anti-neoplastic agentssuch as cyclophosphamide, methotrexate, 5-fluorouracil, doxorubicin, cisplatin, hydroxyurea, leucovorin calcium, tamoxifen, flutamide, asparaginase, altretamine, niitotane, procarbazine hydrochloride, mechlorethamine, thioguanine, carmustine, lomustine, temozolomide, melphalan, chlorambucil, streptozocin, methotrexate, vincristine, bleomycin, vinblastine, vindesine, dactinomycin, 6-MP,daunorubicin, Lenalidomide, L-asparginase, doxorubicin, tamoxifen or anti-pro liferative agents such as rapamycin, paclitaxel or anti- angiogenesis agents such as avastin, or inhibitors of tyrosine kinase including epidermal growth factor receptor (EGFR), vascular endothelial growth factor receptor (VEGFR), platelet derived growth factor receptor (PDGFR), fibroblast growth factor receptor (FGFR), Rous sarcoma oncogene/Breakpoint cluster region/Abl (Src-bcr-abl), Insulin-tike growth factor 1 receptor (1GF- 1 R), FLT-3, HER-2, STAT5 , c- it, c-Met, AL , ETA receptor inhibitor, HIF inhibitor, Syk inhibitor, Tie2 kinase inhibitor and the l ike), Vascular disrupting agents (e.g. plinabulin), cell cycle/check point inhibitors like polo-like kinase (PLK) inhibitor (e.g. volasertib), cyclin dependent kinase (CDK) inhibitors (e.g. selicicl ib, indirubin etc.,), topoisomerase inhibitors (e.g. adriamycin, camptothec in, etoposide, idarubicin, irinotecan, topotecan, mitoxantrone etc.,), microtubule inhibitors (e.g. docetaxel, paclitaxel, vincristine etc.,), antimetabolites (e.g. decitabine, gemcitabine, fiudarabine etc.,), telomerase inhibitors, DNA & RNA replication inhibitors (e.g. clarithromyc in, cytarabine, mitoxantrone HCI etc.,) dihydrofolate reductase inhibitor, HDAC inhibitor, Bc l-2 and TNF-a inhibitors, PARP inhibitors, AP inhibitors, P13 K/Akt/mTOR inhibitors, infegrase and protease inhibitors, Wnt/Hedgehog/Notch inhibitors, cA P, lipide signaling inhibitors (e.g. P C, PI M etc.,), TGF-β inhibitors, chemotherapeutic pro-drugs, antioxidant inhibitors like diethyl- dithiocarbamate, methoxyestradiol, 1-buthionine sulfoxiinine, 3-amino- l ,2,4-triazole and the combinations thereof.
8. The composition of claim 1 , wherein the photosensitize!' loaded to the core or the shell is chosen from, but not limited to ch lorine e6 (Ce6), meso-tetra(3-hydroxypheny))ehlorin (m- THPC), methylene blue, benzoporphyrin derivative monoacid ring A (BPD or verteporfin), photofrin, Rose bengal, metal phthalocyanine, hyperic in, toluidine blue O, pyropheophorbide- a hexyl ether (HPPH), Indium pyropheophorbide, padoporfin, padeliporfin, and combinations of thereof.
9. The composition of claim 1 , wherein the chemo-drug is loaded into the core or the shell as monomers or aggregates or semi-aggregates or its combinations.
10. The composition of claim 1 , wherein the photosensitizer is loaded into the core or shell as monomers or aggregates or semi-aggregates or its combinations.
1 1 . The composition in claim 1 , the photo-chemo nanomedicine will be able to del iver the drugs from the shell and the core sequentially or simultaneously
12. The composition of claim 1 , the photo-chemo nanomedicine is spec ifically delivered to the disease like cancer either by passive or active targeting mechanisms
13. The composition in claim 12, where the active targeting is achieved by conjugating with ligands such as monoclonal antibody, (for example, CD20, CD33 , CD34, CD38, CD44, CD47, CD52 CD90, CD 123, CD 1 33, EGFR, PDGFR, V EGF. HER2, Transferrin receptors and like) peptides, (for example RGD, CR.GD, LyP- 1 peptide, bombes in ( BBNi) pepiide, FSH33 , truncated human basic fibroblast growth factor peptide (tbFG F), octreotide etc.,), small molecules (folid acid, mannose, hyaluronic acid (HA)) , proteins (transferrin, somatoatatin) or aptamers and like.
14. W composition of claim 1 , the photo-chemo nanomedicine is administered either intravenously, orally, parenterally, subcutaneously or by direct local delivery.
15. W claim a method of treatment using photo-chemo nanomedicine by applying chemotherapy followed by photodynamic therapy or photodynamic therapy fol lowed by chemotherapy or simultaneous photodynamic- and chemo-therapy.
16. We claim a process for making core-shell photo-chemo nanomedicine loaded with a photosensitizer and chemo-drugs, either in the core or the shell
17. The process in claim 16, where the polymer core is first formed by a method of spontaneous emulsification or solvent diffusion or salting out or emulsification-diffusion or supercritical fluid technology or micro emulsion or double emulsion or electrospray or electro deposition or ultrasonication or spray drying or like.
18. The process in claim 16, wherein the protein shell is formed over the polymer core by simple desolvation or co-acervation, complex co-acervation, nano-precipitation, sol-gel, spray drying slating-out or cross linking
19. The process in claim 14, wherein the polymer shell over polymer core is formed by the process of nano-precipitation, microemulsion, spontaneous emulsification, double emulsion, emulsion-droplet coalescence, solvent evaporation emulsification, electrospray, solvent diffusion, salting out or emulsification-diffusion or supercritical fluid technology electro deposition or ultrasonication or spray drying or like.
AMRITA VISWAVIDYAPEETHA UNIVERSITY represented by its Director, Amrira Centre for Nanosciences, Dr. Shantikumar Nair
PCT/IN2013/000144 2013-03-12 2013-03-12 Photo - chemo composition on the basis of microcapsules with a core -shell structure WO2014141289A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/IN2013/000144 WO2014141289A1 (en) 2013-03-12 2013-03-12 Photo - chemo composition on the basis of microcapsules with a core -shell structure

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/IN2013/000144 WO2014141289A1 (en) 2013-03-12 2013-03-12 Photo - chemo composition on the basis of microcapsules with a core -shell structure

Publications (1)

Publication Number Publication Date
WO2014141289A1 true WO2014141289A1 (en) 2014-09-18

Family

ID=48699210

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IN2013/000144 WO2014141289A1 (en) 2013-03-12 2013-03-12 Photo - chemo composition on the basis of microcapsules with a core -shell structure

Country Status (1)

Country Link
WO (1) WO2014141289A1 (en)

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104840424A (en) * 2015-05-05 2015-08-19 中国农业科学院兰州畜牧与兽药研究所 Hypericin albumin nanoparticle and escherichia coli serum antibody compound and preparation method and application thereof
CN105343015A (en) * 2015-11-02 2016-02-24 上海交通大学 Preparation method of antineoplastic drug carrier having dual-lymphatic targeting
CN107158393A (en) * 2017-06-20 2017-09-15 苏州乔纳森新材料科技有限公司 A kind of copolymerized material of pulullan polysaccharide PLA and preparation method thereof
WO2017179053A1 (en) * 2016-04-10 2017-10-19 Yeda Research And Development Co. Ltd. Combinational therapies for treatment of cancer comprising a bacteriochlorophyll derivative
RU2637279C2 (en) * 2015-12-25 2017-12-01 Сергей Петрович Кречетов Method for photodynamic therapy
CN108042544A (en) * 2017-11-21 2018-05-18 北京德得创业科技有限公司 A kind of toluidines blue light dynamic pasteurization and promotion healing composition and its application
CN108096583A (en) * 2017-12-17 2018-06-01 宋振川 The preparation method of the cancer target nanoparticulate carriers of mammary cancer chemotherapy drug MTDH siRNA is loaded with altogether
CN108671238A (en) * 2018-05-14 2018-10-19 江苏医药职业学院 A kind of preparation method of the high tumor infiltrating albumin nano system of combination therapy
CN108704134A (en) * 2018-08-31 2018-10-26 重庆医科大学 A kind of targeting multifunctional nano grain containing IR780, using and preparation method thereof
EP3277268A4 (en) * 2015-03-31 2018-12-05 The General Hospital Corporation Self assembling molecules for targeted drug delivery
CN109293738A (en) * 2016-03-10 2019-02-01 福州大学 One kind has the Phthalocyanine Zinc adriamycin conjugate of phototherapy and chemotherapy Synergistic anti-cancer effect
CN109517173A (en) * 2018-11-12 2019-03-26 华南理工大学 A kind of branch polyether acid imide material and the preparation method and application thereof containing Contracting thioketones key
CN110354276A (en) * 2019-07-17 2019-10-22 山东大学 A kind of prodrug and its preparation method and application
CN110755615A (en) * 2019-11-14 2020-02-07 天津大学 Synthesis method of Gd-CuS @ Flu @ RGD nano preparation
CN110975007A (en) * 2019-12-09 2020-04-10 宁夏医科大学 bFGF-loaded guided tissue regeneration membrane with core-shell structure and preparation method thereof
CN111588853A (en) * 2020-06-01 2020-08-28 南京林业大学 Preparation method and application of chemotherapy-phototherapy synergistic antitumor microspheres
CN112121162A (en) * 2020-09-07 2020-12-25 四川康城生物科技有限公司 Carboxymethyl chitosan microcapsule for carrying chemotherapy medicament and thermotherapy sensitizer for sustained release of novel embolism and preparation method and application thereof
CN113143854A (en) * 2021-04-28 2021-07-23 上海健康医学院 Application of human serum albumin in preparation of radioiodine hypericin pharmaceutical preparation
CN113350503A (en) * 2021-05-20 2021-09-07 沈阳药科大学 Carrier-free hybrid nano assembly and preparation method and application thereof
CN113648283A (en) * 2021-07-23 2021-11-16 丽水市中心医院 Preparation method of drug-loaded microsphere for targeted inhibition of HIF-2 alpha, drug-loaded microsphere and application
CN114452406A (en) * 2022-03-16 2022-05-10 安徽工程大学 Antibacterial material and preparation method and application thereof
CN114949208A (en) * 2022-05-06 2022-08-30 温州医科大学 Nanometer photodynamic material and application thereof in treatment of choroidal neovascularization
CN115350283A (en) * 2022-03-22 2022-11-18 四川大学 Carbohydrate functionalized nano-particles and preparation method and application thereof
CN115634293A (en) * 2022-09-27 2023-01-24 石河子大学 Preparation method and application of nano-carrier for targeted delivery of antitumor drugs
CN115804841A (en) * 2022-10-21 2023-03-17 山东大学 Tumor targeted drug-loaded nano preparation based on polyglutamic acid conjugated photosensitizer
WO2023068423A1 (en) * 2021-10-21 2023-04-27 닥터아이앤비(주) Complex anticancer drug composition using photoreactivity and preparation method therefor
US20230210842A1 (en) * 2021-12-30 2023-07-06 The Eighth Affiliated Hospital, Sun Yat-Sen University Preparation method and application of supramolecular nano-drug based on irinotecan and niraparib
CN116396465A (en) * 2022-07-21 2023-07-07 苏州炫景生物科技有限公司 Polymeric nucleic acid delivery vehicles and pharmaceutical composition applications

Citations (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0552802A2 (en) 1992-01-23 1993-07-28 Eastman Kodak Company Chemically fixed micelles comprising copolymer
US5449513A (en) 1992-08-14 1995-09-12 Research Development Corporation Of Japan Physical trapping type polymeric micelle drug preparation
US6080396A (en) 1995-09-29 2000-06-27 Japan Science And Technology Corporation Anthracycline compound derivative and pharmaceutical preparation containing the same
US20050281777A1 (en) 2004-06-18 2005-12-22 Ceramoptec Industries, Inc. Photo-triggered release of active substances from dendrimer-photosensitizer complexes
US20060165440A1 (en) 2005-01-26 2006-07-27 Sharp Kabushiki Kaisha Development apparatus and image forming apparatus comprising the same
WO2006088473A2 (en) * 2004-04-23 2006-08-24 Panduranga Rao Koritala Microcapsules and nanocapsules for the transmucosal delivery of therapeutic and diagnostic agents
US20070009441A1 (en) 2004-07-08 2007-01-11 Molecular Therapeutics, Inc. Biodegradable nanoparticles
US20070253899A1 (en) 2004-06-04 2007-11-01 Hua Ai Dual Function Polymer Micelles
US7311901B2 (en) 2003-10-10 2007-12-25 Samyang Corporation Amphiphilic block copolymer and polymeric composition comprising the same for drug delivery
US20080181852A1 (en) 2007-01-29 2008-07-31 Nitto Denko Corporation Multi-functional Drug Carriers
US20080182776A1 (en) 2007-01-31 2008-07-31 Tong Shen Enterprise Co., Ltd. Drug-loaded poly (alkyl-cyanoacrylate) nanoparticles and process foe the preparation thereof
US20080248126A1 (en) 2007-03-02 2008-10-09 Jianjun Cheng Particulate drug delivery
US20080253969A1 (en) 2007-04-10 2008-10-16 Nitto Denko Corporation Multi-functional polyglutamate drug carriers
US20090012033A1 (en) 2006-03-03 2009-01-08 Demattei Cordell R Delivery of Biologically Active Materials Using Core-Shell Tecto(Dendritic Polymers)
US20090214633A1 (en) 2006-04-05 2009-08-27 Hunnam University Institute for Industry Academia Cooperation Nanoparticles with lipid core and polymer shell structures for protein drug delivery prepared by nanoencapsulation
US20090226393A1 (en) 2008-03-06 2009-09-10 Nitto Denko Corporation Polymer paclitaxel conjugates and methods for treating cancer
US7638558B2 (en) 2005-04-01 2009-12-29 Intezyne Technologies, Inc. Polymeric micelles for drug delivery
US20100159019A1 (en) 2006-07-05 2010-06-24 Yi-Yan Yang Micelles For Drug Delivery
US20100203149A1 (en) 2004-09-10 2010-08-12 University Of Wyoming Nanoparticles for Cytoplasmic Drug Delivery to Cancer Cells
WO2010143942A1 (en) * 2009-06-12 2010-12-16 Erasmus University Medical Center Rotterdam Targeted nano-photomedicines for photodynamic therapy of cancer
US20110020457A1 (en) 2006-08-14 2011-01-27 Wayne State University Polymer-surfactant nanoparticles for sustained release of compounds
US20110022129A1 (en) 2007-11-05 2011-01-27 Prud Homme Robert K Nanoparticles for photodynamic therapy
US20110027172A1 (en) 2007-12-10 2011-02-03 Zhuang Wang Drug delivery system for pharmaceuticals and radiation
US20110052709A1 (en) 2006-03-21 2011-03-03 Morehouse School Of Medicine Novel nanoparticles for delivery of active agents
US20110165258A1 (en) 2008-06-26 2011-07-07 Technische Universitat Munchen Polymeric delivery systems for active agents
US20110217363A1 (en) 2010-03-05 2011-09-08 Bionanox Two-step targeted tumor therapy with prodrug encapsulated in nanocarrier
US8021652B2 (en) 2003-12-04 2011-09-20 Samyang Corporation Biodegradable branched polylactide derivatives capable of forming polymeric micelles, and their preparation method and use
US20110229556A1 (en) 2010-03-19 2011-09-22 Massachusetts Institute Of Technology Lipid-coated polymer particles for immune stimulation
US20110238001A1 (en) 2005-02-02 2011-09-29 Wei Chen Nanoparticle based photodynamic therapy and methods of making and using same

Patent Citations (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0552802A2 (en) 1992-01-23 1993-07-28 Eastman Kodak Company Chemically fixed micelles comprising copolymer
US5449513A (en) 1992-08-14 1995-09-12 Research Development Corporation Of Japan Physical trapping type polymeric micelle drug preparation
US6080396A (en) 1995-09-29 2000-06-27 Japan Science And Technology Corporation Anthracycline compound derivative and pharmaceutical preparation containing the same
US7311901B2 (en) 2003-10-10 2007-12-25 Samyang Corporation Amphiphilic block copolymer and polymeric composition comprising the same for drug delivery
US20080152616A1 (en) 2003-10-10 2008-06-26 Min Hyo Seo Amphiphilic block copolymer and polymeric compostition comprising the same for drug delivery
US8021652B2 (en) 2003-12-04 2011-09-20 Samyang Corporation Biodegradable branched polylactide derivatives capable of forming polymeric micelles, and their preparation method and use
WO2006088473A2 (en) * 2004-04-23 2006-08-24 Panduranga Rao Koritala Microcapsules and nanocapsules for the transmucosal delivery of therapeutic and diagnostic agents
US20070253899A1 (en) 2004-06-04 2007-11-01 Hua Ai Dual Function Polymer Micelles
US20050281777A1 (en) 2004-06-18 2005-12-22 Ceramoptec Industries, Inc. Photo-triggered release of active substances from dendrimer-photosensitizer complexes
US20070009441A1 (en) 2004-07-08 2007-01-11 Molecular Therapeutics, Inc. Biodegradable nanoparticles
US20100203149A1 (en) 2004-09-10 2010-08-12 University Of Wyoming Nanoparticles for Cytoplasmic Drug Delivery to Cancer Cells
US20060165440A1 (en) 2005-01-26 2006-07-27 Sharp Kabushiki Kaisha Development apparatus and image forming apparatus comprising the same
US20110238001A1 (en) 2005-02-02 2011-09-29 Wei Chen Nanoparticle based photodynamic therapy and methods of making and using same
US20100159020A1 (en) 2005-04-01 2010-06-24 Intezyne Technologies, Inc. Polymeric micelles for drug delivery
US7638558B2 (en) 2005-04-01 2009-12-29 Intezyne Technologies, Inc. Polymeric micelles for drug delivery
US20110091534A1 (en) 2005-04-01 2011-04-21 Intezyne Technologies, Inc. Compositions comprising polymeric micelles for drug delivery
US20090012033A1 (en) 2006-03-03 2009-01-08 Demattei Cordell R Delivery of Biologically Active Materials Using Core-Shell Tecto(Dendritic Polymers)
US20110052709A1 (en) 2006-03-21 2011-03-03 Morehouse School Of Medicine Novel nanoparticles for delivery of active agents
US20090214633A1 (en) 2006-04-05 2009-08-27 Hunnam University Institute for Industry Academia Cooperation Nanoparticles with lipid core and polymer shell structures for protein drug delivery prepared by nanoencapsulation
US20100159019A1 (en) 2006-07-05 2010-06-24 Yi-Yan Yang Micelles For Drug Delivery
US20110020457A1 (en) 2006-08-14 2011-01-27 Wayne State University Polymer-surfactant nanoparticles for sustained release of compounds
US20080181852A1 (en) 2007-01-29 2008-07-31 Nitto Denko Corporation Multi-functional Drug Carriers
US20080182776A1 (en) 2007-01-31 2008-07-31 Tong Shen Enterprise Co., Ltd. Drug-loaded poly (alkyl-cyanoacrylate) nanoparticles and process foe the preparation thereof
US20080248126A1 (en) 2007-03-02 2008-10-09 Jianjun Cheng Particulate drug delivery
US20080253969A1 (en) 2007-04-10 2008-10-16 Nitto Denko Corporation Multi-functional polyglutamate drug carriers
US20110022129A1 (en) 2007-11-05 2011-01-27 Prud Homme Robert K Nanoparticles for photodynamic therapy
US20110027172A1 (en) 2007-12-10 2011-02-03 Zhuang Wang Drug delivery system for pharmaceuticals and radiation
US20090226393A1 (en) 2008-03-06 2009-09-10 Nitto Denko Corporation Polymer paclitaxel conjugates and methods for treating cancer
US20110165258A1 (en) 2008-06-26 2011-07-07 Technische Universitat Munchen Polymeric delivery systems for active agents
WO2010143942A1 (en) * 2009-06-12 2010-12-16 Erasmus University Medical Center Rotterdam Targeted nano-photomedicines for photodynamic therapy of cancer
US20110217363A1 (en) 2010-03-05 2011-09-08 Bionanox Two-step targeted tumor therapy with prodrug encapsulated in nanocarrier
US20110229556A1 (en) 2010-03-19 2011-09-22 Massachusetts Institute Of Technology Lipid-coated polymer particles for immune stimulation

Cited By (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3277268A4 (en) * 2015-03-31 2018-12-05 The General Hospital Corporation Self assembling molecules for targeted drug delivery
AU2016242920B2 (en) * 2015-03-31 2021-10-21 The General Hospital Corporation Self assembling molecules for targeted drug delivery
EP3936121A1 (en) * 2015-03-31 2022-01-12 The General Hospital Corporation Self assembling molecules for targeted drug delivery
US11666664B2 (en) 2015-03-31 2023-06-06 The General Hospital Corporation Self assembling molecules for targeted drug delivery
CN104840424B (en) * 2015-05-05 2017-09-15 中国农业科学院兰州畜牧与兽药研究所 A kind of hypericin albumin nanoparticle Escherichia coli serum antibody compound and its preparation method and application
CN104840424A (en) * 2015-05-05 2015-08-19 中国农业科学院兰州畜牧与兽药研究所 Hypericin albumin nanoparticle and escherichia coli serum antibody compound and preparation method and application thereof
CN105343015A (en) * 2015-11-02 2016-02-24 上海交通大学 Preparation method of antineoplastic drug carrier having dual-lymphatic targeting
RU2637279C2 (en) * 2015-12-25 2017-12-01 Сергей Петрович Кречетов Method for photodynamic therapy
CN109293738B (en) * 2016-03-10 2020-11-27 福州大学 Zinc phthalocyanine adriamycin conjugate with phototherapy and chemotherapy synergistic anticancer effect
CN109293738A (en) * 2016-03-10 2019-02-01 福州大学 One kind has the Phthalocyanine Zinc adriamycin conjugate of phototherapy and chemotherapy Synergistic anti-cancer effect
WO2017179053A1 (en) * 2016-04-10 2017-10-19 Yeda Research And Development Co. Ltd. Combinational therapies for treatment of cancer comprising a bacteriochlorophyll derivative
CN109862918A (en) * 2016-04-10 2019-06-07 耶达研究及发展有限公司 The conjoint therapy for treating cancer including bacteriochlorophll derivatives
US11278555B2 (en) 2016-04-10 2022-03-22 Yeda Research And Development Co. Ltd. Combinational therapies for treatment of cancer comprising a bacteriochlorophyll derivative
CN107158393A (en) * 2017-06-20 2017-09-15 苏州乔纳森新材料科技有限公司 A kind of copolymerized material of pulullan polysaccharide PLA and preparation method thereof
CN108042544A (en) * 2017-11-21 2018-05-18 北京德得创业科技有限公司 A kind of toluidines blue light dynamic pasteurization and promotion healing composition and its application
CN108096583A (en) * 2017-12-17 2018-06-01 宋振川 The preparation method of the cancer target nanoparticulate carriers of mammary cancer chemotherapy drug MTDH siRNA is loaded with altogether
CN108671238A (en) * 2018-05-14 2018-10-19 江苏医药职业学院 A kind of preparation method of the high tumor infiltrating albumin nano system of combination therapy
CN108704134A (en) * 2018-08-31 2018-10-26 重庆医科大学 A kind of targeting multifunctional nano grain containing IR780, using and preparation method thereof
CN109517173B (en) * 2018-11-12 2021-03-30 华南理工大学 Branched polyethyleneimine material containing thioketal bond and preparation method and application thereof
CN109517173A (en) * 2018-11-12 2019-03-26 华南理工大学 A kind of branch polyether acid imide material and the preparation method and application thereof containing Contracting thioketones key
CN110354276A (en) * 2019-07-17 2019-10-22 山东大学 A kind of prodrug and its preparation method and application
CN110755615A (en) * 2019-11-14 2020-02-07 天津大学 Synthesis method of Gd-CuS @ Flu @ RGD nano preparation
CN110975007A (en) * 2019-12-09 2020-04-10 宁夏医科大学 bFGF-loaded guided tissue regeneration membrane with core-shell structure and preparation method thereof
CN111588853A (en) * 2020-06-01 2020-08-28 南京林业大学 Preparation method and application of chemotherapy-phototherapy synergistic antitumor microspheres
CN112121162A (en) * 2020-09-07 2020-12-25 四川康城生物科技有限公司 Carboxymethyl chitosan microcapsule for carrying chemotherapy medicament and thermotherapy sensitizer for sustained release of novel embolism and preparation method and application thereof
CN113143854A (en) * 2021-04-28 2021-07-23 上海健康医学院 Application of human serum albumin in preparation of radioiodine hypericin pharmaceutical preparation
CN113350503A (en) * 2021-05-20 2021-09-07 沈阳药科大学 Carrier-free hybrid nano assembly and preparation method and application thereof
CN113648283A (en) * 2021-07-23 2021-11-16 丽水市中心医院 Preparation method of drug-loaded microsphere for targeted inhibition of HIF-2 alpha, drug-loaded microsphere and application
CN113648283B (en) * 2021-07-23 2023-11-07 丽水市中心医院 Preparation method of drug-loaded microsphere for targeted inhibition of HIF-2 alpha, drug-loaded microsphere and application
WO2023068423A1 (en) * 2021-10-21 2023-04-27 닥터아이앤비(주) Complex anticancer drug composition using photoreactivity and preparation method therefor
US20230210842A1 (en) * 2021-12-30 2023-07-06 The Eighth Affiliated Hospital, Sun Yat-Sen University Preparation method and application of supramolecular nano-drug based on irinotecan and niraparib
CN114452406B (en) * 2022-03-16 2023-05-23 安徽工程大学 Antibacterial material and preparation method and application thereof
CN114452406A (en) * 2022-03-16 2022-05-10 安徽工程大学 Antibacterial material and preparation method and application thereof
CN115350283A (en) * 2022-03-22 2022-11-18 四川大学 Carbohydrate functionalized nano-particles and preparation method and application thereof
CN115350283B (en) * 2022-03-22 2024-01-26 四川大学 Carbohydrate functionalized nano-particle and preparation method and application thereof
CN114949208A (en) * 2022-05-06 2022-08-30 温州医科大学 Nanometer photodynamic material and application thereof in treatment of choroidal neovascularization
CN114949208B (en) * 2022-05-06 2023-07-04 温州医科大学 Nanometer photodynamic material and application of same in treatment of choroidal neovascularization
CN116396465A (en) * 2022-07-21 2023-07-07 苏州炫景生物科技有限公司 Polymeric nucleic acid delivery vehicles and pharmaceutical composition applications
CN116396465B (en) * 2022-07-21 2024-02-13 苏州炫景生物科技有限公司 Polymeric nucleic acid delivery vehicles and pharmaceutical composition applications
CN115634293A (en) * 2022-09-27 2023-01-24 石河子大学 Preparation method and application of nano-carrier for targeted delivery of antitumor drugs
CN115804841A (en) * 2022-10-21 2023-03-17 山东大学 Tumor targeted drug-loaded nano preparation based on polyglutamic acid conjugated photosensitizer

Similar Documents

Publication Publication Date Title
WO2014141289A1 (en) Photo - chemo composition on the basis of microcapsules with a core -shell structure
Wang et al. High co-loading capacity and stimuli-responsive release based on cascade reaction of self-destructive polymer for improved chemo-photodynamic therapy
Rahikkala et al. Mesoporous silica nanoparticles for targeted and stimuli‐responsive delivery of chemotherapeutics: A review
US9545382B2 (en) Nanoparticle formulations for delivering multiple therapeutic agents
Sun et al. Stimuli-responsive nanoparticles based on co-assembly of naturally-occurring biomacromolecules for in vitro photodynamic therapy
US10143700B2 (en) Nanoparticle formulations for delivering multiple therapeutic agents
Teng et al. Development and application of nanoparticles synthesized with folic acid conjugated soy protein
Pulakkat et al. Surface engineered protein nanoparticles with hyaluronic acid based multilayers for targeted delivery of anticancer agents
Poudel et al. Macrophage-membrane-camouflaged disintegrable and excretable nanoconstruct for deep tumor penetration
Bhattacharya Fabrication of poly (sarcosine), poly (ethylene glycol), and poly (lactic-co-glycolic acid) polymeric nanoparticles for cancer drug delivery
Grigore Organic and inorganic nano-systems used in cancer treatment
Gao et al. Surface PEGylated cancer cell membrane-coated nanoparticles for codelivery of curcumin and doxorubicin for the treatment of multidrug resistant esophageal carcinoma
Gu et al. Polymeric nanocarriers for drug delivery in osteosarcoma treatment
Sebak et al. Targeted photodynamic-induced singlet oxygen production by peptide-conjugated biodegradable nanoparticles for treatment of skin melanoma
Hou et al. Construction of near infrared light triggered nanodumbbell for cancer photodynamic therapy
Zhou et al. Alternative and injectable preformed albumin-bound anticancer drug delivery system for anticancer and antimetastasis treatment
Loya-Castro et al. Preparation of PLGA/Rose Bengal colloidal particles by double emulsion and layer-by-layer for breast cancer treatment
Banik et al. Polymeric biomaterials in nanomedicine
Fathi-Karkan et al. Recent advancements in the targeted delivery of etoposide nanomedicine for cancer therapy: A comprehensive review
Truong-Dinh Tran et al. Nanoparticulate drug delivery to colorectal cancer: formulation strategies and surface engineering
Rizi et al. Polymeric nanoparticles in cancer chemotherapy: a narrative review
Abdelsalam et al. Surface tailored zein as a novel delivery system for hypericin: Application in photodynamic therapy
Bai et al. Progress and principle of drug nanocrystals for tumor targeted delivery
Ackova et al. Contemporary formulations for drug delivery of anticancer bioactive compounds
Chen et al. Cooperative coordination-mediated multi-component self-assembly of “all-in-one” nanospike theranostic nano-platform for MRI-guided synergistic therapy against breast cancer

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: 13731498

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13731498

Country of ref document: EP

Kind code of ref document: A1